1
|
Huang H, Wang S, Guan Y, Ren J, Liu X. Molecular basis and current insights of atypical Rho small GTPase in cancer. Mol Biol Rep 2024; 51:141. [PMID: 38236467 DOI: 10.1007/s11033-023-09140-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Accepted: 12/11/2023] [Indexed: 01/19/2024]
Abstract
Atypical Rho GTPases are a subtype of the Rho GTPase family that are involved in diverse cellular processes. The typical Rho GTPases, led by RhoA, Rac1 and Cdc42, have been well studied, while relative studies on atypical Rho GTPases are relatively still limited and have great exploration potential. With the increase in studies, current evidence suggests that atypical Rho GTPases regulate multiple biological processes and play important roles in the occurrence and development of human cancers. Therefore, this review mainly discusses the molecular basis of atypical Rho GTPases and their roles in cancer. We summarize the sequence characteristics, subcellular localization and biological functions of each atypical Rho GTPase. Moreover, we review the recent advances and potential mechanisms of atypical Rho GTPases in the development of multiple cancers. A comprehensive understanding and extensive exploration of the biological functions of atypical Rho GTPases and their molecular mechanisms in tumors will provide important insights into the pathophysiology of tumors and the development of cancer therapeutic strategies.
Collapse
Affiliation(s)
- Hua Huang
- Center of Excellence for Environmental Safety and Biological Effects, Faculty of Environment and Life, Beijing International Science and Technology Cooperation Base for Antiviral Drugs, Beijing University of Technology, Beijing, 100124, China
| | - Sijia Wang
- Center of Excellence for Environmental Safety and Biological Effects, Faculty of Environment and Life, Beijing International Science and Technology Cooperation Base for Antiviral Drugs, Beijing University of Technology, Beijing, 100124, China
| | - Yifei Guan
- Center of Excellence for Environmental Safety and Biological Effects, Faculty of Environment and Life, Beijing International Science and Technology Cooperation Base for Antiviral Drugs, Beijing University of Technology, Beijing, 100124, China
| | - Jing Ren
- Department of Plastic and Reconstructive Surgery, The First Medical Center, Chinese PLA (People's Liberation Army) General Hospital, Beijing, 100853, China.
| | - Xinhui Liu
- Center of Excellence for Environmental Safety and Biological Effects, Faculty of Environment and Life, Beijing International Science and Technology Cooperation Base for Antiviral Drugs, Beijing University of Technology, Beijing, 100124, China.
- Faculty of Environment and Life, Beijing University of Technology, Beijing, 100124, China.
| |
Collapse
|
2
|
Elhariri A, Alhaj A, Ahn D, Sonbol MB, Bekaii-Saab T, Wu C, Rutenberg MS, Stauffer J, Starr J, Majeed U, Jones J, Borad M, Babiker H. Targeting KRAS in pancreatic adenocarcinoma: Progress in demystifying the holy grail. World J Clin Oncol 2023; 14:285-296. [PMID: 37700806 PMCID: PMC10494558 DOI: 10.5306/wjco.v14.i8.285] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 07/05/2023] [Accepted: 07/27/2023] [Indexed: 08/22/2023] Open
Abstract
Pancreatic cancer (PC) remains one of the most challenging diseases, with a very poor 5-year overall survival of around 11.5%. Kirsten rat sarcoma virus (KRAS) mutation is seen in 90%-95% of PC patients and plays an important role in cancer cell proliferation, differentiation, metabolism, and survival, making it an essential mutation for targeted therapy. Despite extensive efforts in studying this oncogene, there has been little success in finding a drug to target this pathway, labelling it for decades as "undruggable". In this article we summarize some of the efforts made to target the KRAS pathway in PC, discuss the challenges, and shed light on promising clinical trials.
Collapse
Affiliation(s)
- Ahmed Elhariri
- Division of Hematology-Oncology, Department of Medicine, Mayo Clinic Florida, Mayo Clinic Cancer Center, Jacksonville, FL 32224, United States
| | - Ahmed Alhaj
- Division of Hematology-Oncology, Department of Medicine, Mayo Clinic Florida, Mayo Clinic Cancer Center, Jacksonville, FL 32224, United States
| | - Daniel Ahn
- Division of Hematology-Oncology, Department of Medicine, Mayo Clinic Arizona, Mayo Clinic Cancer Center, Phoenix, AZ 85054, United States
| | - Mohamad Bassam Sonbol
- Division of Hematology-Oncology, Department of Medicine, Mayo Clinic Arizona, Mayo Clinic Cancer Center, Phoenix, AZ 85054, United States
| | - Tanios Bekaii-Saab
- Division of Hematology-Oncology, Department of Medicine, Mayo Clinic Arizona, Mayo Clinic Cancer Center, Phoenix, AZ 85054, United States
| | - Christina Wu
- Division of Hematology-Oncology, Department of Medicine, Mayo Clinic Arizona, Mayo Clinic Cancer Center, Phoenix, AZ 85054, United States
| | - Michael Scott Rutenberg
- Department of Radiation-Oncology, Mayo Clinic Florida, Mayo Clinic Cancer Center, Jacksonville, FL 32224, United States
| | - John Stauffer
- Department of Surgical Oncology, Hepatopancreatobiliary Surgery, Mayo Clinic Florida, Jacksonville, FL 32224, United States
| | - Jason Starr
- Division of Hematology-Oncology, Department of Medicine, Mayo Clinic Florida, Mayo Clinic Cancer Center, Jacksonville, FL 32224, United States
| | - Umair Majeed
- Division of Hematology-Oncology, Department of Medicine, Mayo Clinic Florida, Mayo Clinic Cancer Center, Jacksonville, FL 32224, United States
| | - Jeremy Jones
- Division of Hematology-Oncology, Department of Medicine, Mayo Clinic Florida, Mayo Clinic Cancer Center, Jacksonville, FL 32224, United States
| | - Mitesh Borad
- Division of Hematology-Oncology, Department of Medicine, Mayo Clinic Arizona, Mayo Clinic Cancer Center, Phoenix, AZ 85054, United States
| | - Hani Babiker
- Division of Hematology-Oncology, Department of Medicine, Mayo Clinic Florida, Mayo Clinic Cancer Center, Jacksonville, FL 32224, United States
| |
Collapse
|
3
|
Zhang H, Hu Z, Luo X, Wang Y, Wang Y, Liu T, Zhang Y, Chu L, Wang X, Zhen Y, Zhang J, Yu Y. ZmRop1 participates in maize defense response to the damage of Spodoptera frugiperda larvae through mediating ROS and soluble phenol production. PLANT DIRECT 2022; 6:e468. [PMID: 36540415 PMCID: PMC9751866 DOI: 10.1002/pld3.468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 11/05/2022] [Accepted: 11/07/2022] [Indexed: 06/17/2023]
Abstract
As plant-specific molecular switches, Rho-like GTPases (Rops) are vital for plant survival in response to biotic and abiotic stresses. However, their roles in plant defense response to phytophagous insect's damage are largely unknown. In this study, the expression levels of nine maize RAC family genes were analyzed after fall armyworm (FAW) larvae infestation. Among the analyzed genes, ZmRop1 was specifically and highly expressed, and its role in maize response to FAW larvae damage was studied. The results showed that upon FAW larvae infestation, salicylic acid and methyl jasmonate treatment ZmRop1 gene transcripts were all down-regulated. However, upon mechanical injury, the expression level of ZmRop1 was up-regulated. Overexpression of ZmRop1 gene in maize plants could improve maize plant resistance to FAW larvae damage. Conversely, silencing of ZmRop1 increased maize plant susceptibility to FAW larvae damage. The analysis of the potential anti-herbivore metabolites, showed that ZmRop1 promoted the enzyme activities of catalase, peroxidase and the expression levels of ZmCAT, ZmPOD, ZmRBOHA and ZmRBOHB, thereby enhancing the reactive oxygen species (ROS) production, including the content of O2- and H2O2. In addition, overexpression or silencing of ZmRop1 could have influence on the content of the total soluble phenol through mediating the activity of polyphenol oxidase. In summary, the results illuminated our understanding of how ZmRop1 participate in maize defense response to FAW larvae damage as a positive regulator through mediating ROS production and can be used as a reference for the green prevention and control of FAW larvae.
Collapse
Affiliation(s)
- Haoran Zhang
- College of AgricultureYangtze UniversityJingzhouChina
| | - Zongwei Hu
- College of AgricultureYangtze UniversityJingzhouChina
| | - Xincheng Luo
- College of Life SciencesYangtze UniversityJingzhouChina
| | - Yuxue Wang
- College of AgricultureYangtze UniversityJingzhouChina
| | - Yi Wang
- College of AgricultureYangtze UniversityJingzhouChina
| | - Ting Liu
- College of AgricultureYangtze UniversityJingzhouChina
| | - Yi Zhang
- College of AgricultureYangtze UniversityJingzhouChina
| | - Longyan Chu
- College of AgricultureYangtze UniversityJingzhouChina
| | | | - Yangya Zhen
- College of Life SciencesYangtze UniversityJingzhouChina
| | - Jianmin Zhang
- College of AgricultureYangtze UniversityJingzhouChina
| | - Yonghao Yu
- Guangxi Key Laboratory of Biology for Crop Diseases and Insect PestsNanningChina
| |
Collapse
|
4
|
Zhu MC, Li XM, Zhao N, Yang L, Zhang KQ, Yang JK. Regulatory Mechanism of Trap Formation in the Nematode-Trapping Fungi. J Fungi (Basel) 2022; 8:jof8040406. [PMID: 35448637 PMCID: PMC9031305 DOI: 10.3390/jof8040406] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Revised: 04/13/2022] [Accepted: 04/14/2022] [Indexed: 01/21/2023] Open
Abstract
Nematode-trapping (NT) fungi play a significant role in the biological control of plant- parasitic nematodes. NT fungi, as a predator, can differentiate into specialized structures called “traps” to capture, kill, and consume nematodes at a nutrient-deprived condition. Therefore, trap formation is also an important indicator that NT fungi transition from a saprophytic to a predacious lifestyle. With the development of gene knockout and multiple omics such as genomics, transcriptomics, and metabolomics, increasing studies have tried to investigate the regulation mechanism of trap formation in NT fungi. This review summarizes the potential regulatory mechanism of trap formation in NT fungi based on the latest findings in this field. Signaling pathways have been confirmed to play an especially vital role in trap formation based on phenotypes of various mutants and multi-omics analysis, and the involvement of small molecule compounds, woronin body, peroxisome, autophagy, and pH-sensing receptors in the formation of traps are also discussed. In addition, we also highlight the research focus for elucidating the mechanism underlying trap formation of NT fungi in the future.
Collapse
|
5
|
Classical RAS proteins are not essential for paradoxical ERK activation induced by RAF inhibitors. Proc Natl Acad Sci U S A 2022; 119:2113491119. [PMID: 35091470 PMCID: PMC8812530 DOI: 10.1073/pnas.2113491119] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/22/2021] [Indexed: 11/21/2022] Open
Abstract
RAF inhibitors unexpectedly induce ERK activation in normal and oncogenic RAS tumor cells, making them unsuitable for treating RAS-driven cancers. The precise mechanism of this paradox is not fully understood but is believed to be RAS dependent. In this study, we discovered that classical RAS proteins are not essential for RAF inhibitor-induced ERK activation in H/N/KRAS-less mouse embryonic fibroblasts. We further showed that the MRAS/SHOC2 complex is required for the classical RAS-independent paradoxical ERK activation. Our findings provide new insights into the mechanism of paradoxical ERK activation by RAF inhibitors, and they have important therapeutic implications for developing effective RAF inhibitors. RAF inhibitors unexpectedly induce ERK signaling in normal and tumor cells with elevated RAS activity. Paradoxical activation is believed to be RAS dependent. In this study, we showed that LY3009120, a pan-RAF inhibitor, can unexpectedly cause paradoxical ERK activation in KRASG12C-dependent lung cancer cell lines, when KRAS is inhibited by ARS1620, a KRASG12C inhibitor. Using H/N/KRAS-less mouse embryonic fibroblasts, we discovered that classical RAS proteins are not essential for RAF inhibitor-induced paradoxical ERK signaling. In their absence, RAF inhibitors can induce ERK phosphorylation, ERK target gene transcription, and cell proliferation. We further showed that the MRAS/SHOC2 complex is required for this process. This study highlights the complexity of the allosteric RAF regulation by RAF inhibitors, and the importance of other RAS-related proteins in this process.
