1
|
Seshire A, Duan Y, Lang K. Reaching Outer Space and Enabling the mRNA Revolution: A Critical Role of Partnerships for Successful Drug and Vaccine Development. Handb Exp Pharmacol 2024; 286:51-81. [PMID: 39254747 DOI: 10.1007/164_2024_723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
mRNA and targeted delivery of mRNA carry the promise to enable targeted treatment of undruggable diseases with high unmet medical needs. The transient nature of mRNA opens options for safe influencing of protein biology, immune responses, and complex ailments without impacting DNA heritage. Technical challenges such as mRNA stability and targeted delivery require next generation solutions, which attracted substantial funding and research interests. To build an integrated mRNA value chain and enable the development of novel therapeutics, Merck KGaA Darmstadt, Germany has initiated an internally incubated program, "Targeted mRNA Delivery" (TMD). This collaborative approach brings together scientists, researchers, engineers, and commercial experts from diverse backgrounds to overcome the multidimensional challenges associated with mRNA technology. In this chapter, the multiple opportunities and challenges for the development of mRNA formulations and therapeutics are described comprehensively. Specifically, the TMD program is presented as a use case to show how intrapreneurs were gathered to establish internal mRNA capabilities and foster collaborations for technology development. In the realm of targeted mRNA delivery, partnerships, encompassing internal partnership and external private, public, and hybrid collaborations, play a crucial role in driving innovation and addressing these hurdles. Within multinational pharmaceutical companies, the establishment of "internal startups" is an effective solution to drive innovation to the next level with support from different business sectors, where existing capabilities and positioning are seamlessly blended with the agility and speed of a startup.
Collapse
Affiliation(s)
| | - Yukun Duan
- Merck Pte. Ltd., an affiliate of Merck KGaA, Darmstadt, Germany, Singapore, Singapore
| | | |
Collapse
|
2
|
Goswami B, Nag S, Ray PS. Fates and functions of RNA-binding proteins under stress. WILEY INTERDISCIPLINARY REVIEWS. RNA 2023:e1825. [PMID: 38014833 DOI: 10.1002/wrna.1825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 10/03/2023] [Accepted: 10/30/2023] [Indexed: 11/29/2023]
Abstract
Exposure to stress activates a well-orchestrated set of changes in gene expression programs that allow the cell to cope with and adapt to the stress, or undergo programmed cell death. RNA-protein interactions, mediating all aspects of post-transcriptional regulation of gene expression, play crucial roles in cellular stress responses. RNA-binding proteins (RBPs), which interact with sequence/structural elements in RNAs to control the steps of RNA metabolism, have therefore emerged as central regulators of post-transcriptional responses to stress. Following exposure to a variety of stresses, the dynamic alterations in the RNA-protein interactome enable cells to respond to intracellular or extracellular perturbations by causing changes in mRNA splicing, polyadenylation, stability, translation, and localization. As RBPs play a central role in determining the cellular proteome both qualitatively and quantitatively, it has become increasingly evident that their abundance, availability, and functions are also highly regulated in response to stress. Exposure to stress initiates a series of signaling cascades that converge on post-translational modifications (PTMs) of RBPs, resulting in changes in their subcellular localization, association with stress granules, extracellular export, proteasomal degradation, and RNA-binding activities. These alterations in the fate and function of RBPs directly impact their post-transcriptional regulatory roles in cells under stress. Adopting the ubiquitous RBP HuR as a prototype, three scenarios illustrating the changes in nuclear-cytoplasmic localization, RNA-binding activity, export and degradation of HuR in response to inflammation, genotoxic stress, and heat shock depict the complex and interlinked regulatory mechanisms that control the fate and functions of RBPs under stress. This article is categorized under: RNA Interactions with Proteins and Other Molecules > Protein-RNA Interactions: Functional Implications.
Collapse
Affiliation(s)
- Binita Goswami
- Department of Biological Sciences, Indian Institute of Science Education and Research, Mohanpur, West Bengal, India
| | - Sharanya Nag
- Department of Biological Sciences, Indian Institute of Science Education and Research, Mohanpur, West Bengal, India
| | - Partho Sarothi Ray
- Department of Biological Sciences, Indian Institute of Science Education and Research, Mohanpur, West Bengal, India
| |
Collapse
|
3
|
Palacios-Luna JE, López-Marrufo MV, Bautista-Bautista G, Velarde-Guerra CS, Villeda-Gabriel G, Flores-Herrera O, Osorio-Caballero M, Aguilar-Carrasco JC, Palafox-Vargas ML, García-López G, Díaz-Ruíz O, Arechavaleta-Velasco F, Flores-Herrera H. Progesterone modulates extracellular heat-shock proteins and interlukin-1β in human choriodecidual after Escherichia coli infection. Placenta 2023; 142:85-94. [PMID: 37659254 DOI: 10.1016/j.placenta.2023.08.074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 08/25/2023] [Accepted: 08/27/2023] [Indexed: 09/04/2023]
Abstract
INTRODUCTION Chorioamnionitis is an adverse condition in human pregnancy caused by many bacterial pathogens including Escherichia coli (E. coli); which has been associated with higher risk of preterm birth. We recently reported that human maternal decidua (MDec) tissue responds to E. coli infection by secreting extracellular heat-shock proteins (eHsp)-60, -70 and interlukin-1β (IL-1β). Previous studies have shown that progesterone (P4) regulates the immune response, but it is unknown whether P4 inhibits the secretion of eHsp. The aim of this investigation was to determine the role of P4 on the secretion of eHsp-27, -60, -70 and IL-1β in MDec after 3, 6, and 24 h of E. coli infection. METHODS Nine human feto-maternal interface (HFMi) tissues were included and mounted in the Transwell culture system. Only the maternal decidua (MDec) was stimulated for 3, 6 and 24 h with E. coli alone or in combination with progesterone and RU486. After each treatment, the HFMi tissue was recovered to determine histological changes and the culture medium recovered to evaluate the levels of eHsp-27, -60, -70 and IL-1β by ELISA and mRNA expression by RT-PCR. RESULTS No structural changes were observed in the HFMi tissue treated with P4 and RU486. However, stimulation with E. coli produces diffuse inflammation and ischemic necrosis. E. coli induced infection decreases, in time- and dose-dependent manner, eHsp-27 and increases eHsp-60, eHsp-70 and IL-1β levels. In contrast, incubation of HFMi tissue with E. coli + P4 reversed eHsp and IL-1β secretion levels relative to E. coli stimulation group but not relative to the control group. The same profile was observed on the expression of eHsp-27 and eHsp-60. DISCUSSION we found that progesterone modulates the anti-inflammatory (eHsp-27) and pro-inflammatory (eHsp-60 and eHsp-70) levels of eHsp induced by E. coli infection in human choriodecidual tissue. eHsp-60 and eHsp-70 levels were not completely reversed; maintaining the secretion of IL-1β, which has been associated with adverse events during pregnancy.
Collapse
Affiliation(s)
- Janelly Estefania Palacios-Luna
- Departamento de Inmunobioquímica. Instituto Nacional de Perinatología "Isidro Espinosa de Los Reyes" (INPerIER), Ciudad de México, Mexico
| | - Mariana Victoria López-Marrufo
- Departamento de Ginecología y Obstetricia. Instituto Nacional de Perinatología "Isidro Espinosa de Los Reyes" (INPerIER), Ciudad de México, Mexico
| | - Gerardo Bautista-Bautista
- Departamento de Inmunobioquímica. Instituto Nacional de Perinatología "Isidro Espinosa de Los Reyes" (INPerIER), Ciudad de México, Mexico
| | - Cinthia Selene Velarde-Guerra
- Departamento de Inmunobioquímica. Instituto Nacional de Perinatología "Isidro Espinosa de Los Reyes" (INPerIER), Ciudad de México, Mexico
| | - Graciela Villeda-Gabriel
- Departamento de Inmunología e Infectología, Instituto Nacional de Perinatología "Isidro Espinosa de Los Reyes" (INPerIER), Ciudad de México, Mexico
| | - Oscar Flores-Herrera
- Departamento de Bioquímica, Facultad de Medicina. Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Mauricio Osorio-Caballero
- Departamento de Salud Sexual y Reproductiva. Instituto Nacional de Perinatología "Isidro Espinosa de Los Reyes" (INPerIER), Ciudad de México, Mexico
| | - Jose Carlos Aguilar-Carrasco
- Departamento de Fisiología y Desarrollo Celular, Instituto Nacional de Perinatología "Isidro Espinosa de Los Reyes" (INPerIER), Ciudad de México. Mexico
| | - Martha Leticia Palafox-Vargas
- Departamento de Anatomía Patológica. Instituto Nacional de Perinatología "Isidro Espinosa de Los Reyes" (INPerIER), Ciudad de México, Mexico
| | - Guadalupe García-López
- Departamento de Fisiología y Desarrollo Celular, Instituto Nacional de Perinatología "Isidro Espinosa de Los Reyes" (INPerIER), Ciudad de México. Mexico
| | - Oscar Díaz-Ruíz
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Fabián Arechavaleta-Velasco
- Unidad de Investigación en Medicina Reproductiva. Hospital de Gineco-Obstetricia No. 4 "Luis Castelazo Ayala" Instituto Mexicano Del Seguro Social, Ciudad de México. Mexico.