Collapse
|
6
|
Liu X, Xie Z, Li S, He J, Cao S, Xiao Z. PRG-1 relieves pain and depressive-like behaviors in rats of bone cancer pain by regulation of dendritic spine in hippocampus. Int J Biol Sci 2021; 17:4005-4020. [PMID: 34671215 PMCID: PMC8495398 DOI: 10.7150/ijbs.59032] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 09/06/2021] [Indexed: 12/15/2022] Open
Abstract
Rationale: Pain and depression, which tend to occur simultaneously and share some common neural circuits and neurotransmitters, are highly prevalent complication in patients with advanced cancer. Exploring the underlying mechanisms is the cornerstone to prevent the comorbidity of chronic pain and depression in cancer patients. Plasticity-related gene 1 (PRG-1) protein regulates synaptic plasticity and brain functional reorganization during neuronal development or after cerebral lesion. Purinergic P2X7 receptor has been proposed as a therapeutic target for various pain and neurological disorders like depression in rodents. In this study, we investigated the roles of PRG-1 in the hippocampus in the comorbidity of pain and depressive-like behaviors in rats with bone cancer pain (BCP). Methods: The bone cancer pain rat model was established by intra-tibial cell inoculation of SHZ-88 mammary gland carcinoma cells. The animal pain behaviors were assessed by measuring the thermal withdrawal latency values by using radiant heat stimulation and mechanical withdrawal threshold by using electronic von Frey anesthesiometer, and depressive-like behavior was assessed by sucrose preference test and forced swim test. Alterations in the expression levels of PRG-1 and P2X7 receptor in hippocampus were separately detected by using western blot, immunofluorescence and immunohistochemistry analysis. The effects of intra-hippocampal injection of FTY720 (a PRG-1/PP2A interaction activator), PRG-1 overexpression or intra-hippocampal injection of A438079 (a selective competitive P2X7 receptor antagonist) were also observed. Results: Carcinoma intra-tibia injection caused thermal hyperalgesia, mechanical allodynia and depressive-like behaviors in rats, and also induced the deactivation of neurons and dendritic spine structural anomalies in the hippocampus. Western blot, immunofluorescence and immunohistochemistry analysis showed an increased expression of PRG-1 and P2X7 receptor in the hippocampus of BCP rats. Intra-hippocampal injection of FTY720 or A438079 attenuated both pain and depressive-like behaviors. Furthermore, overexpression of PRG-1 in hippocampus has similar analgesic efficacy to FTY720. In addition, they rescued neuron deactivation and dendritic spine anomalies. Conclusion: The results suggest that both PRG-1 and P2X7 receptor in the hippocampus play important roles in the development of pain and depressive-like behaviors in bone cancer condition in rats by dendritic spine regulation via P2X7R/PRG-1/PP2A pathway.
Collapse
Affiliation(s)
- Xingfeng Liu
- Guizhou Key Laboratory of Brain Science, Zunyi Medical University, Zunyi 563000, China.,Guizhou Key Laboratory of Anesthesia and Organ Protection, Zunyi Medical University, Zunyi 563000, China
| | - Zhuo Xie
- Graduate School, Zunyi Medical University, Zunyi 563000, China
| | - Site Li
- Graduate School, Zunyi Medical University, Zunyi 563000, China
| | - Jingxin He
- Graduate School, Zunyi Medical University, Zunyi 563000, China
| | - Song Cao
- Department of Pain Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, China
| | - Zhi Xiao
- Guizhou Key Laboratory of Brain Science, Zunyi Medical University, Zunyi 563000, China.,Guizhou Key Laboratory of Anesthesia and Organ Protection, Zunyi Medical University, Zunyi 563000, China
| |
Collapse
|
7
|
Zhou X, Shafique K, Sajid M, Ali Q, Khalili E, Javed MA, Haider MS, Zhou G, Zhu G. Era-like GTP protein gene expression in rice. BRAZ J BIOL 2021; 82:e250700. [PMID: 34259718 DOI: 10.1590/1519-6984.250700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 06/19/2021] [Indexed: 11/22/2022] Open
Abstract
The mutations are genetic changes in the genome sequences and have a significant role in biotechnology, genetics, and molecular biology even to find out the genome sequences of a cell DNA along with the viral RNA sequencing. The mutations are the alterations in DNA that may be natural or spontaneous and induced due to biochemical reactions or radiations which damage cell DNA. There is another cause of mutations which is known as transposons or jumping genes which can change their position in the genome during meiosis or DNA replication. The transposable elements can induce by self in the genome due to cellular and molecular mechanisms including hypermutation which caused the localization of transposable elements to move within the genome. The use of induced mutations for studying the mutagenesis in crop plants is very common as well as a promising method for screening crop plants with new and enhanced traits for the improvement of yield and production. The utilization of insertional mutations through transposons or jumping genes usually generates stable mutant alleles which are mostly tagged for the presence or absence of jumping genes or transposable elements. The transposable elements may be used for the identification of mutated genes in crop plants and even for the stable insertion of transposable elements in mutated crop plants. The guanine nucleotide-binding (GTP) proteins have an important role in inducing tolerance in rice plants to combat abiotic stress conditions.
Collapse
Affiliation(s)
- X Zhou
- Linyi University, College of Life Science, Linyi, Shandong, China
| | - K Shafique
- Government Sadiq College Women University, Department of Botany, Bahawalpur, Pakistan
| | - M Sajid
- University of Okara, Faculty of Life Sciences, Department of Biotechnology, Okara, Pakistan
| | - Q Ali
- University of Lahore, Institute of Molecular Biology and Biotechnology, Lahore, Pakistan
| | - E Khalili
- Tarbiat Modarres University, Faculty of Science, Department of Plant Science, Tehran, Iran
| | - M A Javed
- University of the Punjab Lahore, Department of Plant Breeding and Genetics, Lahore, Pakistan
| | - M S Haider
- University of the Punjab Lahore, Department of Plant Pathology, Lahore, Pakistan
| | - G Zhou
- Yangzhou University, The Ministry of Education of China, Joint International Research Laboratory of Agriculture and Agri-Product Safety, Yangzhou, Jiangsu, China
| | - G Zhu
- Yangzhou University, The Ministry of Education of China, Joint International Research Laboratory of Agriculture and Agri-Product Safety, Yangzhou, Jiangsu, China
| |
Collapse
|
8
|
Kilian LS, Voran J, Frank D, Rangrez AY. RhoA: a dubious molecule in cardiac pathophysiology. J Biomed Sci 2021; 28:33. [PMID: 33906663 PMCID: PMC8080415 DOI: 10.1186/s12929-021-00730-w] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 04/23/2021] [Indexed: 02/08/2023] Open
Abstract
The Ras homolog gene family member A (RhoA) is the founding member of Rho GTPase superfamily originally studied in cancer cells where it was found to stimulate cell cycle progression and migration. RhoA acts as a master switch control of actin dynamics essential for maintaining cytoarchitecture of a cell. In the last two decades, however, RhoA has been coined and increasingly investigated as an essential molecule involved in signal transduction and regulation of gene transcription thereby affecting physiological functions such as cell division, survival, proliferation and migration. RhoA has been shown to play an important role in cardiac remodeling and cardiomyopathies; underlying mechanisms are however still poorly understood since the results derived from in vitro and in vivo experiments are still inconclusive. Interestingly its role in the development of cardiomyopathies or heart failure remains largely unclear due to anomalies in the current data available that indicate both cardioprotective and deleterious effects. In this review, we aimed to outline the molecular mechanisms of RhoA activation, to give an overview of its regulators, and the probable mechanisms of signal transduction leading to RhoA activation and induction of downstream effector pathways and corresponding cellular responses in cardiac (patho)physiology. Furthermore, we discuss the existing studies assessing the presented results and shedding light on the often-ambiguous data. Overall, we provide an update of the molecular, physiological and pathological functions of RhoA in the heart and its potential in cardiac therapeutics.
Collapse
Affiliation(s)
- Lucia Sophie Kilian
- Department of Internal Medicine III (Cardiology, Angiology, Intensive Care), University Medical Center Kiel, Rosalind-Franklin Str. 12, 24105, Kiel, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, 24105, Kiel, Germany
| | - Jakob Voran
- Department of Internal Medicine III (Cardiology, Angiology, Intensive Care), University Medical Center Kiel, Rosalind-Franklin Str. 12, 24105, Kiel, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, 24105, Kiel, Germany
| | - Derk Frank
- Department of Internal Medicine III (Cardiology, Angiology, Intensive Care), University Medical Center Kiel, Rosalind-Franklin Str. 12, 24105, Kiel, Germany. .,DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, 24105, Kiel, Germany.
| | - Ashraf Yusuf Rangrez
- Department of Internal Medicine III (Cardiology, Angiology, Intensive Care), University Medical Center Kiel, Rosalind-Franklin Str. 12, 24105, Kiel, Germany. .,DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, 24105, Kiel, Germany. .,Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany.
| |
Collapse
|
9
|
Ayuzawa N, Fujita T. The Mineralocorticoid Receptor in Salt-Sensitive Hypertension and Renal Injury. J Am Soc Nephrol 2021; 32:279-289. [PMID: 33397690 PMCID: PMC8054893 DOI: 10.1681/asn.2020071041] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Hypertension and its comorbidities pose a major public health problem associated with disease-associated factors related to a modern lifestyle, such high salt intake or obesity. Accumulating evidence has demonstrated that aldosterone and its receptor, the mineralocorticoid receptor (MR), have crucial roles in the development of salt-sensitive hypertension and coexisting cardiovascular and renal injuries. Accordingly, clinical trials have repetitively shown the promising effects of MR blockers in these diseases. We and other researchers have identified novel mechanisms of MR activation involved in salt-sensitive hypertension and renal injury, including the obesity-derived overproduction of aldosterone and ligand-independent signaling. Moreover, recent advances in the analysis of cell-specific and context-dependent mechanisms of MR activation in various tissues-including a classic target of aldosterone, aldosterone-sensitive distal nephrons-are now providing new insights. In this review, we summarize recent updates to our understanding of aldosterone-MR signaling, focusing on its role in salt-sensitive hypertension and renal injury.
Collapse
Affiliation(s)
- Nobuhiro Ayuzawa
- Division of Clinical Epigenetics, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Toshiro Fujita
- Division of Clinical Epigenetics, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan,Shinshu University School of Medicine, Nagano, Japan,Research Center for Social Systems, Shinshu University, Nagano, Japan
| |
Collapse
|
10
|
Paez PA, Kolawole M, Taruselli MT, Ajith S, Dailey JM, Kee SA, Haque TT, Barnstein BO, McLeod JJA, Caslin HL, Kiwanuka KN, Fukuoka Y, Le QT, Schwartz LB, Straus DB, Gewirtz DA, Martin RK, Ryan JJ. Fluvastatin Induces Apoptosis in Primary and Transformed Mast Cells. J Pharmacol Exp Ther 2020; 374:104-112. [PMID: 32434944 DOI: 10.1124/jpet.119.264234] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 04/01/2020] [Indexed: 12/18/2022] Open
Abstract
Statin drugs are widely employed in the clinic to reduce serum cholesterol. Because of their hydroxymethylglutaryl coenzyme A reductase antagonism, statins also reduce isoprenyl lipids necessary for the membrane anchorage and signaling of small G-proteins in the Ras superfamily. We previously found that statins suppress immunoglobulin E (IgE)-mediated mast cell activation, suggesting these drugs might be useful in treating allergic disease. Although IgE-induced function is critical to allergic inflammation, mast cell proliferation and survival also impact atopic disease and mast cell neoplasia. In this study, we describe fluvastatin-mediated apoptosis in primary and transformed mast cells. An IC50 was achieved between 0.8 and 3.5 μM in both cell types, concentrations similar to the reported fluvastatin serum Cmax value. Apoptosis was correlated with reduced stem cell factor (SCF)-mediated signal transduction, mitochondrial dysfunction, and caspase activation. Complementing these data, we found that p53 deficiency or Bcl-2 overexpression reduced fluvastatin-induced apoptosis. We also noted evidence of cytoprotective autophagy in primary mast cells treated with fluvastatin. Finally, we found that intraperitoneal fluvastatin treatment reduced peritoneal mast cell numbers in vivo These findings offer insight into the mechanisms of mast cell survival and support the possible utility of statins in mast cell-associated allergic and neoplastic diseases. SIGNIFICANCE STATEMENT: Fluvastatin, a statin drug used to lower cholesterol, induces apoptosis in primary and transformed mast cells by antagonizing protein isoprenylation, effectively inhibiting stem cell factor (SCF)-induced survival signals. This drug may be an effective means of suppressing mast cell survival.