| | - Hector Flores-Herrera
- Departamento de Inmunobioquímica. Instituto Nacional de Perinatología "Isidro Espinosa de Los Reyes" (INPerIER), Ciudad de México, Mexico.
| |
Collapse
|
4
|
Li ML, Ragupathi A, Patel N, Hernandez T, Magsino J, Werlen G, Brewer G, Jacinto E. The RNA-binding protein AUF1 facilitates Akt phosphorylation at the membrane. J Biol Chem 2022; 298:102437. [PMID: 36041631 PMCID: PMC9513781 DOI: 10.1016/j.jbc.2022.102437] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 08/18/2022] [Accepted: 08/19/2022] [Indexed: 11/25/2022] Open
Abstract
Mammalian target of rapamycin (mTOR), which is part of mTOR complex 1 (mTORC1) and mTORC2, controls cellular metabolism in response to levels of nutrients and other growth signals. A hallmark of mTORC2 activation is the phosphorylation of Akt, which becomes upregulated in cancer. How mTORC2 modulates Akt phosphorylation remains poorly understood. Here, we found that the RNA-binding protein, AUF1 (ARE/poly(U)-binding/degradation factor 1), modulates mTORC2/Akt signaling. We determined that AUF1 is required for phosphorylation of Akt at Thr308, Thr450, and Ser473 and that AUF1 also mediates phosphorylation of the mTORC2-modulated metabolic enzyme glutamine fructose-6-phosphate amidotransferase 1 at Ser243. In addition, AUF1 immunoprecipitation followed by quantitative RT–PCR revealed that the mRNAs of Akt, glutamine fructose-6-phosphate amidotransferase 1, and the mTORC2 component SIN1 associate with AUF1. Furthermore, expression of the p40 and p45, but not the p37 or p42, isoforms of AUF1 specifically mediate Akt phosphorylation. In the absence of AUF1, subcellular fractionation indicated that Akt fails to localize to the membrane. However, ectopic expression of a membrane-targeted allele of Akt is sufficient to allow Akt-Ser473 phosphorylation despite AUF1 depletion. Finally, conditions that enhance mTORC2 signaling, such as acute glutamine withdrawal, augment AUF1 phosphorylation, whereas mTOR inhibition abolishes AUF1 phosphorylation. Our findings unravel a role for AUF1 in promoting membrane localization of Akt to facilitate its phosphorylation on this cellular compartment. Targeting AUF1 could have therapeutic benefit for cancers with upregulated mTORC2/Akt signaling.
Collapse
Affiliation(s)
- Mei-Ling Li
- From the Department of Biochemistry and Molecular Biology, Rutgers Biomedical Health Sciences-Robert Wood Johnson Medical School, 683 Hoes Lane, Piscataway, NJ 08854
| | - Aparna Ragupathi
- From the Department of Biochemistry and Molecular Biology, Rutgers Biomedical Health Sciences-Robert Wood Johnson Medical School, 683 Hoes Lane, Piscataway, NJ 08854
| | - Nikhil Patel
- From the Department of Biochemistry and Molecular Biology, Rutgers Biomedical Health Sciences-Robert Wood Johnson Medical School, 683 Hoes Lane, Piscataway, NJ 08854
| | - Tatiana Hernandez
- From the Department of Biochemistry and Molecular Biology, Rutgers Biomedical Health Sciences-Robert Wood Johnson Medical School, 683 Hoes Lane, Piscataway, NJ 08854
| | - Jedrick Magsino
- From the Department of Biochemistry and Molecular Biology, Rutgers Biomedical Health Sciences-Robert Wood Johnson Medical School, 683 Hoes Lane, Piscataway, NJ 08854
| | - Guy Werlen
- From the Department of Biochemistry and Molecular Biology, Rutgers Biomedical Health Sciences-Robert Wood Johnson Medical School, 683 Hoes Lane, Piscataway, NJ 08854
| | - Gary Brewer
- From the Department of Biochemistry and Molecular Biology, Rutgers Biomedical Health Sciences-Robert Wood Johnson Medical School, 683 Hoes Lane, Piscataway, NJ 08854.
| | - Estela Jacinto
- From the Department of Biochemistry and Molecular Biology, Rutgers Biomedical Health Sciences-Robert Wood Johnson Medical School, 683 Hoes Lane, Piscataway, NJ 08854.
| |
Collapse
|
5
|
Li S, Yang P. Relationship between HSPA1A-regulated gene expression and alternative splicing in mouse cardiomyocytes and cardiac hypertrophy. J Thorac Dis 2021; 13:5517-5533. [PMID: 34659818 PMCID: PMC8482330 DOI: 10.21037/jtd-21-1222] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 08/30/2021] [Indexed: 11/30/2022]
Abstract
Background Cardiac hypertrophy may be classified as either physiological or pathological. Pathological hypertrophy has a complex etiology and is genetically regulated. In this study, we used a mouse model of cardiac hypertrophy to explore the mechanisms of gene regulation, in particular, modulation of the expression of target genes through transcription factor activity, regulation of immune and inflammation-associated genes and regulation of the alternative splicing of transcription factors. Methods Mouse models of pathological cardiac hypertrophy were established by transverse aortic constriction (TAC). We overexpressed HSPA1A in mouse cardiac HL-1 cells. GO and KEGG pathway annotation database was used to analyze all DEGs. Results The expression of HSPA1A differed significantly between TAC + dantrolene vs. sham + dantrolene (Sham was the non-TAC group, and DMSO was the contrast agent), and TAC + DMSO vs. sham + DMSO. The RNA-binding protein Zfp36 was found to be differentially expressed between both TAC + dantrolene vs. sham + dantrolene and TAC + DMSO vs. sham + DMSO. The expression of mki67 and gm5619 was significantly different between TAC + dantrolene and TAC + DMSO. HSPA1A was found to selectively regulate the expression of non-coding RNAs related to cardiac hypertrophy, including Rn7sk and RMRP. The downregulated genes were mainly related to inflammation and the immune response. HSPA1A negatively regulated alternative splicing of Asxl2 and positively regulated alternative splicing of Runx1. Conclusions HSPA1A was closely related to cardiac hypertrophy. Zfp36 was also related to cardiac hypertrophy. Dantrolene may delay cardiac hypertrophy and ventricular remodeling by regulating the expression of the RNA-binding protein genes mki67 and gm5619. HSPA1A positively regulated the expression of the non-coding RNAs RN7SK and RMRP while negatively regulating the expression of inflammation- and immune response-related genes. HSPA1A can play a role in cardiac hypertrophy by regulating the alternative splicing of asxl2 and runx1.
Collapse
Affiliation(s)
- Shuai Li
- Jilin Provincial Precision Medicine Key Laboratory for Cardiovascular Genetic Diagnosis, Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Ping Yang
- Jilin Provincial Precision Medicine Key Laboratory for Cardiovascular Genetic Diagnosis, Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
6
|
Velázquez-Cruz A, Baños-Jaime B, Díaz-Quintana A, De la Rosa MA, Díaz-Moreno I. Post-translational Control of RNA-Binding Proteins and Disease-Related Dysregulation. Front Mol Biosci 2021; 8:658852. [PMID: 33987205 PMCID: PMC8111222 DOI: 10.3389/fmolb.2021.658852] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 03/22/2021] [Indexed: 12/20/2022] Open
Abstract
Cell signaling mechanisms modulate gene expression in response to internal and external stimuli. Cellular adaptation requires a precise and coordinated regulation of the transcription and translation processes. The post-transcriptional control of mRNA metabolism is mediated by the so-called RNA-binding proteins (RBPs), which assemble with specific transcripts forming messenger ribonucleoprotein particles of highly dynamic composition. RBPs constitute a class of trans-acting regulatory proteins with affinity for certain consensus elements present in mRNA molecules. However, these regulators are subjected to post-translational modifications (PTMs) that constantly adjust their activity to maintain cell homeostasis. PTMs can dramatically change the subcellular localization, the binding affinity for RNA and protein partners, and the turnover rate of RBPs. Moreover, the ability of many RBPs to undergo phase transition and/or their recruitment to previously formed membrane-less organelles, such as stress granules, is also regulated by specific PTMs. Interestingly, the dysregulation of PTMs in RBPs has been associated with the pathophysiology of many different diseases. Abnormal PTM patterns can lead to the distortion of the physiological role of RBPs due to mislocalization, loss or gain of function, and/or accelerated or disrupted degradation. This Mini Review offers a broad overview of the post-translational regulation of selected RBPs and the involvement of their dysregulation in neurodegenerative disorders, cancer and other pathologies.
Collapse
Affiliation(s)
- Alejandro Velázquez-Cruz
- Instituto de Investigaciones Químicas, Centro de Investigaciones Científicas Isla de la Cartuja, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas, Seville, Spain
| | - Blanca Baños-Jaime
- Instituto de Investigaciones Químicas, Centro de Investigaciones Científicas Isla de la Cartuja, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas, Seville, Spain
| | - Antonio Díaz-Quintana
- Instituto de Investigaciones Químicas, Centro de Investigaciones Científicas Isla de la Cartuja, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas, Seville, Spain
| | - Miguel A De la Rosa
- Instituto de Investigaciones Químicas, Centro de Investigaciones Científicas Isla de la Cartuja, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas, Seville, Spain
| | - Irene Díaz-Moreno
- Instituto de Investigaciones Químicas, Centro de Investigaciones Científicas Isla de la Cartuja, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas, Seville, Spain
| |
Collapse
|
7
|
Uchida Y, Chiba T, Kurimoto R, Asahara H. Post-transcriptional regulation of inflammation by RNA-binding proteins via cis-elements of mRNAs. J Biochem 2019; 166:375-382. [PMID: 31511872 DOI: 10.1093/jb/mvz067] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 08/26/2019] [Indexed: 12/18/2022] Open
Abstract
In human genome, there are approximately 1,500 RNA-binding proteins (RBPs). They can regulate mRNA stability or translational efficiency via ribosomes and these processes are known as 'post-transcriptional regulation'. Accumulating evidences indicate that post-transcriptional regulation is the determinant of the accurate levels of cytokines mRNAs. While transcriptional regulation of cytokines mRNAs has been well studied and found to be important for the rapid induction of mRNA and regulation of the acute phase of inflammation, post-transcriptional regulation by RBPs is essential for resolving inflammation in the later phase, and their dysfunction may lead to severe autoimmune diseases such as rheumatoid arthritis or systemic lupus erythematosus. For post-transcriptional regulation, RBPs recognize and directly bind to cis-regulatory elements in 3' untranslated region of mRNAs such as AU-rich or constitutive decay elements and play various roles. In this review, we summarize the recent findings regarding the role of RBPs in the regulation of inflammation.
Collapse
Affiliation(s)
- Yutaro Uchida
- Department of Systems BioMedicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Tomoki Chiba
- Department of Systems BioMedicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Ryota Kurimoto
- Department of Systems BioMedicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hiroshi Asahara
- Department of Systems BioMedicine, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
8
|
Murali B, Ren Q, Luo X, Faget DV, Wang C, Johnson RM, Gruosso T, Flanagan KC, Fu Y, Leahy K, Alspach E, Su X, Ross MH, Burnette B, Weilbaecher KN, Park M, Mbalaviele G, Monahan JB, Stewart SA. Inhibition of the Stromal p38MAPK/MK2 Pathway Limits Breast Cancer Metastases and Chemotherapy-Induced Bone Loss. Cancer Res 2018; 78:5618-5630. [PMID: 30093561 PMCID: PMC6168362 DOI: 10.1158/0008-5472.can-18-0234] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 06/25/2018] [Accepted: 08/03/2018] [Indexed: 12/13/2022]
Abstract
The role of the stromal compartment in tumor progression is best illustrated in breast cancer bone metastases, where the stromal compartment supports tumor growth, albeit through poorly defined mechanisms. p38MAPKα is frequently expressed in tumor cells and surrounding stromal cells, and its expression levels correlate with poor prognosis. This observation led us to investigate whether inhibition of p38MAPKα could reduce breast cancer metastases in a clinically relevant model. Orally administered, small-molecule inhibitors of p38MAPKα or its downstream kinase MK2 each limited outgrowth of metastatic breast cancer cells in the bone and visceral organs. This effect was primarily mediated by inhibition of the p38MAPKα pathway within the stromal compartment. Beyond effectively limiting metastatic tumor growth, these inhibitors reduced tumor-associated and chemotherapy-induced bone loss, which is a devastating comorbidity that drastically affects quality of life for patients with cancer. These data underscore the vital role played by stromal-derived factors in tumor progression and identify the p38MAPK-MK2 pathway as a promising therapeutic target for metastatic disease and prevention of tumor-induced bone loss.Significance: Pharmacologically targeting the stromal p38MAPK-MK2 pathway limits metastatic breast cancer growth, preserves bone quality, and extends survival. Cancer Res; 78(19); 5618-30. ©2018 AACR.
Collapse
Affiliation(s)
- Bhavna Murali
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri
| | - Qihao Ren
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri
| | - Xianmin Luo
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri
| | - Douglas V Faget
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri
| | - Chun Wang
- Division of Bone and Mineral Diseases, Washington University School of Medicine, St. Louis, Missouri
| | - Radia Marie Johnson
- Goodman Cancer Center, Department of Oncology, Department of Biochemistry, McGill University, Montreal, Quebec, Canada
| | - Tina Gruosso
- Goodman Cancer Center, Department of Oncology, Department of Biochemistry, McGill University, Montreal, Quebec, Canada
| | - Kevin C Flanagan
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri
| | - Yujie Fu
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri
| | - Kathleen Leahy
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri
| | - Elise Alspach
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri
| | - Xinming Su
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Michael H Ross
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | | | | | - Morag Park
- Goodman Cancer Center, Department of Oncology, Department of Biochemistry, McGill University, Montreal, Quebec, Canada
| | - Gabriel Mbalaviele
- Division of Bone and Mineral Diseases, Washington University School of Medicine, St. Louis, Missouri
| | | | - Sheila A Stewart
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri.
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri
- ICCE Institute, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
9
|
Shrestha A, Pun NT, Park PH. ZFP36L1 and AUF1 Induction Contribute to the Suppression of Inflammatory Mediators Expression by Globular Adiponectin via Autophagy Induction in Macrophages. Biomol Ther (Seoul) 2018; 26:446-457. [PMID: 30001609 PMCID: PMC6131013 DOI: 10.4062/biomolther.2018.078] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 05/18/2018] [Accepted: 05/25/2018] [Indexed: 01/07/2023] Open
Abstract
Adiponectin, a hormone predominantly originated from adipose tissue, has exhibited potent anti-inflammatory properties. Accumulating evidence suggests that autophagy induction plays a crucial role in anti-inflammatory responses by adiponectin. However, underlying molecular mechanisms are still largely unknown. Association of Bcl-2 with Beclin-1, an autophagy activating protein, prevents autophagy induction. We have previously shown that adiponectin-induced autophagy activation is mediated through inhibition of interaction between Bcl-2 and Beclin-1. In the present study, we examined the molecular mechanisms by which adiponectin modulates association of Bcl-2 and Beclin-1 in macrophages. Herein, we demonstrated that globular adiponectin (gAcrp) induced increase in the expression of AUF1 and ZFP36L1, which act as mRNA destabilizing proteins, both in RAW 264.7 macrophages and primary peritoneal macrophages. In addition, gene silencing of AUF1 and ZFP36L1 caused restoration of decrease in Bcl-2 expression and Bcl-2 mRNA half-life by gAcrp, indicating crucial roles of AUF1 and ZFP36L1 induction in Bcl-2 mRNA destabilization by gAcrp. Moreover, knock-down of AUF1 and ZFP36L1 enhanced interaction of Bcl-2 with Beclin-1, and subsequently prevented gAcrp-induced autophagy activation, suggesting that AUF1 and ZFP36L1 induction mediates gAcrp-induced autophagy activation via Bcl-2 mRNA destabilization. Furthermore, suppressive effects of gAcrp on LPS-stimulated inflammatory mediators expression were prevented by gene silencing of AUF1 and ZFP36L1 in macrophages. Taken together, these results suggest that AUF1 and ZFP36L1 induction critically contributes to autophagy induction by gAcrp and are promising targets for anti-inflammatory responses by gAcrp.
Collapse
Affiliation(s)
| | | | - Pil-Hoon Park
- Corresponding Author: E-mail: , Tel: +82-53-810-2826, Fax: +82-53-810-4654
| |
Collapse
|
10
|
Cai HQ, Wang PF, Zhang HP, Cheng ZJ, Li SW, He J, Zhang Y, Hao JJ, Wang MR, Yan CX, Wan JH. Phosphorylated Hsp27 is mutually exclusive with ATRX loss and the IDH1 R132H mutation and may predict better prognosis among glioblastomas without the IDH1 mutation and ATRX loss. J Clin Pathol 2018; 71:702-707. [PMID: 29550762 PMCID: PMC6204978 DOI: 10.1136/jclinpath-2018-205000] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 02/02/2018] [Accepted: 02/05/2018] [Indexed: 12/12/2022]
Abstract
Aim To identify biomarkers for accurate classification of glioma. Patients and methods We evaluated the heat shock protein 27 (Hsp27), phosphorylated Hsp27 (p-Hsp27), ATRX and IDH1R132Hproteins using immunohistochemistry in 421 glioma tissues. The χ2 test was used to assess the relationship between molecular alterations and clinico-pathological parameters. Kaplan-Meier survival curves were constructed, and differences were detected by the log-rank test. Results We found that Hsp27 and p-Hsp27 were mainly expressed in aggressive astrocytic gliomas. However, neither Hsp27 nor p-Hsp27 expression was related to survival time for any grade of glioma. Interestingly, p-Hsp27 was mutually exclusive with ATRX loss (ATRX−) and the IDH1R132H mutation, except for one case of anaplastic astrocytoma. We classified glioblastomas (GBMs) into three subtypes: ATRX−/IDH1R132H, high p-Hsp27 expression (p-Hsp27+) and none of these three markers. ATRX-/IDH1R132Hshowed the longest median survival (19.6 months). The prognostic difference between p-Hsp27+ and none of these three markers was significant (15.0 vs 13.1 months, P=0.045). Moreover, p-Hsp27+ predicted better sensitivity for standard therapy among GBMs without the IDH1 mutation and ATRX loss (26.3 vs 15.5 months, P=0.008). Conclusion p-Hsp27 is a novel biomarker of glioma and might have important clinical value for further classification of patients with wild-type IDH1 and normal ATRX expression, for evaluating prognosis and for guidance for adjuvant therapy.