Collapse
Affiliation(s)
- Patrick A Paez
- Departments of Biology (P.A.P., E.M.K., M.T.T., S.A., J.M.D., S.A.K., T.T.H., B.O.B., J.J.A.M., H.L.C., K.N.K., Y.F., D. B.S., J.J.R.), Internal Medicine (Q.T.L., L.B.S.), Pharmacology and Toxicology (D.A.G.), and Microbiology and Immunology (R.K.M.), Virginia Commonwealth University, Richmond, Virginia
| | - Motunrayo Kolawole
- Departments of Biology (P.A.P., E.M.K., M.T.T., S.A., J.M.D., S.A.K., T.T.H., B.O.B., J.J.A.M., H.L.C., K.N.K., Y.F., D. B.S., J.J.R.), Internal Medicine (Q.T.L., L.B.S.), Pharmacology and Toxicology (D.A.G.), and Microbiology and Immunology (R.K.M.), Virginia Commonwealth University, Richmond, Virginia
| | - Marcela T Taruselli
- Departments of Biology (P.A.P., E.M.K., M.T.T., S.A., J.M.D., S.A.K., T.T.H., B.O.B., J.J.A.M., H.L.C., K.N.K., Y.F., D. B.S., J.J.R.), Internal Medicine (Q.T.L., L.B.S.), Pharmacology and Toxicology (D.A.G.), and Microbiology and Immunology (R.K.M.), Virginia Commonwealth University, Richmond, Virginia
| | - Siddarth Ajith
- Departments of Biology (P.A.P., E.M.K., M.T.T., S.A., J.M.D., S.A.K., T.T.H., B.O.B., J.J.A.M., H.L.C., K.N.K., Y.F., D. B.S., J.J.R.), Internal Medicine (Q.T.L., L.B.S.), Pharmacology and Toxicology (D.A.G.), and Microbiology and Immunology (R.K.M.), Virginia Commonwealth University, Richmond, Virginia
| | - Jordan M Dailey
- Departments of Biology (P.A.P., E.M.K., M.T.T., S.A., J.M.D., S.A.K., T.T.H., B.O.B., J.J.A.M., H.L.C., K.N.K., Y.F., D. B.S., J.J.R.), Internal Medicine (Q.T.L., L.B.S.), Pharmacology and Toxicology (D.A.G.), and Microbiology and Immunology (R.K.M.), Virginia Commonwealth University, Richmond, Virginia
| | - Sydney A Kee
- Departments of Biology (P.A.P., E.M.K., M.T.T., S.A., J.M.D., S.A.K., T.T.H., B.O.B., J.J.A.M., H.L.C., K.N.K., Y.F., D. B.S., J.J.R.), Internal Medicine (Q.T.L., L.B.S.), Pharmacology and Toxicology (D.A.G.), and Microbiology and Immunology (R.K.M.), Virginia Commonwealth University, Richmond, Virginia
| | - Tamara T Haque
- Departments of Biology (P.A.P., E.M.K., M.T.T., S.A., J.M.D., S.A.K., T.T.H., B.O.B., J.J.A.M., H.L.C., K.N.K., Y.F., D. B.S., J.J.R.), Internal Medicine (Q.T.L., L.B.S.), Pharmacology and Toxicology (D.A.G.), and Microbiology and Immunology (R.K.M.), Virginia Commonwealth University, Richmond, Virginia
| | - Brian O Barnstein
- Departments of Biology (P.A.P., E.M.K., M.T.T., S.A., J.M.D., S.A.K., T.T.H., B.O.B., J.J.A.M., H.L.C., K.N.K., Y.F., D. B.S., J.J.R.), Internal Medicine (Q.T.L., L.B.S.), Pharmacology and Toxicology (D.A.G.), and Microbiology and Immunology (R.K.M.), Virginia Commonwealth University, Richmond, Virginia
| | - Jamie Josephine Avila McLeod
- Departments of Biology (P.A.P., E.M.K., M.T.T., S.A., J.M.D., S.A.K., T.T.H., B.O.B., J.J.A.M., H.L.C., K.N.K., Y.F., D. B.S., J.J.R.), Internal Medicine (Q.T.L., L.B.S.), Pharmacology and Toxicology (D.A.G.), and Microbiology and Immunology (R.K.M.), Virginia Commonwealth University, Richmond, Virginia
| | - Heather L Caslin
- Departments of Biology (P.A.P., E.M.K., M.T.T., S.A., J.M.D., S.A.K., T.T.H., B.O.B., J.J.A.M., H.L.C., K.N.K., Y.F., D. B.S., J.J.R.), Internal Medicine (Q.T.L., L.B.S.), Pharmacology and Toxicology (D.A.G.), and Microbiology and Immunology (R.K.M.), Virginia Commonwealth University, Richmond, Virginia
| | - Kasalina N Kiwanuka
- Departments of Biology (P.A.P., E.M.K., M.T.T., S.A., J.M.D., S.A.K., T.T.H., B.O.B., J.J.A.M., H.L.C., K.N.K., Y.F., D. B.S., J.J.R.), Internal Medicine (Q.T.L., L.B.S.), Pharmacology and Toxicology (D.A.G.), and Microbiology and Immunology (R.K.M.), Virginia Commonwealth University, Richmond, Virginia
| | - Yoshihiro Fukuoka
- Departments of Biology (P.A.P., E.M.K., M.T.T., S.A., J.M.D., S.A.K., T.T.H., B.O.B., J.J.A.M., H.L.C., K.N.K., Y.F., D. B.S., J.J.R.), Internal Medicine (Q.T.L., L.B.S.), Pharmacology and Toxicology (D.A.G.), and Microbiology and Immunology (R.K.M.), Virginia Commonwealth University, Richmond, Virginia
| | - Quang T Le
- Departments of Biology (P.A.P., E.M.K., M.T.T., S.A., J.M.D., S.A.K., T.T.H., B.O.B., J.J.A.M., H.L.C., K.N.K., Y.F., D. B.S., J.J.R.), Internal Medicine (Q.T.L., L.B.S.), Pharmacology and Toxicology (D.A.G.), and Microbiology and Immunology (R.K.M.), Virginia Commonwealth University, Richmond, Virginia
| | - Lawrence B Schwartz
- Departments of Biology (P.A.P., E.M.K., M.T.T., S.A., J.M.D., S.A.K., T.T.H., B.O.B., J.J.A.M., H.L.C., K.N.K., Y.F., D. B.S., J.J.R.), Internal Medicine (Q.T.L., L.B.S.), Pharmacology and Toxicology (D.A.G.), and Microbiology and Immunology (R.K.M.), Virginia Commonwealth University, Richmond, Virginia
| | - David B Straus
- Departments of Biology (P.A.P., E.M.K., M.T.T., S.A., J.M.D., S.A.K., T.T.H., B.O.B., J.J.A.M., H.L.C., K.N.K., Y.F., D. B.S., J.J.R.), Internal Medicine (Q.T.L., L.B.S.), Pharmacology and Toxicology (D.A.G.), and Microbiology and Immunology (R.K.M.), Virginia Commonwealth University, Richmond, Virginia
| | - David A Gewirtz
- Departments of Biology (P.A.P., E.M.K., M.T.T., S.A., J.M.D., S.A.K., T.T.H., B.O.B., J.J.A.M., H.L.C., K.N.K., Y.F., D. B.S., J.J.R.), Internal Medicine (Q.T.L., L.B.S.), Pharmacology and Toxicology (D.A.G.), and Microbiology and Immunology (R.K.M.), Virginia Commonwealth University, Richmond, Virginia
| | - Rebecca K Martin
- Departments of Biology (P.A.P., E.M.K., M.T.T., S.A., J.M.D., S.A.K., T.T.H., B.O.B., J.J.A.M., H.L.C., K.N.K., Y.F., D. B.S., J.J.R.), Internal Medicine (Q.T.L., L.B.S.), Pharmacology and Toxicology (D.A.G.), and Microbiology and Immunology (R.K.M.), Virginia Commonwealth University, Richmond, Virginia
| | - John J Ryan
- Departments of Biology (P.A.P., E.M.K., M.T.T., S.A., J.M.D., S.A.K., T.T.H., B.O.B., J.J.A.M., H.L.C., K.N.K., Y.F., D. B.S., J.J.R.), Internal Medicine (Q.T.L., L.B.S.), Pharmacology and Toxicology (D.A.G.), and Microbiology and Immunology (R.K.M.), Virginia Commonwealth University, Richmond, Virginia
| |
Collapse
|
11
|
Osanai K. Rab38 Mutation and the Lung Phenotype. Int J Mol Sci 2018; 19:E2203. [PMID: 30060521 PMCID: PMC6122074 DOI: 10.3390/ijms19082203] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Revised: 07/21/2018] [Accepted: 07/23/2018] [Indexed: 12/12/2022] Open
Abstract
Rab38 is highly expressed in alveolar type II cells, melanocytes, and platelets. These cells are specifically-differentiated cells and contain characteristic intracellular organelles called lysosome-related organelles, i.e., lamellar bodies in alveolar type II cells, melanosomes in melanocytes, and dense granules in platelets. There are Rab38-mutant rodents, i.e., chocolate mice and Ruby rats. While chocolate mice only show oculocutaneous albinism, Ruby rats show oculocutaneous albinism and prolonged bleeding time and, hence, are a rat model of Hermansky-Pudlak syndrome (HPS). Most patients with HPS suffer from fatal interstitial pneumonia by middle age. The lungs of both chocolate mice and Ruby rats show remarkably increased amounts of lung surfactant and conspicuously enlarged lysosome-related organelles, i.e., lamellar bodies, which are also characteristic of the lungs in human HPS. There are 16 mutant HPS-mouse strains, of which ten mutant genes have been identified to be causative in patients with HPS thus far. The gene products of eight of the ten genes constitute one of the three protein complexes, i.e., biogenesis of lysosome-related organelle complex-1, -2, -3 (BLOC-1, -2, -3). Patients with HPS of the mutant BLOC-3 genotype develop interstitial pneumonia. Recently, BLOC-3 has been elucidated to be a guanine nucleotide exchange factor for Rab38. Growing evidence suggests that Rab38 is an additional candidate gene of human HPS that displays the lung phenotype.
Collapse
Affiliation(s)
- Kazuhiro Osanai
- Department of Life Science, Medical Research Institute, Kanazawa Medical University, 1-1 Uchinada-Daigaku, Kahokugun, Ishikawa 920-0293, Japan.
- Department of Respiratory Medicine, Kanazawa Medical University, 1-1 Uchinada-Daigaku, Kahokugun, Ishikawa 920-0293, Japan.
| |
Collapse
|
12
|
Peterson YK, Luttrell LM. The Diverse Roles of Arrestin Scaffolds in G Protein-Coupled Receptor Signaling. Pharmacol Rev 2017. [PMID: 28626043 DOI: 10.1124/pr.116.013367] [Citation(s) in RCA: 313] [Impact Index Per Article: 39.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The visual/β-arrestins, a small family of proteins originally described for their role in the desensitization and intracellular trafficking of G protein-coupled receptors (GPCRs), have emerged as key regulators of multiple signaling pathways. Evolutionarily related to a larger group of regulatory scaffolds that share a common arrestin fold, the visual/β-arrestins acquired the capacity to detect and bind activated GPCRs on the plasma membrane, which enables them to control GPCR desensitization, internalization, and intracellular trafficking. By acting as scaffolds that bind key pathway intermediates, visual/β-arrestins both influence the tonic level of pathway activity in cells and, in some cases, serve as ligand-regulated scaffolds for GPCR-mediated signaling. Growing evidence supports the physiologic and pathophysiologic roles of arrestins and underscores their potential as therapeutic targets. Circumventing arrestin-dependent GPCR desensitization may alleviate the problem of tachyphylaxis to drugs that target GPCRs, and find application in the management of chronic pain, asthma, and psychiatric illness. As signaling scaffolds, arrestins are also central regulators of pathways controlling cell growth, migration, and survival, suggesting that manipulating their scaffolding functions may be beneficial in inflammatory diseases, fibrosis, and cancer. In this review we examine the structure-function relationships that enable arrestins to perform their diverse roles, addressing arrestin structure at the molecular level, the relationship between arrestin conformation and function, and sites of interaction between arrestins, GPCRs, and nonreceptor-binding partners. We conclude with a discussion of arrestins as therapeutic targets and the settings in which manipulating arrestin function might be of clinical benefit.
Collapse
Affiliation(s)
- Yuri K Peterson
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy (Y.K.P.), and Departments of Medicine and Biochemistry and Molecular Biology (L.M.L.), Medical University of South Carolina, Charleston, South Carolina; and Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina (L.M.L.)
| | - Louis M Luttrell
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy (Y.K.P.), and Departments of Medicine and Biochemistry and Molecular Biology (L.M.L.), Medical University of South Carolina, Charleston, South Carolina; and Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina (L.M.L.)
| |
Collapse
|
13
|
Park S, Jang H, Kim BS, Hwang C, Jeong GS, Park Y. Directional migration of mesenchymal stem cells under an SDF-1α gradient on a microfluidic device. PLoS One 2017; 12:e0184595. [PMID: 28886159 PMCID: PMC5590985 DOI: 10.1371/journal.pone.0184595] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2017] [Accepted: 08/25/2017] [Indexed: 12/13/2022] Open
Abstract
Homing of peripheral stem cells is regulated by one of the most representative homing factors, stromal cell-derived factor 1 alpha (SDF-1α), which specifically binds to the plasma membrane receptor CXCR4 of mesenchymal stem cells (MSCs) in order to initiate the signaling pathways that lead to directional migration and homing of stem cells. This complex homing process and directional migration of stem cells have been mimicked on a microfluidic device that is capable of generating a chemokine gradient within the collagen matrix and embedding endothelial cell (EC) monolayers to mimic blood vessels. On the microfluidic device, stem cells showed directional migration toward the higher concentration of SDF-1α, whereas treatment with the CXCR4 antagonist AMD3100 caused loss of directionality of stem cells. Furthermore, inhibition of stem cell's main migratory signaling pathways, Rho-ROCK and Rac pathways, caused blockage of actomyosin and lamellipodia formation, decreasing the migration distance but maintaining directionality. Stem cell homing regulated by SDF-1α caused directional migration of stem cells, while the migratory ability was affected by the activation of migration-related signaling pathways.
Collapse
Affiliation(s)
- Siwan Park
- Department of Biomedical Engineering, Biomedical Science of Brain Korea 21, College of Medicine, Korea University, Seoul, Korea
| | - Hwanseok Jang
- Department of Biomedical Engineering, Biomedical Science of Brain Korea 21, College of Medicine, Korea University, Seoul, Korea
| | - Byung Soo Kim
- Department of Biomedical Science, Graduate School of Medicine, Korea University, Seoul Korea
| | - Changmo Hwang
- Biomedical Engineering Research Center, Asan Institute for Life Sciences, Asan Medical Center, Seoul, Korea
| | - Gi Seok Jeong
- Biomedical Engineering Research Center, Asan Institute for Life Sciences, Asan Medical Center, Seoul, Korea
- * E-mail: (YP); (GSJ)
| | - Yongdoo Park
- Department of Biomedical Engineering, Biomedical Science of Brain Korea 21, College of Medicine, Korea University, Seoul, Korea
- * E-mail: (YP); (GSJ)
| |
Collapse
|
14
|
Control of astrocyte morphology by Rho GTPases. Brain Res Bull 2017; 136:44-53. [PMID: 28502648 DOI: 10.1016/j.brainresbull.2017.05.003] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 05/05/2017] [Accepted: 05/10/2017] [Indexed: 12/15/2022]
Abstract
Astrocytes modulate and support neuronal and synapse function via numerous mechanisms that often rely on diffusion of signalling molecules, ions or metabolites through extracellular space. As a consequence, the spatial arrangement and the distance between astrocyte processes and neuronal structures are of functional importance. Likewise, changes of astrocyte structure will affect the ability of astrocytes to interact with neurons. In contrast to neurons, where rapid morphology changes are critically involved in many aspects of physiological brain function, a role of astrocyte restructuring in brain physiology is only beginning to emerge. In neurons, small GTPases of the Rho family are powerful initiators and modulators of structural changes. Less is known about the functional significance of these signalling molecules in astrocytes. Here, we review recent experimental evidence for the role of RhoA, Cdc42 and Rac1 in controlling dynamic astrocyte morphology as well as experimental tools and analytical approaches for studying astrocyte morphology changes.
Collapse
|
15
|
Yang A, Zhang WH. A Small GTPase, OsRab6a, is Involved in the Regulation of Iron Homeostasis in Rice. PLANT & CELL PHYSIOLOGY 2016; 57:1271-80. [PMID: 27257291 DOI: 10.1093/pcp/pcw073] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Accepted: 04/04/2016] [Indexed: 05/16/2023]
Abstract
Plants have evolved two distinct strategies to acquire iron (Fe) from soils. However, the regulatory mechanisms underlying the Fe acquisition remain largely elusive. There is emerging evidence that small GTPases are involved in the responses of plants to environmental cues. Here, we identified a gene encoding a small GTPase, OsRab6a, in rice and characterized its role in Fe acquisition by generating transgenic rice plants with overexpression and knockdown of OsRab6a OsRab6a shared conserved functional domains with other known members of the Rab subfamily and localized ubiquitously in the cytoplasm and nucleus. The expression of OsRab6a was rapidly and transiently up-regulated by Fe deficiency. No differences in growth and development among the OsRab6a-overexpression, OsRab6a-RNAi (RNA interference) and wild-type plants were detected when grown in Fe-sufficient medium. However, overexpression of OsRab6a in rice plants conferred greater tolerance to Fe deficiency than RNAi and wild-type plants, as evidenced by higher seedling height, and greater biomass, Chl contents and Fe concentrations in shoots, roots and grains in the overexpression lines than wild-type and RNAi plants. Moreover, the overexpression lines exhibited larger root systems than wild-type and RNAi plants in Fe-deficient medium. Exposure to Fe-deficient medium led to up-regulation of OsIRO2, OsIRT1, OsNAS1 and OsNAS2 in both wild-type and transgenic rice plants, with the magnitude of up-regulation positively correlated with the expression levels of OsRab6a These results may suggest that OsRab6a plays an important role in the regulation of Fe acquisition in rice plants by modulating physiological processes involved in Fe acquisition and root system architecture in response to Fe-deficient medium.
Collapse
Affiliation(s)
- An Yang
- State Key Laboratory of Vegetation and Environmental Change, Institute of Botany, the Chinese Academy of Sciences, Beijing 100093, PR China
| | - Wen-Hao Zhang
- Research Network of Global Change Biology, Beijing Institutes of Life Sciences, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
16
|
Choi JY, Shin YC, Yoon JH, Kim CM, Lee JH, Jeon JH, Park HH. Molecular mechanism of constitutively active Rab11A was revealed by crystal structure of Rab11A S20V. FEBS Lett 2016; 590:819-27. [PMID: 26879265 DOI: 10.1002/1873-3468.12100] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 02/02/2016] [Accepted: 02/05/2016] [Indexed: 11/06/2022]
Abstract
Rab11A is a small GTP-binding protein involved in the regulation of vesicle trafficking during recycling of endosomes. Substitution of S20 to V (S20V) at Rab11A inhibits the GTP hydrolysis activity of Rab11A. This mutation is known to be constitutively in an active form. Here, we report the crystal structure of the human Rab11A S20V mutant form complexed with GTP at a resolution of 2.4 Å. Without adding any substrate, Rab11A contained non-hydrolyzed natural substrate GTP in the nucleotide binding pocket with Mg(2+). In our observations, substituted V20 of Rab11A was found to interfere with proper localization of the water molecule, which mediated GTP hydrolysis, resulting in GTP being locked in an active form of Rab11A S20V.
Collapse
Affiliation(s)
- Jae Young Choi
- School of Biotechnology and Graduate School of Biochemistry at Yeungnam University, Gyeongsan, South Korea
| | - Young-Cheul Shin
- Department of Physiology, Department of Biomedical Sciences, and Institute of Dermatological Science, Seoul National University College of Medicine, South Korea
| | - Jong Hwan Yoon
- School of Biotechnology and Graduate School of Biochemistry at Yeungnam University, Gyeongsan, South Korea
| | - Chang Min Kim
- School of Biotechnology and Graduate School of Biochemistry at Yeungnam University, Gyeongsan, South Korea
| | - Jun Hyuck Lee
- Division of Polar Life Sciences, Korea Polar Research Institute, Inchon, South Korea
| | - Ju-Hong Jeon
- Department of Physiology, Department of Biomedical Sciences, and Institute of Dermatological Science, Seoul National University College of Medicine, South Korea
| | - Hyun Ho Park
- School of Biotechnology and Graduate School of Biochemistry at Yeungnam University, Gyeongsan, South Korea
| |
Collapse
|
17
|
Shin YC, Kim CM, Choi JY, Jeon JH, Park HH. Occupation of nucleotide in the binding pocket is critical to the stability of Rab11A. Protein Expr Purif 2016; 120:153-9. [PMID: 26767484 DOI: 10.1016/j.pep.2016.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 01/04/2016] [Accepted: 01/04/2016] [Indexed: 11/25/2022]
Abstract
The Ras superfamily of small G proteins is a family of guanosine triphosphatases (GTPases) and each GTPase has conserved amino acid sequences in the enzymatic active site that are responsible for specific interactions with GDP and GTP molecules. Rab GTPases, which belong to the Ras superfamily, are key regulators of intracellular vesicle trafficking via the recruitment of effector molecules. Here, we purified wild type, active mutant and inactive mutant of Rab11A. In this process, we found that the inactive mutant (Rab11A S25N) had low stability compared with wild type and other mutants. Further analysis revealed that the stability of Rab11A S25N is dependent on the occupation of GDP in the nucleotide binding pocket of the protein. We found that the stability of Rab11A S25N is affected by the presence of GDP, not other nucleotides, and is independent of pH or salt in FPLC buffer. Our results provide a better understanding of how GTPase can be stable under in vitro conditions without effector proteins and how proper substrate/cofactor coordination is crucial to the stability of Rab11A. Successful purification and proposed purification methods will provide a valuable guide for investigation of other small GTPase proteins.
Collapse
Affiliation(s)
- Young-Cheul Shin
- Department of Physiology and Biomedical Sciences, Institute of Human-Environment Interface Biology, Seoul National University College of Medicine, Seoul 110-799, South Korea
| | - Chang Min Kim
- School of Biotechnology and Graduate School of Biochemistry at Yeungnam University, Gyeongsan 38541, South Korea
| | - Jae Young Choi
- School of Biotechnology and Graduate School of Biochemistry at Yeungnam University, Gyeongsan 38541, South Korea
| | - Ju-Hong Jeon
- Department of Physiology and Biomedical Sciences, Institute of Human-Environment Interface Biology, Seoul National University College of Medicine, Seoul 110-799, South Korea.
| | - Hyun Ho Park
- School of Biotechnology and Graduate School of Biochemistry at Yeungnam University, Gyeongsan 38541, South Korea.
| |
Collapse
|
18
|
Görtzen J, Schierwagen R, Bierwolf J, Klein S, Uschner FE, van der Ven PF, Fürst DO, Strassburg CP, Laleman W, Pollok JM, Trebicka J. Interplay of Matrix Stiffness and c-SRC in Hepatic Fibrosis. Front Physiol 2015; 6:359. [PMID: 26696895 PMCID: PMC4667086 DOI: 10.3389/fphys.2015.00359] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 11/16/2015] [Indexed: 01/06/2023] Open
Abstract
Introduction: In liver fibrosis activation of hepatic stellate cells (HSC) comprises phenotypical change into profibrotic and myofibroplastic cells with increased contraction and secretion of extracellular matrix (ECM) proteins. The small GTPase RhoA orchestrates cytoskeleton formation, migration, and mobility via non-receptor tyrosine-protein kinase c-SRC (cellular sarcoma) in different cells. Furthermore, RhoA and its downstream effector Rho-kinase also play a crucial role in hepatic stellate cells and hepatic fibrogenesis. Matrix stiffness promotes HSC activation via cytoskeleton modulation. This study investigated the interaction of c-SRC and RhoA under different matrix stiffness conditions. Methods: Liver fibrosis was induced in rats using bile duct ligation (BDL), thioacetamide (TAA) or carbon tetrachloride (CCl4) models. mRNA levels of albumin, PDGF-R, RHOA, COL1A1, and αSMA were analyzed via qRT-PCR. Western Blots using phospho-specific antibodies against p-c-SRC418 and p-c-SRC530 analyzed the levels of activating and inactivating c-SRC, respectively. LX2 cells and hepatocytes were cultured on acrylamide gels of 1 and 12 kPa or on plastic to mimic non-fibrotic, fibrotic, or cirrhotic environments then exposed to SRC-inhibitor PP2. Overexpression of RhoA was performed by transfection using RhoA-plasmids. Additionally, samples from cirrhotic patients and controls were collected at liver transplantations and tumor resections were analyzed for RhoA and c-SRC protein expression by Western Blot. Results: Transcription of albumin and RhoA was decreased, whereas transcription and activation of c-SRC was increased in hepatocytes cultured on 12 kPa compared to 1 kPa gels. LX2 cells cultured on 12 kPa gels showed upregulation of RHOA, COL1A1, and αSMA mRNA levels. Inhibition of c-SRC by PP2 in LX2 cells led to an increase in COL1A1 and αSMA most prominently in 12 kPa gels. In LX2 cells with RhoA overexpression, c-SRC inhibition by PP2 failed to improve fibrosis. RhoA expression was significantly elevated in human and experimental liver fibrosis, while c-SRC was inactivated. Conclusions: This study shows that c-SRC is inactive in activated myofibroblast-like HSC in liver cirrhosis. Inactivation of c-SRC is mediated by a crosstalk with RhoA upon hepatic stellate cell activation and fibrosis progression.