Collapse
Affiliation(s)
- Hong-Qing Cai
- Department of Neurosurgery, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Peng-Fei Wang
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Hai-Peng Zhang
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Zhi-Jian Cheng
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Shou-Wei Li
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Jie He
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yu Zhang
- State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jia-Jie Hao
- State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ming-Rong Wang
- State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chang-Xiang Yan
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Jing-Hai Wan
- Department of Neurosurgery, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
11
|
Inactivation of p38 MAPK contributes to stem cell-like properties of non-small cell lung cancer. Oncotarget 2018; 8:26702-26717. [PMID: 28460458 PMCID: PMC5432291 DOI: 10.18632/oncotarget.15804] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 02/15/2017] [Indexed: 01/07/2023] Open
Abstract
Cancer stem cells (CSCs) are recognized as the major source for cancer initiation and recurrence. Yet, the mechanism by which the cancer stem cell properties are acquired and maintained in a cancer cell population is not well understood. In the current study, we observed that the level of active p38 MAPK is downregulated, while the level of the stemness marker SOX2 is upregulated in lung cancer tissues as compared to normal tissues. We further demonstrated that inactivation of p38 is a potential mechanism contributing to acquisition and maintenance of cancer stem cell properties in non-small cell lung cancer (NSCLC) cells. p38, in particular the p38γ and p38δ isoforms, suppresses the cancer stem cell properties and tumor initiating ability of NSCLC cells by promoting the ubiquitylation and degradation of stemness proteins such as SOX2, Oct4, Nanog, Klf4 and c-Myc, through MK2-mediated phosphorylation of Hsp27 that is an essential component of the proteasomal degradation machinery. In contrast, inactivation of p38 in lung cancer cells leads to upregulation of the stemness proteins, thus promoting the cancer stem cell properties of these cells. These findings have demonstrated a novel mechanism by which cancer stem cell properties are acquired and maintained in a cancer cell population, and have revealed a new function of the p38 pathway in suppressing cancer development. These studies have also identified a new pathway that can potentially serve as a target for cancer therapies aimed at eliminating CSCs.
Collapse
|
12
|
Kennedy D, Mnich K, Oommen D, Chakravarthy R, Almeida-Souza L, Krols M, Saveljeva S, Doyle K, Gupta S, Timmerman V, Janssens S, Gorman AM, Samali A. HSPB1 facilitates ERK-mediated phosphorylation and degradation of BIM to attenuate endoplasmic reticulum stress-induced apoptosis. Cell Death Dis 2017; 8:e3026. [PMID: 29048431 PMCID: PMC5596589 DOI: 10.1038/cddis.2017.408] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 06/30/2017] [Accepted: 07/06/2017] [Indexed: 01/11/2023]
Abstract
BIM, a pro-apoptotic BH3-only protein, is a key regulator of the intrinsic (or mitochondrial) apoptosis pathway. Here, we show that BIM induction by endoplasmic reticulum (ER) stress is suppressed in rat PC12 cells overexpressing heat shock protein B1 (HSPB1 or HSP27) and that this is due to enhanced proteasomal degradation of BIM. HSPB1 and BIM form a complex that immunoprecipitates with p-ERK1/2. We found that HSPB1-mediated proteasomal degradation of BIM is dependent on MEK-ERK signaling. Other studies have shown that several missense mutations in HSPB1 cause the peripheral neuropathy, Charcot-Marie-Tooth (CMT) disease, which is associated with nerve degeneration. Here we show that cells overexpressing CMT-related HSPB1 mutants exhibited increased susceptibility to ER stress-induced cell death and high levels of BIM. These findings identify a novel function for HSPB1 as a negative regulator of BIM protein stability leading to protection against ER stress-induced apoptosis, a function that is absent in CMT-associated HSPB1 mutants.
Collapse
Affiliation(s)
- Donna Kennedy
- Apoptosis Research Centre, Biomedical Sciences, NUI Galway, Galway, Ireland
| | - Katarzyna Mnich
- Apoptosis Research Centre, Biomedical Sciences, NUI Galway, Galway, Ireland
| | - Deepu Oommen
- Apoptosis Research Centre, Biomedical Sciences, NUI Galway, Galway, Ireland
| | - Reka Chakravarthy
- Apoptosis Research Centre, Biomedical Sciences, NUI Galway, Galway, Ireland
| | - Leonardo Almeida-Souza
- Peripheral Neuropathy Research Group, University of Antwerp, Antwerpen, Belgium.,Institute Born Bunge, Antwerpen, Belgium
| | - Michiel Krols
- Peripheral Neuropathy Research Group, University of Antwerp, Antwerpen, Belgium.,Institute Born Bunge, Antwerpen, Belgium
| | - Svetlana Saveljeva
- Apoptosis Research Centre, Biomedical Sciences, NUI Galway, Galway, Ireland
| | - Karen Doyle
- Discipline of Physiology, NUI Galway, Galway, Ireland
| | - Sanjeev Gupta
- Discipline of Pathology, School of Medicine, NUI Galway, Galway, Ireland
| | - Vincent Timmerman
- Peripheral Neuropathy Research Group, University of Antwerp, Antwerpen, Belgium.,Institute Born Bunge, Antwerpen, Belgium
| | - Sophie Janssens
- Unit Immunoregulation and Mucosal Immunology, VIB Inflammation Research Centre, Ghent University, Gent, Belgium.,Department of Internal Medicine, Ghent University, Gent, Belgium
| | - Adrienne M Gorman
- Apoptosis Research Centre, Biomedical Sciences, NUI Galway, Galway, Ireland
| | - Afshin Samali
- Apoptosis Research Centre, Biomedical Sciences, NUI Galway, Galway, Ireland
| |
Collapse
|
13
|
HSPB1 mutations causing hereditary neuropathy in humans disrupt non-cell autonomous protection of motor neurons. Exp Neurol 2017; 297:101-109. [PMID: 28797631 DOI: 10.1016/j.expneurol.2017.08.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 07/22/2017] [Accepted: 08/06/2017] [Indexed: 12/12/2022]
Abstract
Heat shock protein beta-1 (HSPB1), is a ubiquitously expressed, multifunctional protein chaperone. Mutations in HSPB1 result in the development of a late-onset, distal hereditary motor neuropathy type II (dHMN) and axonal Charcot-Marie Tooth disease with sensory involvement (CMT2F). The functional consequences of HSPB1 mutations associated with hereditary neuropathy are unknown. HSPB1 also displays neuroprotective properties in many neuronal disease models, including the motor neuron disease amyotrophic lateral sclerosis (ALS). HSPB1 is upregulated in SOD1-ALS animal models during disease progression, predominately in glial cells. Glial cells are known to contribute to motor neuron loss in ALS through a non-cell autonomous mechanism. In this study, we examined the non-cell autonomous role of wild type and mutant HSPB1 in an astrocyte-motor neuron co-culture model system of ALS. Astrocyte-specific overexpression of wild type HSPB1 was sufficient to attenuate SOD1(G93A) astrocyte-mediated toxicity in motor neurons, whereas, overexpression of mutHSPB1 failed to ameliorate motor neuron toxicity. Expression of a phosphomimetic HSPB1 mutant in SOD1(G93A) astrocytes also reduced toxicity to motor neurons, suggesting that phosphorylation may contribute to HSPB1 mediated-neuroprotection. These data provide evidence that astrocytic HSPB1 expression may play a central role in motor neuron health and maintenance.
Collapse
|
14
|
Hadadi E, Zhang B, Baidžajevas K, Yusof N, Puan KJ, Ong SM, Yeap WH, Rotzschke O, Kiss-Toth E, Wilson H, Wong SC. Differential IL-1β secretion by monocyte subsets is regulated by Hsp27 through modulating mRNA stability. Sci Rep 2016; 6:39035. [PMID: 27976724 PMCID: PMC5157043 DOI: 10.1038/srep39035] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 11/16/2016] [Indexed: 12/11/2022] Open
Abstract
Monocytes play a central role in regulating inflammation in response to infection or injury, and during auto-inflammatory diseases. Human blood contains classical, intermediate and non-classical monocyte subsets that each express characteristic patterns of cell surface CD16 and CD14; each subset also has specific functional properties, but the mechanisms underlying many of their distinctive features are undefined. Of particular interest is how monocyte subsets regulate secretion of the apical pro-inflammatory cytokine IL-1β, which is central to the initiation of immune responses but is also implicated in the pathology of various auto-immune/auto-inflammatory conditions. Here we show that primary human non-classical monocytes, exposed to LPS or LPS + BzATP (3'-O-(4-benzoyl)benzyl-ATP, a P2X7R agonist), produce approx. 80% less IL-1β than intermediate or classical monocytes. Despite their low CD14 expression, LPS-sensing, caspase-1 activation and P2X7R activity were comparable in non-classical monocytes to other subsets: their diminished ability to produce IL-1β instead arose from 50% increased IL-1β mRNA decay rates, mediated by Hsp27. These findings identify the Hsp27 pathway as a novel therapeutic target for the management of conditions featuring dysregulated IL-1β production, and represent an advancement in understanding of both physiological inflammatory responses and the pathogenesis of inflammatory diseases involving monocyte-derived IL-1β.
Collapse
Affiliation(s)
- Eva Hadadi
- University of Sheffield, Dept of Infection, Immunity &Cardiovascular Disease (IICD), Sheffield, UK.,Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (ASTAR), Singapore
| | - Biyan Zhang
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (ASTAR), Singapore
| | - Kajus Baidžajevas
- University of Sheffield, Dept of Infection, Immunity &Cardiovascular Disease (IICD), Sheffield, UK
| | - Nurhashikin Yusof
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (ASTAR), Singapore
| | - Kia Joo Puan
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (ASTAR), Singapore
| | - Siew Min Ong
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (ASTAR), Singapore
| | - Wei Hseun Yeap
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (ASTAR), Singapore
| | - Olaf Rotzschke
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (ASTAR), Singapore
| | - Endre Kiss-Toth
- University of Sheffield, Dept of Infection, Immunity &Cardiovascular Disease (IICD), Sheffield, UK
| | - Heather Wilson
- University of Sheffield, Dept of Infection, Immunity &Cardiovascular Disease (IICD), Sheffield, UK
| | - Siew Cheng Wong
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (ASTAR), Singapore
| |
Collapse
|
15
|
Batulan Z, Pulakazhi Venu VK, Li Y, Koumbadinga G, Alvarez-Olmedo DG, Shi C, O'Brien ER. Extracellular Release and Signaling by Heat Shock Protein 27: Role in Modifying Vascular Inflammation. Front Immunol 2016; 7:285. [PMID: 27507972 PMCID: PMC4960997 DOI: 10.3389/fimmu.2016.00285] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 07/14/2016] [Indexed: 12/31/2022] Open
Abstract
Heat shock protein 27 (HSP27) is traditionally viewed as an intracellular chaperone protein with anti-apoptotic properties. However, recent data indicate that a number of heat shock proteins, including HSP27, are also found in the extracellular space where they may signal via membrane receptors to alter gene transcription and cellular function. Therefore, there is increasing interest in better understanding how HSP27 is released from cells, its levels and composition in the extracellular space, and the cognate cell membrane receptors involved in effecting cell signaling. In this paper, the knowledge to date, as well as some emerging paradigms about the extracellular function of HSP27 is presented. Of particular interest is the role of HSP27 in attenuating atherogenesis by modifying lipid uptake and inflammation in the plaque. Moreover, the abundance of HSP27 in serum is an emerging new biomarker for ischemic events. Finally, HSP27 replacement therapy may represent a novel therapeutic opportunity for chronic inflammatory disorders, such as atherosclerosis.
Collapse
Affiliation(s)
- Zarah Batulan
- Vascular Biology Laboratory, Health Research Innovation Centre, Libin Cardiovascular Institute of Alberta, University of Calgary Cumming School of Medicine , Calgary, AB , Canada
| | - Vivek Krishna Pulakazhi Venu
- Vascular Biology Laboratory, Health Research Innovation Centre, Libin Cardiovascular Institute of Alberta, University of Calgary Cumming School of Medicine , Calgary, AB , Canada
| | - Yumei Li
- Vascular Biology Laboratory, Health Research Innovation Centre, Libin Cardiovascular Institute of Alberta, University of Calgary Cumming School of Medicine , Calgary, AB , Canada
| | - Geremy Koumbadinga
- Vascular Biology Laboratory, Health Research Innovation Centre, Libin Cardiovascular Institute of Alberta, University of Calgary Cumming School of Medicine , Calgary, AB , Canada
| | - Daiana Gisela Alvarez-Olmedo
- Oncology Laboratory, Institute for Experimental Medicine and Biology of Cuyo (IMBECU), CCT CONICET , Mendoza , Argentina
| | - Chunhua Shi
- Vascular Biology Laboratory, Health Research Innovation Centre, Libin Cardiovascular Institute of Alberta, University of Calgary Cumming School of Medicine , Calgary, AB , Canada
| | - Edward R O'Brien
- Vascular Biology Laboratory, Health Research Innovation Centre, Libin Cardiovascular Institute of Alberta, University of Calgary Cumming School of Medicine , Calgary, AB , Canada
| |
Collapse
|
16
|
Corcoran JA, McCormick C. Viral activation of stress-regulated Rho-GTPase signaling pathway disrupts sites of mRNA degradation to influence cellular gene expression. Small GTPases 2015; 6:178-85. [PMID: 26480288 PMCID: PMC4905259 DOI: 10.1080/21541248.2015.1093068] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Revised: 09/07/2015] [Accepted: 09/08/2015] [Indexed: 01/09/2023] Open
Abstract
Viruses are useful tools that often reveal previously unrecognized levels of control within a cell. By studying the oncogenic Kaposi's sarcoma-associated herpesvirus (KSHV), we discovered a new signaling axis in endothelial cells (ECs) that links actin cytoskeleton dynamics to post-transcriptional control of gene expression. Translational repression and rapid decay of mRNAs containing AU-rich elements (AREs) occurs in cytoplasmic RNA granules known as processing bodies (PBs). Rho-GTPase activity influences PB dynamics but mechanistic details remain obscure. We have previously shown that the KSHV Kaposin B protein blocks the degradation of ARE-mRNAs that encode potent cytokines and angiogenic factors, at least in part by preventing PB formation. Moreover, Kaposin B is sufficient to cause marked alterations in endothelial cell physiology including the formation of long parallel actin stress fibers and accelerated migration and angiogenic phenotypes. All of these phenotypes depend on Kaposin B-mediated activation of a non-canonical signaling pathway comprising the stress-inducible kinase MK2, hsp27, p115RhoGEF and RhoA. Accelerated endothelial cell migration and angiogenesis depends on the subsequent activation of the RhoA-dependent kinase ROCK, but PB disruption is ROCK-independent. In this Commentary, we discuss implications of the activation of this signaling axis, and propose mechanistic links between RhoA activation and PB dynamics.
Collapse
Affiliation(s)
- Jennifer A Corcoran
- Department of Microbiology and Immunology; Dalhousie University; Halifax NS, Canada
| | - Craig McCormick
- Department of Microbiology and Immunology; Dalhousie University; Halifax NS, Canada
| |
Collapse
|
17
|
Isacke CM, Barcellos-Hoff MH. Soil amendments that slow cancer growth. Cancer Discov 2015; 4:637-9. [PMID: 24891361 DOI: 10.1158/2159-8290.cd-14-0416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The recognition that the tumor microenvironment contributes to tumor survival, growth, and response to therapy provides the rationale for considering it a therapeutic target. The article by Alspach and colleagues in this issue provides evidence that p38MAPK acts posttranscriptionally to promote the tumor-permissive secretory phenotype of both cancer-associated and senescent fibroblasts, and that p38MAPK inhibitors already in clinical trials have significant therapeutic potential.
Collapse
Affiliation(s)
- Clare M Isacke
- Authors' Affiliations:Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London, United Kingdom; and Departments of Radiation Oncology and Cell Biology, New York University School of Medicine, New York, New York
| | - Mary Helen Barcellos-Hoff
- Authors' Affiliations:Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London, United Kingdom; and Departments of Radiation Oncology and Cell Biology, New York University School of Medicine, New York, New York
| |
Collapse
|
18
|
Mutant glucocerebrosidase in Gaucher disease recruits Hsp27 to the Hsp90 chaperone complex for proteasomal degradation. Proc Natl Acad Sci U S A 2015; 112:1137-42. [PMID: 25583479 DOI: 10.1073/pnas.1424288112] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Gaucher disease is caused by mutations of the GBA1 gene, which encodes the lysosomal anchored gluococerebrosidase (GCase). GBA1 mutations commonly result in protein misfolding, abnormal chaperone recognition, and premature degradation, but are less likely to affect catalytic activity. In the present study, we demonstrate that the Hsp90/HOP/Cdc37 complex recruits Hsp27 after recognition of GCase mutants with subsequent targeting of GCase mutant peptides to degradation mechanisms such as VCP and the 26S proteasome. Inhibition of Hsp27 not only increased the quantity of enzyme but also enhanced GCase activity in fibroblasts derived from patients with Gaucher disease. These findings provide insight into a possible therapeutic strategy for protein misfolding diseases by correcting chaperone binding and altering subsequent downstream patterns of protein degradation.