Collapse
Affiliation(s)
- Jan Görtzen
- Department of Internal Medicine I, University of Bonn Bonn, Germany
| | | | - Jeanette Bierwolf
- Department of General, Visceral, Thoracic, and Vascular Surgery, University of Bonn Bonn, Germany
| | - Sabine Klein
- Department of Internal Medicine I, University of Bonn Bonn, Germany
| | - Frank E Uschner
- Department of Internal Medicine I, University of Bonn Bonn, Germany
| | - Peter F van der Ven
- Department of Molecular Cell Biology, Institute for Cell Biology, University of Bonn Bonn, Germany
| | - Dieter O Fürst
- Department of Molecular Cell Biology, Institute for Cell Biology, University of Bonn Bonn, Germany
| | | | - Wim Laleman
- Department of Internal Medicine, University Hospital Gasthuisberg Leuven, Belgium
| | - Jörg-Matthias Pollok
- Department of General, Visceral, Thoracic, and Vascular Surgery, University of Bonn Bonn, Germany
| | - Jonel Trebicka
- Department of Internal Medicine I, University of Bonn Bonn, Germany ; Faculty of Health Sciences, University of Southern Denmark Odense, Denmark
| |
Collapse
|
19
|
Ouellette MH, Martin E, Lacoste-Caron G, Hamiche K, Jenna S. Spatial control of active CDC-42 during collective migration of hypodermal cells in Caenorhabditis elegans. J Mol Cell Biol 2015; 8:313-27. [PMID: 26578656 DOI: 10.1093/jmcb/mjv062] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Accepted: 08/12/2015] [Indexed: 12/11/2022] Open
Abstract
Collective epithelial cell migration requires the maintenance of cell-cell junctions while enabling the generation of actin-rich protrusions at the leading edge of migrating cells. Ventral enclosure of Caenorhabditis elegans embryos depends on the collective migration of anterior-positioned leading hypodermal cells towards the ventral midline where they form new junctions with their contralateral neighbours. In this study, we characterized the zygotic function of RGA-7/SPV-1, a CDC-42/Cdc42 and RHO-1/RhoA-specific Rho GTPase-activating protein, which controls the formation of actin-rich protrusions at the leading edge of leading hypodermal cells and the formation of new junctions between contralateral cells. We show that RGA-7 controls these processes in an antagonistic manner with the CDC-42's effector WSP-1/N-WASP and the CDC-42-binding proteins TOCA-1/2/TOCA1. RGA-7 is recruited to spatially distinct locations at junctions between adjacent leading cells, where it promotes the accumulation of clusters of activated CDC-42. It also inhibits the spreading of these clusters towards the leading edge of the junctions and regulates their accumulation and distribution at new junctions formed between contralateral leading cells. Our study suggests that RGA-7 controls collective migration and junction formation between epithelial cells by spatially restricting active CDC-42 within cell-cell junctions.
Collapse
Affiliation(s)
- Marie-Hélène Ouellette
- Department of Chemistry, Pharmaqam, Biomed, Université du Québec à Montréal, Montréal, Québec, Canada
| | - Emmanuel Martin
- Department of Chemistry, Pharmaqam, Biomed, Université du Québec à Montréal, Montréal, Québec, Canada
| | - Germain Lacoste-Caron
- Department of Chemistry, Pharmaqam, Biomed, Université du Québec à Montréal, Montréal, Québec, Canada
| | - Karim Hamiche
- Department of Chemistry, Pharmaqam, Biomed, Université du Québec à Montréal, Montréal, Québec, Canada
| | - Sarah Jenna
- Department of Chemistry, Pharmaqam, Biomed, Université du Québec à Montréal, Montréal, Québec, Canada
| |
Collapse
|
20
|
Kim CM, Choi JY, Yoon JH, Park HH. Purification, crystallization and X-ray crystallographic analysis of human RAB11(S20V), a constitutively active GTP-binding form. ACTA CRYSTALLOGRAPHICA SECTION F-STRUCTURAL BIOLOGY COMMUNICATIONS 2015; 71:1247-50. [PMID: 26457514 DOI: 10.1107/s2053230x15015447] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 08/18/2015] [Indexed: 11/10/2022]
Abstract
RAB11, a member of the Ras superfamily of small G proteins, is involved in the regulation of vesicle trafficking during endosome recycling. Substitution of Ser20 by Val20 in Rab11 [RAB11(S20V)] inhibits its GTP hydrolysis activity and produces a constitutively active GTP-binding form. In this study, the RAB11(S20V) mutant was overexpressed in Escherichia coli with an engineered C-terminal His tag. RAB11(S20V) was then purified to homogeneity and was crystallized at 293 K. X-ray diffraction data were collected to a resolution of 2.4 Å from a crystal belonging to space group I4, with unit-cell parameters a = 74.11, b = 74.11, c = 149.44 Å. The asymmetric unit was estimated to contain two molecules of RAB11(S20V).
Collapse
Affiliation(s)
- Chang Min Kim
- Department of Biochemistry, Yeungnam University, Gyeongsan, Republic of Korea
| | - Jae Young Choi
- Department of Biochemistry, Yeungnam University, Gyeongsan, Republic of Korea
| | - Jong Hwan Yoon
- Department of Biochemistry, Yeungnam University, Gyeongsan, Republic of Korea
| | - Hyun Ho Park
- Department of Biochemistry, Yeungnam University, Gyeongsan, Republic of Korea
| |
Collapse
|
21
|
Soltan Ghoraie L, Burkowski F, Zhu M. Using kernelized partial canonical correlation analysis to study directly coupled side chains and allostery in small G proteins. Bioinformatics 2015; 31:i124-32. [PMID: 26072474 PMCID: PMC4765857 DOI: 10.1093/bioinformatics/btv241] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Motivation: Inferring structural dependencies among a protein’s side chains helps us understand their coupled motions. It is known that coupled fluctuations can reveal pathways of communication used for information propagation in a molecule. Side-chain conformations are commonly represented by multivariate angular variables, but existing partial correlation methods that can be applied to this inference task are not capable of handling multivariate angular data. We propose a novel method to infer direct couplings from this type of data, and show that this method is useful for identifying functional regions and their interactions in allosteric proteins. Results: We developed a novel extension of canonical correlation analysis (CCA), which we call ‘kernelized partial CCA’ (or simply KPCCA), and used it to infer direct couplings between side chains, while disentangling these couplings from indirect ones. Using the conformational information and fluctuations of the inactive structure alone for allosteric proteins in the Ras and other Ras-like families, our method identified allosterically important residues not only as strongly coupled ones but also in densely connected regions of the interaction graph formed by the inferred couplings. Our results were in good agreement with other empirical findings. By studying distinct members of the Ras, Rho and Rab sub-families, we show further that KPCCA was capable of inferring common allosteric characteristics in the small G protein super-family. Availability and implementation:https://github.com/lsgh/ismb15 Contact:lsoltang@uwaterloo.ca
Collapse
Affiliation(s)
- Laleh Soltan Ghoraie
- Department of Computer Science and Department of Statistics and Actuarial Science, University of Waterloo, Waterloo, ON, Canada
| | - Forbes Burkowski
- Department of Computer Science and Department of Statistics and Actuarial Science, University of Waterloo, Waterloo, ON, Canada
| | - Mu Zhu
- Department of Computer Science and Department of Statistics and Actuarial Science, University of Waterloo, Waterloo, ON, Canada
| |
Collapse
|
22
|
Shigella flexneri regulation of ARF6 activation during bacterial entry via an IpgD-mediated positive feedback loop. mBio 2015; 6:e02584. [PMID: 25736891 PMCID: PMC4358011 DOI: 10.1128/mbio.02584-14] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
UNLABELLED Entry into cells is critical for virulence of the human bacterial pathogens Shigella spp. Shigella spp. induce membrane ruffle formation and macropinocytic uptake, but the events instigating this process are incompletely understood. The host small GTPase ADP-ribosylation factor 6 (ARF6) functions in membrane trafficking at the plasma membrane and activates membrane ruffle formation. We demonstrate that ARF6 is required for efficient Shigella flexneri entry, is activated by S. flexneri dependent on the phosphatase activity of the type III secreted effector IpgD, and depends on cytohesin guanine nucleotide exchange factors (GEFs) for recruitment to entry sites. The cytohesin GEF ARF nucleotide binding site opener (ARNO) is recruited to these sites, also dependent on IpgD phosphatase activity. ARNO recruitment is independent of ARF6, indicating that, in addition to the described recruitment of ARNO by ARF6, ARNO is recruited upstream of ARF6. Our data provide evidence that ARF6, IpgD, phosphoinositide species, and ARNO constitute a previously undescribed positive feedback loop that amplifies ARF6 activation at bacterial entry sites, thereby promoting efficient S. flexneri uptake. IMPORTANCE Shigella spp. cause diarrhea and dysentery by infection of epithelial cells in the human colon. Critical to disease is the ability of Shigella to enter into cells, yet the mechanisms involved in entry are incompletely understood. We demonstrate that the small GTPase ADP-ribosylation factor 6 (ARF6) is required for efficient cellular entry of Shigella flexneri and that activation of ARF6 depends on the phosphatase activity of the Shigella protein IpgD, which is introduced into cells via the bacterial type III secretion system. We further show that IpgD phosphatase activity is required for recruitment of the ARF6 guanine nucleotide exchange factor (GEF) ARF nucleotide binding site opener (ARNO) to bacterial entry sites and that ARNO lies upstream of ARF6 activation. These relationships define a positive feedback loop that contributes to activation of ARF6 at S. flexneri entry sites and leads to local amplification of signals that promote bacterial entry.
Collapse
|
23
|
Ong ST, Freeley M, Skubis-Zegadło J, Fazil MHUT, Kelleher D, Fresser F, Baier G, Verma NK, Long A. Phosphorylation of Rab5a protein by protein kinase Cϵ is crucial for T-cell migration. J Biol Chem 2014; 289:19420-34. [PMID: 24872409 DOI: 10.1074/jbc.m113.545863] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Rab GTPases control membrane traffic and receptor-mediated endocytosis. Within this context, Rab5a plays an important role in the spatial regulation of intracellular transport and signal transduction processes. Here, we report a previously uncharacterized role for Rab5a in the regulation of T-cell motility. We show that Rab5a physically associates with protein kinase Cϵ (PKCϵ) in migrating T-cells. After stimulation of T-cells through the integrin LFA-1 or the chemokine receptor CXCR4, Rab5a is phosphorylated on an N-terminal Thr-7 site by PKCϵ. Both Rab5a and PKCϵ dynamically interact at the centrosomal region of migrating cells, and PKCϵ-mediated phosphorylation on Thr-7 regulates Rab5a trafficking to the cell leading edge. Furthermore, we demonstrate that Rab5a Thr-7 phosphorylation is functionally necessary for Rac1 activation, actin rearrangement, and T-cell motility. We present a novel mechanism by which a PKCϵ-Rab5a-Rac1 axis regulates cytoskeleton remodeling and T-cell migration, both of which are central for the adaptive immune response.
Collapse
Affiliation(s)
- Seow Theng Ong
- From the From the Department of Clinical Medicine, Institute of Molecular Medicine, Trinity College Dublin, Dublin 8, Ireland
| | - Michael Freeley
- From the From the Department of Clinical Medicine, Institute of Molecular Medicine, Trinity College Dublin, Dublin 8, Ireland
| | - Joanna Skubis-Zegadło
- From the From the Department of Clinical Medicine, Institute of Molecular Medicine, Trinity College Dublin, Dublin 8, Ireland, Department of Applied Pharmacy and Bioengineering, Medical University of Warsaw, 02-091 Warsaw, Poland
| | | | - Dermot Kelleher
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 637553, Faculty of Medicine, Imperial College London, Exhibition Road, London SW7 2AZ, United Kingdom, and
| | - Friedrich Fresser
- the Department of Medical Genetics, Molecular and Clinical Pharmacology, Innsbruck Medical University, A-6020 Innsbruck, Austria
| | - Gottfried Baier
- the Department of Medical Genetics, Molecular and Clinical Pharmacology, Innsbruck Medical University, A-6020 Innsbruck, Austria
| | - Navin Kumar Verma
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 637553,
| | - Aideen Long
- From the From the Department of Clinical Medicine, Institute of Molecular Medicine, Trinity College Dublin, Dublin 8, Ireland,
| |
Collapse
|
24
|
Barbar A, Couture M, Sen SE, Béliveau C, Nisole A, Bipfubusa M, Cusson M. Cloning, expression and characterization of an insect geranylgeranyl diphosphate synthase from Choristoneura fumiferana. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2013; 43:947-958. [PMID: 23907071 DOI: 10.1016/j.ibmb.2013.07.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Revised: 07/20/2013] [Accepted: 07/22/2013] [Indexed: 06/02/2023]
Abstract
Geranylgeranyl diphosphate synthase (GGPPS) catalyzes the condensation of the non-allylic diphosphate, isopentenyl diphosphate (IPP; C5), with allylic diphosphates to generate the C20 prenyl chain (GGPP) used for protein prenylation and diterpenoid biosynthesis. Here, we cloned the cDNA of a GGPPS from the spruce budworm, Choristoneura fumiferana, and characterized the corresponding recombinant protein (rCfGGPPS). As shown for other type-III GGPPSs, rCfGGPPS preferred farnesyl diphosphate (FPP; C15) over other allylic substrates for coupling with IPP. Unexpectedly, rCfGGPPS displayed inhibition by its FPP substrate at low IPP concentration, suggesting the existence of a mechanism that may regulate intracellular FPP pools. rCfGGPPS was also inhibited by its product, GGPP, in a competitive manner with respect to FPP, as reported for human and bovine brain GGPPSs. A homology model of CfGGPPS was prepared and compared to human and yeast GGPPSs. Consistent with its enzymological properties, CfGGPPS displayed a larger active site cavity that can accommodate the binding of FPP and GGPP in the region normally occupied by IPP and the allylic isoprenoid tail, and the binding of GGPP in an alternate orientation seen for GGPP binding to the human protein. To begin exploring the role of CfGGPPS in protein prenylation, its transcripts were quantified by qPCR in whole insects, along with those of other genes involved in this pathway. CfGGPPS was expressed throughout insect development and the abundance of its transcripts covaried with that of other prenylation-related genes. Our qPCR results suggest that geranylgeranylation is the predominant form of prenylation in whole C. fumiferana.
Collapse
Affiliation(s)
- Aline Barbar
- Département de biochimie, de microbiologie et de bio-informatique, Université Laval, Québec, QC G1V 0A6, Canada; Natural Resources Canada, Canadian Forest Service, Laurentian Forestry Centre, 1055 du P.E.P.S., C.P. 10380, Succ. Sainte-Foy, Québec, QC G1V 4C7, Canada
| | | | | | | | | | | | | |
Collapse
|
25
|
Srivastava DP, Woolfrey KM, Penzes P. Insights into rapid modulation of neuroplasticity by brain estrogens. Pharmacol Rev 2013; 65:1318-50. [PMID: 24076546 PMCID: PMC3799233 DOI: 10.1124/pr.111.005272] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Converging evidence from cellular, electrophysiological, anatomic, and behavioral studies suggests that the remodeling of synapse structure and function is a critical component of cognition. This modulation of neuroplasticity can be achieved through the actions of numerous extracellular signals. Moreover, it is thought that it is the integration of different extracellular signals regulation of neuroplasticity that greatly influences cognitive function. One group of signals that exerts powerful effects on multiple neurologic processes is estrogens. Classically, estrogens have been described to exert their effects over a period of hours to days. However, there is now increasing evidence that estrogens can rapidly influence multiple behaviors, including those that require forebrain neural circuitry. Moreover, these effects are found in both sexes. Critically, it is now emerging that the modulation of cognition by rapid estrogenic signaling is achieved by activation of specific signaling cascades and regulation of synapse structure and function, cumulating in the rewiring of neural circuits. The importance of understanding the rapid effects of estrogens on forebrain function and circuitry is further emphasized as investigations continue to consider the potential of estrogenic-based therapies for neuropathologies. This review focuses on how estrogens can rapidly influence cognition and the emerging mechanisms that underlie these effects. We discuss the potential sources and the biosynthesis of estrogens within the brain and the consequences of rapid estrogenic-signaling on the remodeling of neural circuits. Furthermore, we argue that estrogens act via distinct signaling pathways to modulate synapse structure and function in a manner that may vary with cell type, developmental stage, and sex. Finally, we present a model in which the coordination of rapid estrogenic-signaling and activity-dependent stimuli can result in long-lasting changes in neural circuits, contributing to cognition, with potential relevance for the development of novel estrogenic-based therapies for neurodevelopmental or neurodegenerative disorders.
Collapse
Affiliation(s)
- Deepak P Srivastava
- Department of Neuroscience & Centre for the Cellular Basis of Behaviour, 125 Coldharbour Lane, The James Black Centre, Institute of Psychiatry, King's College London, London, SE5 9NU, UK.
| | | | | |
Collapse
|
26
|
Shin YC, Jang TH, Yoon JH, Jeon JH, Park HH. Crystallization and preliminary X-ray crystallographic studies of Rab6A'(Q72L): a GTP-locked form. Acta Crystallogr Sect F Struct Biol Cryst Commun 2012; 68:1077-80. [PMID: 22949199 DOI: 10.1107/s1744309112030874] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2012] [Accepted: 07/06/2012] [Indexed: 12/27/2022]
Abstract
Rab6A, a member of the Ras superfamily of small G proteins, is involved in the regulation of vesicle trafficking, which is critical for endocytosis, cell differentiation and cell growth. Rab6A can exist in two isoforms termed Rab6A and Rab6A'. The substitution of Gln72 by Leu (Q72L) in the Rab6A family blocks GTP-hydrolysis activity, and this mutation usually causes the Rab6A protein to be in a constitutively active form. In this study, in order to understand the functional uniqueness of Rab6A' and the molecular mechanism of the control of activity by GTP and GDP from the crystal structure, a Rab6A'(Q72L) mutant form was overexpressed in Escherichia coli with an engineered N-terminal His tag. Rab6A'(Q72L) was then purified to homogeneity and crystallized at 293 K. X-ray diffraction data were collected to a resolution of 1.9 Å from a crystal belonging to space group P22(1)2(1) with unit-cell parameters a = 36.84, b = 96.78, c = 109.99 Å. The asymmetric unit was estimated to contain two molecules.
Collapse
Affiliation(s)
- Young-Cheul Shin
- Department of Physiology, Seoul National University College of Medicine, Seoul 110-799, Republic of Korea
| | | | | | | | | |
Collapse
|
27
|
Crystal structure of Rab6A'(Q72L) mutant reveals unexpected GDP/Mg²⁺ binding with opened GTP-binding domain. Biochem Biophys Res Commun 2012; 424:269-73. [PMID: 22750005 DOI: 10.1016/j.bbrc.2012.06.102] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2012] [Accepted: 06/20/2012] [Indexed: 01/14/2023]
Abstract
The Ras small G protein-superfamily is a family of GTP hydrolases whose activity is regulated by GTP/GDP binding states. Rab6A, a member of the Ras superfamily, is involved in the regulation of vesicle trafficking, which is critical for endocytosis, biosynthesis, secretion, cell differentiation and cell growth. Rab6A exists in two isoforms, termed RabA and Rab6A'. Substitution of Gln72 to Leu72 (Q72L) at Rab6 family blocks GTP hydrolysis activity and this mutation usually causes the Rab6 protein to be constitutively in an active form. Here, we report the crystal structure of the human Rab6A'(Q72L) mutant form at 1.9Å resolution. Unexpectedly, we found that Rab6A'(Q72L) possesses GDP/Mg(2+) in the GTP binding pockets, which is formed by a flexible switch I and switch II. Large conformational changes were also detected in the switch I and switch II regions. Our structure revealed that the non-hydrolysable, constitutively active form of Rab6A' can accommodate GDP/Mg(2+) in the open conformation.
Collapse
|
28
|
Gazzerro P, Proto MC, Gangemi G, Malfitano AM, Ciaglia E, Pisanti S, Santoro A, Laezza C, Bifulco M. Pharmacological actions of statins: a critical appraisal in the management of cancer. Pharmacol Rev 2011; 64:102-46. [PMID: 22106090 DOI: 10.1124/pr.111.004994] [Citation(s) in RCA: 321] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Statins, among the most commonly prescribed drugs worldwide, are cholesterol-lowering agents used to manage and prevent cardiovascular and coronary heart diseases. Recently, a multifaceted action in different physiological and pathological conditions has been also proposed for statins, beyond anti-inflammation and neuroprotection. Statins have been shown to act through cholesterol-dependent and -independent mechanisms and are able to affect several tissue functions and modulate specific signal transduction pathways that could account for statin pleiotropic effects. Typically, statins are prescribed in middle-aged or elderly patients in a therapeutic regimen covering a long life span during which metabolic processes, aging, and concomitant novel diseases, including cancer, could occur. In this context, safety, toxicity, interaction with other drugs, and the state of health have to be taken into account in subjects treated with statins. Some evidence has shown a dichotomous effect of statins with either cancer-inhibiting or -promoting effects. To date, clinical trials failed to demonstrate a reduced cancer occurrence in statin users and no sufficient data are available to define the long-term effects of statin use over a period of 10 years. Moreover, results from clinical trials performed to evaluate the therapeutic efficacy of statins in cancer did not suggest statin use as chemotherapeutic or adjuvant agents. Here, we reviewed the pharmacology of the statins, providing a comprehensive update of the current knowledge of their effects on tissues, biological processes, and pathological conditions, and we dissected the disappointing evidence on the possible future use of statin-based drugs in cancer therapy.
Collapse
Affiliation(s)
- Patrizia Gazzerro
- Department of Pharmaceutical and Biomedical Sciences, University of Salerno, Via Ponte Don Melillo, 84084 Fisciano (Salerno), Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
A rit GTPase-p38 mitogen-activated protein kinase survival pathway confers resistance to cellular stress. Mol Cell Biol 2011; 31:1938-48. [PMID: 21444726 DOI: 10.1128/mcb.01380-10] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Cells mobilize diverse signaling cascades to protect against stress-mediated injury. Ras family GTPases play a pivotal role in cell fate determination, serving as molecular switches to control the integration of multiple signaling pathways. p38 mitogen-activated protein kinase (MAPK) signaling serves as a critical fulcrum in this process, regulating networks that stimulate cellular apoptosis but also have the capacity to promote cell survival. However, relatively little is known concerning this functional dichotomy, particularly the regulation of p38-dependent survival pathways. Here, we demonstrate that the Rit GTPase promotes cell survival by directing an unexpected p38 MAPK-dependent AKT survival pathway. Following stress exposure, Rit small hairpin RNA interference (shRNAi)-treated cells display increased apoptosis and selective disruption of p38 MAPK signaling, while expression of constitutively activated Rit promotes p38-AKT-dependent cell survival. Rit, but not Ras or Rap GTPases, can associate with, and is critical for, stress-mediated activation of the scaffolded p38-MK2-HSP27-AKT prosurvival signaling complex. Together, our studies establish Rit as a central regulator of a p38 MAPK-dependent signaling cascade that functions as a critical cellular survival mechanism in response to stress.
Collapse
|
30
|
Chen W, Wang X, Deng C, Lv X, Fan Y, Men J, Liang C, Yu X. Molecular cloning and characterization of a novel ras-related protein (rap2) from Clonorchis sinensis. Parasitol Res 2010; 108:1021-6. [DOI: 10.1007/s00436-010-2147-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2010] [Accepted: 10/28/2010] [Indexed: 11/28/2022]
|
31
|
Jackson AJ, Coats P, Orr DJ, Teenan RP, Wadsworth RM. Pharmacotherapy to Improve Outcomes in Infrainguinal Bypass Graft Surgery: A Review of Current Treatment Strategies. Ann Vasc Surg 2010; 24:562-72. [DOI: 10.1016/j.avsg.2010.02.028] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2009] [Revised: 10/14/2009] [Accepted: 02/11/2010] [Indexed: 11/15/2022]
|
32
|
He YD, Liu DD, Xi DM, Yang LY, Tan YW, Liu Q, Mao HM, Deng WD. Isolation, sequence identification and expression profile of three novel genes Rab2A, Rab3A and Rab7A from black-boned sheep (Ovis aries). Mol Biol 2010. [DOI: 10.1134/s0026893310010036] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
33
|
Nagata K, Okano Y, Suzuki T, Nozawa Y. Evidence for the Presence of a LowMrGTP-binding Protein,ramp25, in Human Platelet Membranes. Platelets 2009; 4:268-74. [DOI: 10.3109/09537109309013227] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
34
|
Bhuin T, Roy JK. Rab11 is required for myoblast fusion in Drosophila. Cell Tissue Res 2009; 336:489-99. [PMID: 19370361 DOI: 10.1007/s00441-009-0782-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2008] [Accepted: 02/11/2009] [Indexed: 01/06/2023]
Abstract
Rab11, an evolutionarily conserved, ubiquitously expressed subfamily of small monomeric Rab GTPases, has been implicated in regulating vesicular trafficking through the recycling of endosomal compartment. In order to gain an insight into the role of this gene in myogenesis during embryonic development, we have studied the expression pattern of Rab11 in mesoderm during muscle differentiation in Drosophila embryo. When dominant-negative or constitutively active Drosophila Rab11 proteins are expressed or Rab11 is reduced via double-stranded RNA in muscle precursors, they cause partial failure of myoblast fusion and show anomalies in the shape of the muscle fibres. Our results suggest that Rab11 plays no role in cell fate specification in muscle precursors but is required late in the process of myoblast fusion.
Collapse
Affiliation(s)
- Tanmay Bhuin
- Cytogenetics Laboratory, Department of Zoology, Banaras Hindu University, Varanasi, 221 005, India
| | | |
Collapse
|
35
|
Mruk DD, Silvestrini B, Cheng CY. Anchoring junctions as drug targets: role in contraceptive development. Pharmacol Rev 2008; 60:146-80. [PMID: 18483144 DOI: 10.1124/pr.107.07105] [Citation(s) in RCA: 117] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
In multicellular organisms, cell-cell interactions are mediated in part by cell junctions, which underlie tissue architecture. Throughout spermatogenesis, for instance, preleptotene leptotene spermatocytes residing in the basal compartment of the seminiferous epithelium must traverse the blood-testis barrier to enter the adluminal compartment for continued development. At the same time, germ cells must also remain attached to Sertoli cells, and numerous studies have reported extensive restructuring at the Sertoli-Sertoli and Sertoli-germ cell interface during germ cell movement across the seminiferous epithelium. Furthermore, the proteins and signaling cascades that regulate adhesion between testicular cells have been largely delineated. These findings have unveiled a number of potential "druggable" targets that can be used to induce premature release of germ cells from the seminiferous epithelium, resulting in transient infertility. Herein, we discuss a novel approach with the aim of developing a nonhormonal male contraceptive for future human use, one that involves perturbing adhesion between Sertoli and germ cells in the testis.