Collapse
|
19
|
Corcoran JA, Johnston BP, McCormick C. Viral activation of MK2-hsp27-p115RhoGEF-RhoA signaling axis causes cytoskeletal rearrangements, p-body disruption and ARE-mRNA stabilization. PLoS Pathog 2015; 11:e1004597. [PMID: 25569678 PMCID: PMC4287613 DOI: 10.1371/journal.ppat.1004597] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Accepted: 12/02/2014] [Indexed: 11/30/2022] Open
Abstract
Kaposi's sarcoma-associated herpesvirus (KSHV) is the infectious cause of several AIDS-related cancers, including the endothelial cell (EC) neoplasm Kaposi's sarcoma (KS). KSHV-infected ECs secrete abundant host-derived pro-inflammatory molecules and angiogenic factors that contribute to tumorigenesis. The precise contributions of viral gene products to this secretory phenotype remain to be elucidated, but there is emerging evidence for post-transcriptional regulation. The Kaposin B (KapB) protein is thought to contribute to the secretory phenotype in infected cells by binding and activating the stress-responsive kinase MK2, thereby selectively blocking decay of AU-rich mRNAs (ARE-mRNAs) encoding pro-inflammatory cytokines and angiogenic factors. Processing bodies (PBs) are cytoplasmic ribonucleoprotein foci in which ARE-mRNAs normally undergo rapid 5′ to 3′ decay. Here, we demonstrate that PB dispersion is a feature of latent KSHV infection, which is dependent on kaposin protein expression. KapB is sufficient to disperse PBs, and KapB-mediated ARE-mRNA stabilization could be partially reversed by treatments that restore PBs. Using a combination of genetic and chemical approaches we provide evidence that KapB-mediated PB dispersion is dependent on activation of a non-canonical Rho-GTPase signaling axis involving MK2, hsp27, p115RhoGEF and RhoA. PB dispersion in latently infected cells is likewise dependent on p115RhoGEF. In addition to PB dispersion, KapB-mediated RhoA activation in primary ECs caused actin stress fiber formation, increased cell motility and angiogenesis; these effects were dependent on the activity of the RhoA substrate kinases ROCK1/2. By contrast, KapB-mediated PB dispersion occurred in a ROCK1/2-independent manner. Taken together, these observations position KapB as a key contributor to viral reprogramming of ECs, capable of eliciting many of the phenotypes characteristic of KS tumor cells, and strongly contributing to the post-transcriptional control of EC gene expression and secretion. We have only scratched the surface in understanding how viruses control host gene expression. Several viruses disrupt important sites of post-transcriptional control of gene expression known as processing bodies (PBs), but underlying regulatory mechanisms and biological relevance remain poorly understood in most cases. Our study shows that the Kaposin B (KapB) protein of Kaposi's sarcoma (KS)-associated herpesvirus, known to block the degradation of a class of labile host mRNAs, does so by constitutively activating a signaling axis involving MK2, hsp27, p115RhoGEF and RhoA, thereby dispersing PBs. Thus, PB disruption may support the secretion of host pro-inflammatory cytokines and angiogenic factors that underlies KS tumor formation. Furthermore, by activating RhoA, KapB also causes cytoskeletal rearrangements, accelerated cell migration and angiogenesis in an endothelial cell model. Our findings position KapB as a key contributor to viral reprogramming of endothelial cells.
Collapse
Affiliation(s)
- Jennifer A. Corcoran
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
- Beatrice Hunter Cancer Research Institute, Halifax, Nova Scotia, Canada
| | - Benjamin P. Johnston
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
- Beatrice Hunter Cancer Research Institute, Halifax, Nova Scotia, Canada
| | - Craig McCormick
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
- Beatrice Hunter Cancer Research Institute, Halifax, Nova Scotia, Canada
- * E-mail:
| |
Collapse
|
20
|
Arrigo AP, Ducarouge B, Lavial F, Gibert B. Immense Cellular Implications Associated to Small Stress Proteins Expression: Impacts on Human Pathologies. HEAT SHOCK PROTEINS 2015. [DOI: 10.1007/978-3-319-16077-1_2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
21
|
Lin JY, Li ML, Brewer G. mRNA decay factor AUF1 binds the internal ribosomal entry site of enterovirus 71 and inhibits virus replication. PLoS One 2014; 9:e103827. [PMID: 25077793 PMCID: PMC4117571 DOI: 10.1371/journal.pone.0103827] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Accepted: 07/07/2014] [Indexed: 02/05/2023] Open
Abstract
AU-rich element binding factor 1 (AUF1) has a role in the replication cycles of different viruses. Here we demonstrate that AUF1 binds the internal ribosome entry site (IRES) of enterovirus 71 (EV71) and negatively regulates IRES-dependent translation. During EV71 infection, AUF1 accumulates in the cytoplasm where viral replication occurs, whereas AUF1 localizes predominantly in the nucleus in mock-infected cells. AUF1 knockdown in infected cells increases IRES activity and synthesis of viral proteins. Taken together, the results suggest that AUF1 interacts with the EV71 IRES to negatively regulate viral translation and replication.
Collapse
Affiliation(s)
- Jing-Yi Lin
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Mei-Ling Li
- Department of Biochemistry & Molecular Biology, Rutgers Robert Wood Johnson Medical School, Piscataway, New Jersey, United States of America
| | - Gary Brewer
- Department of Biochemistry & Molecular Biology, Rutgers Robert Wood Johnson Medical School, Piscataway, New Jersey, United States of America
| |
Collapse
|
22
|
Wang R, Ferraris JD, Izumi Y, Dmitrieva N, Ramkissoon K, Wang G, Gucek M, Burg MB. Global discovery of high-NaCl-induced changes of protein phosphorylation. Am J Physiol Cell Physiol 2014; 307:C442-54. [PMID: 24965592 DOI: 10.1152/ajpcell.00379.2013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
High extracellular NaCl, such as in the renal medulla, can perturb and even kill cells, but cells mount protective responses that enable them to survive and function. Many high-NaCl-induced perturbations and protective responses are known, but the signaling pathways involved are less clear. Change in protein phosphorylation is a common mode of cell signaling, but there was no unbiased survey of protein phosphorylation in response to high NaCl. We used stable isotopic labeling of amino acids in cell culture coupled to mass spectrometry to identify changes in protein phosphorylation in human embryonic kidney (HEK 293) cells exposed to high NaCl. We reproducibly identify >8,000 unique phosphopeptides in 4 biological replicate samples with a 1% false discovery rate. High NaCl significantly changed phosphorylation of 253 proteins. Western analysis and targeted ion selection mass spectrometry confirm a representative sample of the phosphorylation events. We analyze the affected proteins by functional category to infer how altered protein phosphorylation might signal cellular responses to high NaCl, including alteration of cell cycle, cyto/nucleoskeletal organization, DNA double-strand breaks, transcription, proteostasis, metabolism of mRNA, and cell death.
Collapse
Affiliation(s)
- Rong Wang
- Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Joan D Ferraris
- Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Yuichiro Izumi
- Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Natalia Dmitrieva
- Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Kevin Ramkissoon
- Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Guanghui Wang
- Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Marjan Gucek
- Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Maurice B Burg
- Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
23
|
Moore AE, Chenette DM, Larkin LC, Schneider RJ. Physiological networks and disease functions of RNA-binding protein AUF1. WILEY INTERDISCIPLINARY REVIEWS-RNA 2014; 5:549-64. [PMID: 24687816 DOI: 10.1002/wrna.1230] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 02/20/2014] [Accepted: 02/24/2014] [Indexed: 11/09/2022]
Abstract
Regulated messenger RNA (mRNA) decay is an essential mechanism that governs proper control of gene expression. In fact, many of the most physiologically potent proteins are encoded by short-lived mRNAs, many of which contain AU-rich elements (AREs) in their 3'-untranslated region (3'-UTR). AREs target mRNAs for post-transcriptional regulation, generally rapid decay, but also stabilization and translation inhibition. AREs control mRNA turnover and translation activities through association with trans-acting RNA-binding proteins that display high affinity for these AU-rich regulatory elements. AU-rich element RNA-binding protein (AUF1), also known as heterogeneous nuclear ribonucleoprotein D (HNRNPD), is an extensively studied AU-rich binding protein (AUBP). AUF1 has been shown to regulate ARE-mRNA turnover, primarily functioning to promote rapid ARE-mRNA degradation. In certain cellular contexts, AUF1 has also been shown to regulate gene expression at the translational and even the transcriptional level. AUF1 comprises a family of four related protein isoforms derived from a common pre-mRNA by differential exon splicing. AUF1 isoforms have been shown to display multiple and distinct functions that include the ability to target ARE-mRNA stability or decay, and transcriptional activation of certain genes that is controlled by their differential subcellular locations, expression levels, and post-translational modifications. AUF1 has been implicated in controlling a variety of physiological functions through its ability to regulate the expression of numerous mRNAs containing 3'-UTR AREs, thereby coordinating functionally related pathways. This review highlights the physiological functions of AUF1-mediated regulation of mRNA and gene expression, and the consequences of deficient AUF1 levels in different physiological settings.
Collapse
Affiliation(s)
- Ashleigh E Moore
- Alexandria Center for Life Sciences, New York University School of Medicine, New York, NY, USA
| | | | | | | |
Collapse
|
24
|
Alspach E, Flanagan KC, Luo X, Ruhland MK, Huang H, Pazolli E, Donlin MJ, Marsh T, Piwnica-Worms D, Monahan J, Novack DV, McAllister SS, Stewart SA. p38MAPK plays a crucial role in stromal-mediated tumorigenesis. Cancer Discov 2014; 4:716-29. [PMID: 24670723 DOI: 10.1158/2159-8290.cd-13-0743] [Citation(s) in RCA: 132] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
UNLABELLED Neoplastic cells rely on the tumor microenvironment (TME) for survival and progression factors. Indeed, senescent and cancer-associated fibroblasts (CAF) express factors that promote tumorigenesis that are collectively referred to as the senescence-associated secretory phenotype (SASP). Despite their importance in tumorigenesis, the mechanisms that control TME-derived factor expression remain poorly understood. Here, we address a key unanswered question: how the SASP is sustained in senescent fibroblasts and CAFs. We find that the mitogen-activated protein kinase p38 (p38MAPK) controls AUF1 occupancy on SASP mRNAs and thus controls their stability. The importance of this regulatory mechanism is underscored by our findings that stromal-specific p38MAPK inhibition abrogates the tumor-promoting activities of CAFs and senescent fibroblasts. Our data suggest that targeting SASP mRNA stability through inhibition of p38MAPK will significantly aid the development of clinical strategies to target the TME. SIGNIFICANCE The TME plays a key role in tumorigenesis. We demonstrate that p38MAPK governs a posttranscriptional mechanism that sustains the protumorigenic SASP. Inhibition of p38MAPK abrogates the tumor-promoting activities of CAFs and senescent fibroblasts. Thus, p38MAPK is a TME-specific Achilles' heel that may be exploited as a new therapeutic target.