Collapse
Affiliation(s)
- Dolores D Mruk
- Population Council, Center for Biomedical Research, The Mary M Wohlford Laboratory for Male Contraceptive Research, 1230 York Avenue, New York, NY 10065, USA.
| | | | | |
Collapse
|
36
|
Monteiro HP, Arai RJ, Travassos LR. Protein tyrosine phosphorylation and protein tyrosine nitration in redox signaling. Antioxid Redox Signal 2008; 10:843-89. [PMID: 18220476 DOI: 10.1089/ars.2007.1853] [Citation(s) in RCA: 124] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Reversible phosphorylation of protein tyrosine residues by polypeptide growth factor-receptor protein tyrosine kinases is implicated in the control of fundamental cellular processes including the cell cycle, cell adhesion, and cell survival, as well as cell proliferation and differentiation. During the last decade, it has become apparent that receptor protein tyrosine kinases and the signaling pathways they activate belong to a large signaling network. Such a network can be regulated by various extracellular cues, which include cell adhesion, agonists of G protein-coupled receptors, and oxidants. It is well documented that signaling initiated by receptor protein tyrosine kinases is directly dependent on the intracellular production of oxidants, including reactive oxygen and nitrogen species. Accumulated evidence indicates that the intracellular redox environment plays a major role in the mechanisms underlying the actions of growth factors. Oxidation of cysteine thiols and nitration of tyrosine residues on signaling proteins are described as posttranslational modifications that regulate, positively or negatively, protein tyrosine phosphorylation (PTP). Early observations described the inhibition of PTP activities by oxidants, resulting in increased levels of proteins phosphorylated on tyrosine. Therefore, a redox circuitry involving the increasing production of intracellular oxidants associated with growth-factor stimulation/cell adhesion, oxidative reversible inhibition of protein tyrosine phosphatases, and the activation of protein tyrosine kinases can be delineated.
Collapse
Affiliation(s)
- Hugo P Monteiro
- Department of Biochemistry/Molecular Biology and CINTERGEN, Universidade Federal de São Paulo, São Paulo, Brazil.
| | | | | |
Collapse
|
37
|
Tao Y, Chen YC, Li YY, Yang SQ, Xu WR. Localization and translocation of RhoA protein in the human gastric cancer cell line SGC-7901. World J Gastroenterol 2008; 14:1175-81. [PMID: 18300342 PMCID: PMC2690664 DOI: 10.3748/wjg.14.1175] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To elucidate the localization of RhoA in gastric SGC-7901 cancer cells and its translocation by lysophosphatidic acid (LPA) and/or 8-chlorophenylthio-cAMP (CPT-cAMP).
METHODS: Immunofluorescence microscopy was used to determine the localization of RhoA. Western blotting was used to detect both endogenous and exogenous RhoA in different cellular compartments (membrane, cytosol, nucleus) and the translocation of RhoA following treatment with LPA, CPT-cAMP, or CPT-cAMP + LPA.
RESULTS: Immunofluorescence staining revealed endogenous RhoA to be localized in the membrane, the cytosol, and the nucleus, and its precise localization within the nucleus to be the nucleolus. Western blotting identified both endogenous and exogenous RhoA within different cellular compartments (membrane, cytosol, nucleus, nucleolus). After stimulation with LPA, the amount of RhoA within membrane and nuclear extracts increased, while it decreased in the cytosol fractions. After treatment with CPT-cAMP the amount of RhoA within the membrane and the nuclear extracts decreased, while it increased within the cytosol fraction. Treatment with a combination of both substances led to a decrease in RhoA in the membrane and the nucleus but to an increase in the cytosol.
CONCLUSION: In SGC-7901 cells RhoA was found to be localized within the membrane, the cytosol, and the nucleus. Within the nucleus its precise localization could be demonstrated to be the nucleolus. Stimulation with LPA caused a translocation of RhoA from the cytosol towards the membrane and the nucleus; treatment with CPT-cAMP caused the opposite effect. Furthermore, pre-treatment with CPT-cAMP was found to block the effect of LPA.
Collapse
|
38
|
Dudakovic A, Wiemer AJ, Lamb KM, Vonnahme LA, Dietz SE, Hohl RJ. Inhibition of geranylgeranyl diphosphate synthase induces apoptosis through multiple mechanisms and displays synergy with inhibition of other isoprenoid biosynthetic enzymes. J Pharmacol Exp Ther 2007; 324:1028-36. [PMID: 18083912 DOI: 10.1124/jpet.107.132217] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Inhibitors of isoprenoid synthesis are widely used for treatment of human diseases, including hypercholesterolemia and osteoporosis, and they have the potential to be useful for treatment of cancer. Statin drugs inhibit the enzyme HMG-CoA reductase, whereas nitrogenous bisphosphonates have more recently been shown to inhibit farnesyl disphosphate synthase. In addition, our laboratory has recently developed several potent and specific bisphosphonate inhibitors of geranylgeranyl diphosphate synthase, including digeranyl bisphosphonate. Because all three enzymes fall in the same biosynthetic pathway and many of the biological effects are due to depletion of downstream products, we hypothesized that simultaneous inhibition of these enzymes would result in synergistic growth inhibition. In this study, we show that inhibition of geranylgeranyl diphosphate synthase induces apoptosis in K562 leukemia cells. This induction of apoptosis is in part dependent upon both geranylgeranyl diphosphate depletion and accumulation of farnesyl diphosphate. Combinations of either lovastatin or zoledronate with digeranyl bisphosphonate synergistically inhibited growth and induced apoptosis. These combinations also potently inhibited cellular geranylgeranylation. These results support the potential for combinations of multiple inhibitors of isoprene biosynthesis to inhibit cancer cell growth or metastasis at clinically achievable concentrations.
Collapse
|
39
|
Takai Y, Kaibuchi K, Kikuchi A, Sasaki T, Shirataki H. Regulators of small GTPases. CIBA FOUNDATION SYMPOSIUM 2007; 176:128-38; discussion 138-46. [PMID: 8299416 DOI: 10.1002/9780470514450.ch9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Small GTPases are converted from the GDP-bound inactive form to the GTP-bound active form by a GDP/GTP exchange reaction which is regulated by GDP/GTP exchange proteins (GEPs). We have found both stimulatory and inhibitory GEPs, which we have named GDP dissociation stimulators (GDSs) and GDP dissociation inhibitors (GDIs) respectively. We have isolated Smg GDS, Rho GDI and Rab GDI, cloned them, and determined their primary structures. These GEPs are active on a group of small GTPases: Smg GDS on at least K-Ras, Rap1/Smg21, Rho and Rac; Rho GDI on at least Rho, Rac and Cdc42; Rab GDI on most of the Rab family members. These GEPs have an additional function, regulating the translocation of their substrate small GTPases between the membrane and the cytosol. The GEPs interact only with the post-translationally modified form of their substrate small GTPases.
Collapse
Affiliation(s)
- Y Takai
- Department of Biochemistry, Kobe University School of Medicine, Japan
| | | | | | | | | |
Collapse
|
40
|
Lemonnier M, Landraud L, Lemichez E. Rho GTPase-activating bacterial toxins: from bacterial virulence regulation to eukaryotic cell biology. FEMS Microbiol Rev 2007; 31:515-34. [PMID: 17680807 DOI: 10.1111/j.1574-6976.2007.00078.x] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Studies on the interactions of bacterial pathogens with their host have provided an invaluable source of information on the major functions of eukaryotic and prokaryotic cell biology. In addition, this expanding field of research, known as cellular microbiology, has revealed fascinating examples of trans-kingdom functional interplay. Bacterial factors actually exploit eukaryotic cell machineries using refined molecular strategies to promote invasion and proliferation within their host. Here, we review a family of bacterial toxins that modulate their activity in eukaryotic cells by activating Rho GTPases and exploiting the ubiquitin/proteasome machineries. This family, found in human and animal pathogenic Gram-negative bacteria, encompasses the cytotoxic necrotizing factors (CNFs) from Escherichia coli and Yersinia species as well as dermonecrotic toxins from Bordetella species. We survey the genetics, biochemistry, molecular and cellular biology of these bacterial factors from the standpoint of the CNF1 toxin, the paradigm of Rho GTPase-activating toxins produced by urinary tract infections causing pathogenic Escherichia coli. Because it reveals important connections between bacterial invasion and the host inflammatory response, the mode of action of CNF1 and its related Rho GTPase-targetting toxins addresses major issues of basic and medical research and constitutes a privileged experimental model for host-pathogen interaction.
Collapse
Affiliation(s)
- Marc Lemonnier
- INSERM U627, UNSA, Faculté de Médecine, 28 Avenue de Valombrose, 06107 Nice cedex 2, France.
| | | | | |
Collapse
|
41
|
Filipeanu CM, Zhou F, Fugetta EK, Wu G. Differential regulation of the cell-surface targeting and function of beta- and alpha1-adrenergic receptors by Rab1 GTPase in cardiac myocytes. Mol Pharmacol 2006; 69:1571-8. [PMID: 16461589 DOI: 10.1124/mol.105.019984] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The molecular mechanism underlying the export from the endoplasmic reticulum (ER) to the cell surface and its role in the regulation of signaling of adrenergic receptors (ARs) remain largely unknown. In this report, we determined the role of Rab1, a Ras-like GTPase that coordinates protein transport specifically from the ER to the Golgi, in the cell surface targeting and function of endogenous beta- and alpha1-ARs in neonatal rat ventricular myocytes. Adenovirus-driven expression of Rab1 into myocytes selectively increased the cell-surface number of alpha1-AR, but not beta-AR, whereas the dominant-negative mutant Rab1N124I significantly reduced the cell-surface expression of beta-AR and alpha1-AR. Brefeldin A inhibited beta-AR and alpha1-AR export and antagonized the Rab1 effect on alpha1-AR expression. Manipulation of Rab1 function similarly influenced the transport of alpha1A- and alpha1B-ARs as well as beta1- and beta2-ARs. Fluorescent microscopy analysis demonstrated that expression of Rab1N124I and Rab1 small interfering RNA induced a marked accumulation of GFP-tagged beta2-AR and alpha1B-AR in the ER. Consistent with the effects on receptor cell-surface targeting, Rab1 selectively enhanced ERK1/2 activation and hypertrophic growth in response to the alpha1-AR agonist phenylephrine but not to the beta-AR agonist isoproterenol. Rab1N124I inhibited both agonist-mediated ERK1/2 activation and hypertrophic growth in neonatal myocytes. These results demonstrate that the cell-surface targeting and signaling of beta- and alpha1-ARs require Rab1 and are differentially modulated by augmentation of Rab1 function. Our data provide strong evidence implicating the ER-to-Golgi traffic as a site for selective manipulation of distinct AR function in cardiac myocytes.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Cell Membrane/physiology
- Heart/physiology
- Humans
- MAP Kinase Signaling System/physiology
- Microscopy, Fluorescence
- Muscle Cells/drug effects
- Muscle Cells/enzymology
- Muscle Cells/physiology
- Myocardium/metabolism
- RNA, Small Interfering
- Rats
- Receptors, Adrenergic, alpha-1/physiology
- Receptors, Adrenergic, beta/drug effects
- Receptors, Adrenergic, beta/physiology
- Receptors, Cell Surface/physiology
- rab1 GTP-Binding Proteins/metabolism
Collapse
Affiliation(s)
- Catalin M Filipeanu
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, 1901 Perdido St., New Orleans, LA 70112, USA
| | | | | | | |
Collapse
|
42
|
Makioka A, Kumagai M, Takeuchi T, Nozaki T. Characterization of protein geranylgeranyltransferase I from the enteric protist Entamoeba histolytica. Mol Biochem Parasitol 2006; 145:216-25. [PMID: 16300841 DOI: 10.1016/j.molbiopara.2005.10.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2005] [Revised: 09/28/2005] [Accepted: 10/04/2005] [Indexed: 11/27/2022]
Abstract
Entamoeba histolytica is a unique protozoan parasite possessing both protein farnesyltransferase and geranylgeranyltrasferase I (GGT-I) for isoprenylation of small GTPases. In this study, we demonstrated unique enzymological properties of the amebic GGT-I (EhGGT-I), including substrate specificity and insensitivity to known mammalian inhibitors. Some of important residues of the catalytic beta subunit implicated in the specificity for GTPase acceptors and prenyl donors are substituted in EhGGT-I. Recombinant alpha and beta subunits of EhGGT-I, co-expressed in Escherichia coli, showed activity to transfer geranylgeranyl to both human wild-type (CVLS) and mutant (CVLL) H-Ras, while the mammalian GGT-I geranylgeranylated, but not farnesylated, only mutant H-Ras. All the representative amebic Ras and Rho/Rac small GTPases with phenylalanine, leucine, methionine, or alanine terminus were preferentially geranylgeranylated by EhGGT-I. This indicates that the acceptor specificity of the amebic GGT-I is remarkably broader than that of its mammalian counterpart. In contrast to EhFT, which farnesylates but not geranylgeranlylates solely EhRas4-CVVA, EhGGT-I also showed significant farnesyltransferase activity against Ras GTPase acceptors. EhGGT-I showed remarkable resistance to peptidomimetics known to inhibit mammalian GGT-I. Together with our previous observation that this parasite does not appear to depend on farnesylation for a majority of Ras and Rho/Rac, these data indicate that biological and biochemical advantages leading to the evolutional selection of this isoprenyl modification must exist uniquely in this parasitic protist. Finally, remarkable biochemical differences in binding to substrates and inhibitors between amebic and mammalian GGT-I highlight this enzyme as an attractive target for the development of new chemotherapeutics against amebiasis.
Collapse
Affiliation(s)
- Asao Makioka
- Department of Tropical Medicine, Jikei University School of Medicine, 3-25-8 Nishi-shinbashi, Minato-ku, Tokyo 105-8461, Japan
| | | | | | | |
Collapse
|
43
|
Glenn MP, Chang SY, Hucke O, Verlinde CLMJ, Rivas K, Hornéy C, Yokoyama K, Buckner FS, Pendyala PR, Chakrabarti D, Gelb M, Van Voorhis WC, Sebti SM, Hamilton AD. Structurally Simple Farnesyltransferase Inhibitors Arrest the Growth of Malaria Parasites. Angew Chem Int Ed Engl 2005. [DOI: 10.1002/ange.200500674] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
44
|
Glenn MP, Chang SY, Hucke O, Verlinde CLMJ, Rivas K, Hornéy C, Yokoyama K, Buckner FS, Pendyala PR, Chakrabarti D, Gelb M, Van Voorhis WC, Sebti SM, Hamilton AD. Structurally Simple Farnesyltransferase Inhibitors Arrest the Growth of Malaria Parasites. Angew Chem Int Ed Engl 2005; 44:4903-6. [PMID: 16007716 DOI: 10.1002/anie.200500674] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Matthew P Glenn
- Department of Chemistry, Yale University, 225 Prospect St., New Haven, CT 06511, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Cytotoxic Necrotizing Factors: Rho-Activating Toxins from Escherichia coli. EcoSal Plus 2004; 1. [PMID: 26443355 DOI: 10.1128/ecosalplus.8.7.4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
This article reviews the Escherichia coli toxins called cytotoxic necrotizing factors (CNFs), which cause activation of Rho GTPases. It describes their modes of action, structure-function relationships, and roles in disease. Rho GTPases, the targets of CNFs, belong to the Ras superfamily of low molecular mass GTPases and act as molecular switches in various signaling pathways. Low molecular mass GTPases of the Rho family are known as master regulators of the actin cytoskeleton. Moreover, they are involved in various signal transduction processes, from transcriptional activation, cell cycle progression, and cell transformation to apoptosis. CNFs are cytotoxic for a wide variety of cells, including 3T3 fibroblasts, Chinese hamster ovary cells, Vero cells, HeLa cells, and cell lines of neuronal origin. This implies that a commonly expressed receptor is responsible for the uptake of CNF1. Cultured mammalian cells treated with CNFs are characterized by dramatic changes in actin-containing structures, including stress fibers, lamellipodia, and filopodia. Most striking is the formation of multinucleation in these cells. Rho GTPases are increasingly recognized as essential factors in the development of cancer and metastasis. This fact has initiated a discussion as to whether activation of Rho proteins by CNFs might be involved in tumorigenesis. Moreover, CNF1 increases the expression of the cyclooxygenase 2 (Cox2) gene in fibroblasts. Increased expression of Cox2 is observed in some types of tumors, e.g., colon carcinoma. Lipid-mediators produced by the enzyme are suggested to be responsible for tumor progression.
Collapse
|
46
|
Nijhara R, van Hennik PB, Gignac ML, Kruhlak MJ, Hordijk PL, Delon J, Shaw S. Rac1 mediates collapse of microvilli on chemokine-activated T lymphocytes. THE JOURNAL OF IMMUNOLOGY 2004; 173:4985-93. [PMID: 15470041 DOI: 10.4049/jimmunol.173.8.4985] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Lymphocytes circulate in the blood and upon chemokine activation rapidly bind, where needed, to microvasculature to mediate immune surveillance. Resorption of microvilli is an early morphological alteration induced by chemokines that facilitates lymphocyte emigration. However, the antecedent molecular mechanisms remain largely undefined. We demonstrate that Rac1 plays a fundamental role in chemokine-induced microvillar breakdown in human T lymphocytes. The supporting evidence includes: first, chemokine induces Rac1 activation within 5 s via a signaling pathway that involves Galphai. Second, constitutively active Rac1 mediates microvilli disintegration. Third, blocking Rac1 function by cell permeant C-terminal "Trojan" peptides corresponding to Rac1 (but not Rac2, Rho, or Cdc42) blocks microvillar loss induced by the chemokine stromal cell-derived factor 1alpha (SDF-1alpha). Furthermore, we demonstrate that the molecular mechanism of Rac1 action involves dephosphorylation-induced inactivation of the ezrin/radixin/moesin (ERM) family of actin regulators; such inactivation is known to detach the membrane from the underlying actin cytoskeleton, thereby facilitating disassembly of actin-based peripheral processes. Specifically, ERM dephosphorylation is induced by constitutively active Rac1 and stromal cell-derived factor 1alpha-induced ERM dephosphorylation is blocked by either the dominant negative Rac1 construct or by Rac1 C-terminal peptides. Importantly, the basic residues at the C terminus of Rac1 are critical to Rac1's participation in ERM dephosphorylation and in microvillar retraction. Together, these data elucidate new roles for Rac1 in early signal transduction and cytoskeletal rearrangement of T lymphocytes responding to chemokine.
Collapse
Affiliation(s)
- Ruchika Nijhara
- National Cancer Institute, Experimental Immunology Branch, Bethesda, MD 20892, USA
| | | | | | | | | | | | | |
Collapse
|
47
|
Nishimura Y, Mabuchi I. An IQGAP-like protein is involved in actin assembly together with Cdc42 in the sea urchin egg. ACTA ACUST UNITED AC 2004; 56:207-18. [PMID: 14584023 DOI: 10.1002/cm.10146] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
We isolated a gene homologous to human cdc42 (ucdc42) from a sea urchin cDNA library. The GTPgammaS-bound UCdc42 induced actin assembly in sea urchin egg extract. Proteins that are involved in this actin assembly system were searched using UCdc42-bound agarose beads. A 180-kDa protein (p180), which showed a homology to human IQGAPs, bound to the GTPgammaS-UCdc42 beads. Immunodepletion of p180 from the sea urchin egg extract abolished this actin assembly on the UCdc42 beads. Immunofluorescent localization of p180 was similar to that of the actin cytoskeleton in the egg cortex and it was concentrated in the cleavage furrow during cytokinesis. A possible role of p180 in actin assembly is discussed.
Collapse
Affiliation(s)
- Yukako Nishimura
- Division of Biology, Department of Life Sciences, Graduate School of Arts and Sciences, University of Tokyo, Komaba, Meguro-ku, Tokyo
| | | |
Collapse
|
48
|
Molon A, Di Giovanni S, Chen YW, Clarkson PM, Angelini C, Pegoraro E, Hoffman EP. Large-scale disruption of microtubule pathways in morphologically normal human spastin muscle. Neurology 2004; 62:1097-104. [PMID: 15079007 DOI: 10.1212/01.wnl.0000118204.90814.5a] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To investigate the molecular pathways disrupted by dominant spastin mutations in apparently unaffected skeletal muscle from patients with motor neuron disease (SPG4). METHODS The authors studied muscle of three individuals from two unrelated families affected by spastic paraplegia caused by spastin mutations. The authors compared RNA expression profiles to 7 normal and 13 pathologic muscle U95A profiles (Duchenne dystrophy, acute quadriplegic myopathy, and spinal muscular atrophy). Data were validated with U133A arrays with seven different control specimens. mRNA and protein confirmations were done for a subset of genes. RESULTS Both nonsense and missense mutations in the spastin gene disrupted microtubule pathways in nonpathologic tissue, including microtubule dynamics, stability, exocytosis, and endocytosis. CONCLUSIONS Normal muscle can be used to uncover biochemical perturbation in motor neuron disease. Altered microtubule metabolism in SPG4-linked hereditary spastic paraplegia patients leads to pathology of the long descending tracks of motor neurons that likely have a stringent need for efficient microtubular transport. As many inherited neurologic conditions show a systemic biochemical defect with disease limited to neurons, our data have broader implications for biochemical pathway studies of many neurologic disorders.
Collapse
Affiliation(s)
- A Molon
- Center for Genetic Medicine, Children's National Medical Center, Washington, DC 20010, USA
| | | | | | | | | | | | | |
Collapse
|
49
|
Kumagai M, Makioka A, Takeuchi T, Nozaki T. Molecular Cloning and Characterization of a Protein Farnesyltransferase from the Enteric Protozoan Parasite Entamoeba histolytica. J Biol Chem 2004; 279:2316-23. [PMID: 14583615 DOI: 10.1074/jbc.m311478200] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Genes encoding alpha- and beta-subunits of a putative protein farnesyltransferase (FT) from the enteric protozoan parasite Entamoeba histolytica were obtained and their biochemical properties were characterized. Deduced amino acid sequences of the alpha- and beta-subunit of E. histolytica FT (EhFT) were 298- and 375-residues long with a molecular mass of 35.6 and 42.6 kDa, and a pI of 5.43 and 5.65, respectively. They showed 24% to 36% identity to and shared common signature domains and repeats with those from other organisms. Recombinant alpha- and beta-subunits, co-expressed in Escherichia coli, formed a heterodimer and showed activity to transfer farnesyl using farnesylpyrophosphate as a donor to human H-Ras possessing a C-terminal CVLS, but not a mutant H-Ras possessing CVLL. Among a number of small GTPases that belong to the Ras superfamily from this parasite, we identified EhRas4, which possesses CVVA at the C terminus, as a sole farnesyl acceptor for EhFT. This is in contrast to mammalian FT, which utilizes a variety of small GTPases that possess a C-terminal CaaX motif, where X is serine, methionine, glutamine, cysteine, or alanine. EhFT also showed remarkable resistance against a variety of known inhibitors of mammalian FT. These results suggest that remarkable biochemical differences in binding to substrates and inhibitors exist between amebic and mammalian FTs, which highlights this enzyme as a novel target for the development of new chemotherapeutics against amebiasis.
Collapse
Affiliation(s)
- Masahiro Kumagai
- Department of Tropical Medicine, Jikei University School of Medicine, 3-25-8 Nishi-shinbashi, Minato-ku, Tokyo 105-8461, Japan
| | | | | | | |
Collapse
|
50
|
Betancor-Hernández E, Pérez-Machín R, Henríquez-Hernández L, Mateos-Díaz C, Novoa-Mogollón J, Fernández-Pérez L. Photoaffinity labeling identification of thyroid hormone-regulated glucocorticoid-binding peptides in rat liver endoplasmic reticulum: an oligomeric protein with high affinity for 16beta-hydroxylated stanozolol. J Steroid Biochem Mol Biol 2003; 87:253-64. [PMID: 14698206 DOI: 10.1016/j.jsbmb.2003.09.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Steroid-binding proteins unrelated to the classical nuclear receptors have been proposed to play a role in non-genomic actions of the17alpha-alkylated testosterone derivative (17alpha-AA) stanozolol (ST). We have previously reported that male rat liver endoplasmic reticulum contains two steroid-binding sites associated with high molecular mass oligomeric proteins: (1) the ST-binding protein (STBP); and (2) the low-affinity glucocorticoid-binding protein (LAGS). To further explore the role of LAGS on the mechanism of action of ST, we have now studied: (1) the interaction of ST and its hydroxylated metabolites with solubilized LAGS and the cytosolic glucocorticoid receptor (GR); and (2) the effects of hormones on the capability of STBP to bind ST. We found that, unlike 17alpha-methyltestosterone, neither ST nor its hydroxylated metabolites bind to GR. However, the 16beta-hydroxylation of ST significantly increases the capability of LAGS to bind ST. Interestingly, 3'-hydroxylation of ST abrogates the capability of LAGS to bind ST. ST (k(i)=30 nM) and 16beta-hydroxystanozolol (k(i)=13 nM) bind with high affinity to LAGS, and are capable of accelerating the rate of dissociation of previously bound dexamethasone from the LAGS. STBP and LAGS are strongly induced by ethinylestradiol. However, unlike STBP, LAGS is regulated by thyroid hormones and growth hormone, which proves that these steroid-binding activities are associated with different binding sites. These findings seem to suggest a novel mechanism for ST whereby membrane-associated glucocorticoid-binding activity is targeted by the 16beta-hydroxylated metabolite of ST. ST and its 16beta-hydroxylated metabolite modulate glucocorticoid activity in the liver through negative allosteric modulation of LAGS, with the result of this interaction an effective increase in classical GR-signaling by increasing glucocorticoid availability to the cytosolic GR.
Collapse
Affiliation(s)
- Eva Betancor-Hernández
- Department of Clinical Sciences, Health Sciences Center, University of Las Palmas de Gran Canaria, and Instituto Canario de Investigación del Cáncer, P.O. Box 550, 35080 Canary Islands, Spain
| | | | | | | | | | | |
Collapse
|