Collapse
Affiliation(s)
- Elise Alspach
- Authors' Affiliations: Departments of Cell Biology and Physiology, Medicine, and Pathology and Immunology; BRIGHT Institute, Washington University School of Medicine; Departments of Biochemistry & Molecular Biology and Molecular Microbiology & Immunology, Saint Louis University School of Medicine; Confluence Life Sciences, Inc., St. Louis, Missouri; Hematology Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston; Harvard Stem Cell Institute and Broad Institute of Harvard and MIT, Cambridge, Massachusetts; and Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TexasAuthors' Affiliations: Departments of Cell Biology and Physiology, Medicine, and Pathology and Immunology; BRIGHT Institute, Washington University School of Medicine; Departments of Biochemistry & Molecular Biology and Molecular Microbiology & Immunology, Saint Louis University School of Medicine; Confluence Life Sciences, Inc., St. Louis, Missouri; Hematology Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston; Harvard Stem Cell Institute and Broad Institute of Harvard and MIT, Cambridge, Massachusetts; and Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Kevin C Flanagan
- Authors' Affiliations: Departments of Cell Biology and Physiology, Medicine, and Pathology and Immunology; BRIGHT Institute, Washington University School of Medicine; Departments of Biochemistry & Molecular Biology and Molecular Microbiology & Immunology, Saint Louis University School of Medicine; Confluence Life Sciences, Inc., St. Louis, Missouri; Hematology Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston; Harvard Stem Cell Institute and Broad Institute of Harvard and MIT, Cambridge, Massachusetts; and Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TexasAuthors' Affiliations: Departments of Cell Biology and Physiology, Medicine, and Pathology and Immunology; BRIGHT Institute, Washington University School of Medicine; Departments of Biochemistry & Molecular Biology and Molecular Microbiology & Immunology, Saint Louis University School of Medicine; Confluence Life Sciences, Inc., St. Louis, Missouri; Hematology Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston; Harvard Stem Cell Institute and Broad Institute of Harvard and MIT, Cambridge, Massachusetts; and Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Xianmin Luo
- Authors' Affiliations: Departments of Cell Biology and Physiology, Medicine, and Pathology and Immunology; BRIGHT Institute, Washington University School of Medicine; Departments of Biochemistry & Molecular Biology and Molecular Microbiology & Immunology, Saint Louis University School of Medicine; Confluence Life Sciences, Inc., St. Louis, Missouri; Hematology Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston; Harvard Stem Cell Institute and Broad Institute of Harvard and MIT, Cambridge, Massachusetts; and Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TexasAuthors' Affiliations: Departments of Cell Biology and Physiology, Medicine, and Pathology and Immunology; BRIGHT Institute, Washington University School of Medicine; Departments of Biochemistry & Molecular Biology and Molecular Microbiology & Immunology, Saint Louis University School of Medicine; Confluence Life Sciences, Inc., St. Louis, Missouri; Hematology Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston; Harvard Stem Cell Institute and Broad Institute of Harvard and MIT, Cambridge, Massachusetts; and Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Megan K Ruhland
- Authors' Affiliations: Departments of Cell Biology and Physiology, Medicine, and Pathology and Immunology; BRIGHT Institute, Washington University School of Medicine; Departments of Biochemistry & Molecular Biology and Molecular Microbiology & Immunology, Saint Louis University School of Medicine; Confluence Life Sciences, Inc., St. Louis, Missouri; Hematology Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston; Harvard Stem Cell Institute and Broad Institute of Harvard and MIT, Cambridge, Massachusetts; and Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TexasAuthors' Affiliations: Departments of Cell Biology and Physiology, Medicine, and Pathology and Immunology; BRIGHT Institute, Washington University School of Medicine; Departments of Biochemistry & Molecular Biology and Molecular Microbiology & Immunology, Saint Louis University School of Medicine; Confluence Life Sciences, Inc., St. Louis, Missouri; Hematology Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston; Harvard Stem Cell Institute and Broad Institute of Harvard and MIT, Cambridge, Massachusetts; and Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Hui Huang
- Authors' Affiliations: Departments of Cell Biology and Physiology, Medicine, and Pathology and Immunology; BRIGHT Institute, Washington University School of Medicine; Departments of Biochemistry & Molecular Biology and Molecular Microbiology & Immunology, Saint Louis University School of Medicine; Confluence Life Sciences, Inc., St. Louis, Missouri; Hematology Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston; Harvard Stem Cell Institute and Broad Institute of Harvard and MIT, Cambridge, Massachusetts; and Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TexasAuthors' Affiliations: Departments of Cell Biology and Physiology, Medicine, and Pathology and Immunology; BRIGHT Institute, Washington University School of Medicine; Departments of Biochemistry & Molecular Biology and Molecular Microbiology & Immunology, Saint Louis University School of Medicine; Confluence Life Sciences, Inc., St. Louis, Missouri; Hematology Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston; Harvard Stem Cell Institute and Broad Institute of Harvard and MIT, Cambridge, Massachusetts; and Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ermira Pazolli
- Authors' Affiliations: Departments of Cell Biology and Physiology, Medicine, and Pathology and Immunology; BRIGHT Institute, Washington University School of Medicine; Departments of Biochemistry & Molecular Biology and Molecular Microbiology & Immunology, Saint Louis University School of Medicine; Confluence Life Sciences, Inc., St. Louis, Missouri; Hematology Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston; Harvard Stem Cell Institute and Broad Institute of Harvard and MIT, Cambridge, Massachusetts; and Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Maureen J Donlin
- Authors' Affiliations: Departments of Cell Biology and Physiology, Medicine, and Pathology and Immunology; BRIGHT Institute, Washington University School of Medicine; Departments of Biochemistry & Molecular Biology and Molecular Microbiology & Immunology, Saint Louis University School of Medicine; Confluence Life Sciences, Inc., St. Louis, Missouri; Hematology Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston; Harvard Stem Cell Institute and Broad Institute of Harvard and MIT, Cambridge, Massachusetts; and Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TexasAuthors' Affiliations: Departments of Cell Biology and Physiology, Medicine, and Pathology and Immunology; BRIGHT Institute, Washington University School of Medicine; Departments of Biochemistry & Molecular Biology and Molecular Microbiology & Immunology, Saint Louis University School of Medicine; Confluence Life Sciences, Inc., St. Louis, Missouri; Hematology Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston; Harvard Stem Cell Institute and Broad Institute of Harvard and MIT, Cambridge, Massachusetts; and Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Timothy Marsh
- Authors' Affiliations: Departments of Cell Biology and Physiology, Medicine, and Pathology and Immunology; BRIGHT Institute, Washington University School of Medicine; Departments of Biochemistry & Molecular Biology and Molecular Microbiology & Immunology, Saint Louis University School of Medicine; Confluence Life Sciences, Inc., St. Louis, Missouri; Hematology Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston; Harvard Stem Cell Institute and Broad Institute of Harvard and MIT, Cambridge, Massachusetts; and Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - David Piwnica-Worms
- Authors' Affiliations: Departments of Cell Biology and Physiology, Medicine, and Pathology and Immunology; BRIGHT Institute, Washington University School of Medicine; Departments of Biochemistry & Molecular Biology and Molecular Microbiology & Immunology, Saint Louis University School of Medicine; Confluence Life Sciences, Inc., St. Louis, Missouri; Hematology Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston; Harvard Stem Cell Institute and Broad Institute of Harvard and MIT, Cambridge, Massachusetts; and Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Joseph Monahan
- Authors' Affiliations: Departments of Cell Biology and Physiology, Medicine, and Pathology and Immunology; BRIGHT Institute, Washington University School of Medicine; Departments of Biochemistry & Molecular Biology and Molecular Microbiology & Immunology, Saint Louis University School of Medicine; Confluence Life Sciences, Inc., St. Louis, Missouri; Hematology Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston; Harvard Stem Cell Institute and Broad Institute of Harvard and MIT, Cambridge, Massachusetts; and Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Deborah V Novack
- Authors' Affiliations: Departments of Cell Biology and Physiology, Medicine, and Pathology and Immunology; BRIGHT Institute, Washington University School of Medicine; Departments of Biochemistry & Molecular Biology and Molecular Microbiology & Immunology, Saint Louis University School of Medicine; Confluence Life Sciences, Inc., St. Louis, Missouri; Hematology Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston; Harvard Stem Cell Institute and Broad Institute of Harvard and MIT, Cambridge, Massachusetts; and Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TexasAuthors' Affiliations: Departments of Cell Biology and Physiology, Medicine, and Pathology and Immunology; BRIGHT Institute, Washington University School of Medicine; Departments of Biochemistry & Molecular Biology and Molecular Microbiology & Immunology, Saint Louis University School of Medicine; Confluence Life Sciences, Inc., St. Louis, Missouri; Hematology Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston; Harvard Stem Cell Institute and Broad Institute of Harvard and MIT, Cambridge, Massachusetts; and Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sandra S McAllister
- Authors' Affiliations: Departments of Cell Biology and Physiology, Medicine, and Pathology and Immunology; BRIGHT Institute, Washington University School of Medicine; Departments of Biochemistry & Molecular Biology and Molecular Microbiology & Immunology, Saint Louis University School of Medicine; Confluence Life Sciences, Inc., St. Louis, Missouri; Hematology Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston; Harvard Stem Cell Institute and Broad Institute of Harvard and MIT, Cambridge, Massachusetts; and Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TexasAuthors' Affiliations: Departments of Cell Biology and Physiology, Medicine, and Pathology and Immunology; BRIGHT Institute, Washington University School of Medicine; Departments of Biochemistry & Molecular Biology and Molecular Microbiology & Immunology, Saint Louis University School of Medicine; Confluence Life Sciences, Inc., St. Louis, Missouri; Hematology Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston; Harvard Stem Cell Institute and Broad Institute of Harvard and MIT, Cambridge, Massachusetts; and Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sheila A Stewart
- Authors' Affiliations: Departments of Cell Biology and Physiology, Medicine, and Pathology and Immunology; BRIGHT Institute, Washington University School of Medicine; Departments of Biochemistry & Molecular Biology and Molecular Microbiology & Immunology, Saint Louis University School of Medicine; Confluence Life Sciences, Inc., St. Louis, Missouri; Hematology Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston; Harvard Stem Cell Institute and Broad Institute of Harvard and MIT, Cambridge, Massachusetts; and Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TexasAuthors' Affiliations: Departments of Cell Biology and Physiology, Medicine, and Pathology and Immunology; BRIGHT Institute, Washington University School of Medicine; Departments of Biochemistry & Molecular Biology and Molecular Microbiology & Immunology, Saint Louis University School of Medicine; Confluence Life Sciences, Inc., St. Louis, Missouri; Hematology Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston; Harvard Stem Cell Institute and Broad Institute of Harvard and MIT, Cambridge, Massachusetts; and Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TexasAuthors' Affiliations: Departments of Cell Biology and Physiology, Medicine, and Pathology and Immunology; BRIGHT Institute, Washington University School of Medicine; Departments of Biochemistry & Molecular Biology and Molecular Microbiology & Immunology, Saint Louis University School of Medicine; Confluence Life Sciences, Inc., St. Louis, Missouri; Hematology Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston; Harvard Stem Cell Institute and Broad Institute of Harvard and MIT, Cambridge, Massachusetts; and Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas sheila.stewart@wust
| |
Collapse
|
25
|
Arrigo AP, Gibert B. HspB1, HspB5 and HspB4 in Human Cancers: Potent Oncogenic Role of Some of Their Client Proteins. Cancers (Basel) 2014; 6:333-65. [PMID: 24514166 PMCID: PMC3980596 DOI: 10.3390/cancers6010333] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Revised: 01/03/2014] [Accepted: 01/17/2014] [Indexed: 12/20/2022] Open
Abstract
Human small heat shock proteins are molecular chaperones that regulate fundamental cellular processes in normal unstressed cells as well as in many cancer cells where they are over-expressed. These proteins are characterized by cell physiology dependent changes in their oligomerization and phosphorylation status. These structural changes allow them to interact with many different client proteins that subsequently display modified activity and/or half-life. Nowdays, the protein interactomes of small Hsps are under intense investigations and will represent, when completed, key parameters to elaborate therapeutic strategies aimed at modulating the functions of these chaperones. Here, we have analyzed the potential pro-cancerous roles of several client proteins that have been described so far to interact with HspB1 (Hsp27) and its close members HspB5 (αB-crystallin) and HspB4 (αA-crystallin).
Collapse
Affiliation(s)
- André-Patrick Arrigo
- Apoptosis, Cancer and Development Laboratory, Lyon Cancer Research Center, INSERM U1052-CNRS UMR5286, Claude Bernard University Lyon 1, Lyon 69008, France.
| | - Benjamin Gibert
- Apoptosis, Cancer and Development Laboratory, Lyon Cancer Research Center, INSERM U1052-CNRS UMR5286, Claude Bernard University Lyon 1, Lyon 69008, France.
| |
Collapse
|
26
|
Seibel BA, Häfker NS, Trübenbach K, Zhang J, Tessier SN, Pörtner HO, Rosa R, Storey KB. Metabolic suppression during protracted exposure to hypoxia in the jumbo squid, Dosidicus gigas, living in an oxygen minimum zone. J Exp Biol 2014; 217:2555-68. [DOI: 10.1242/jeb.100487] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Abstract
The jumbo squid, Dosidicus gigas, can survive extended forays into the oxygen minimum zone (OMZ) of the Eastern Pacific Ocean. Previous studies have demonstrated reduced oxygen consumption and a limited anaerobic contribution to ATP production, suggesting the capacity for substantial metabolic suppression during hypoxic exposure. Here we provide a more complete description of energy metabolism and explore the expression of proteins indicative of transcriptional and translational arrest that may contribute to metabolic suppression. We demonstrate a suppression of total ATP demand under hypoxic conditions (1% oxygen, PO2 = 0.8 kPa) in both juveniles (52%) and adults (35%) of the jumbo squid. Oxygen consumption rates are reduced to 20% under hypoxia relative to air-saturated controls. Concentrations of Arg-P and ATP declined initially, reaching a new steady state (~30% of controls) after the first hour of hypoxic exposure. Octopine began accumulating after the first hour of hypoxic exposure, once Arg-P breakdown resulted in sufficient free arginine for substrate. Octopine reached levels near 30 mmoles g-1 after 3.4 h of hypoxic exposure. Succinate did increase through hypoxia but contributed minimally to total ATP production. Glycogenolysis in mantle muscle presumably serves to maintain muscle functionality and balance energetics during hypoxia. We provide evidence that post-translational modifications on histone proteins and translation factors serve as a primary means of energy conservation and that select components of the stress response are altered in hypoxic squids. Reduced ATP consumption under hypoxia serves to maintain ATP levels, prolong fuel store use and minimize the accumulation of acidic intermediates of anaerobic ATP generating pathways during prolonged diel forays into the OMZ. Metabolic suppression likely limits active, daytime foraging at depth in the core of the OMZ, but confers an energetic advantage over competitors that must remain in warm, oxygenated surface waters. Moreover, capacity for metabolic suppression provides habitat flexibility as oxygen minimum zones expand due to climate change.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Rui Rosa
- Universidade de Lisboa, Portugal
| | | |
Collapse
|
27
|
Sun X, Zhou Z, Fink DJ, Mata M. HspB1 silences translation of PDZ-RhoGEF by enhancing miR-20a and miR-128 expression to promote neurite extension. Mol Cell Neurosci 2013; 57:111-9. [PMID: 24141048 DOI: 10.1016/j.mcn.2013.10.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Revised: 10/03/2013] [Accepted: 10/08/2013] [Indexed: 11/17/2022] Open
Abstract
HspB1 is a small heat shock protein implicated in neuronal survival and neurite growth; mutations in HspB1 have been identified in hereditary motor neuronopathies and Charcot Marie Tooth Type 2 neuropathies. In cortical neurons we found that expression of HspB1 decreased RhoA activity and RhoA-GTP protein, and reversed the inhibition of neurite extension induced by NogoA. HspB1 decreased PDZ-RhoGEF, a RhoA specific guanine nucleotide exchange factor, while other regulators of RhoA activity were unchanged. The decrease in PDZ-RhoGEF was independent of proteasomal or lysosomal degradation pathways and was not associated with changes in PDZ-RhoGEF mRNA. We sequenced the 3'UTR of rat PDZ-RhoGEF and found binding sites for miRNAs miR-20a, miR-128 and miR-132. Expression of these microRNAs was substantially increased in cortical neurons transfected with HspB1. Co-transfection of HspB1 with specific inhibitors of miR-20a or miR-128 prevented the decrease in PDZ-RhoGEF and blocked the neurite growth promoting effects of HspB1. Using the 3'UTR of PDZ-RhoGEF mRNA in a luciferase reporter construct we observed that HspB1, miR-20a and miR-128 each inhibited luciferase expression. We conclude that HspB1 regulates RhoA activity through modulation of PDZ-RhoGEF levels achieved by translational control through enhanced expression of specific miRNAs (miR-20a and miR-128). Regulation of RhoA activity by translational silencing of PDZ-RhoGEF may be the mechanism through which HspB1 is involved in regulation of neurite growth. As RhoA-GTPase plays a regulatory role in the organization and stability of cytoskeletal networks through its downstream effectors, the results suggest a possible mechanism linking HspB1 mutations and axonal cytoskeletal pathology.
Collapse
Affiliation(s)
- Xiankui Sun
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA; GRECC VA Ann Arbor Healthcare System, Ann Arbor, MI 48109, USA
| | | | | | | |
Collapse
|
28
|
Krause CD, Izotova LS, Pestka S. Analytical use of multi-protein Fluorescence Resonance Energy Transfer to demonstrate membrane-facilitated interactions within cytokine receptor complexes. Cytokine 2013; 64:298-309. [PMID: 23769803 PMCID: PMC3770794 DOI: 10.1016/j.cyto.2013.05.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Revised: 05/17/2013] [Accepted: 05/18/2013] [Indexed: 12/17/2022]
Abstract
Experiments measuring Fluorescence Resonance Energy Transfer (FRET) between cytokine receptor chains and their associated proteins led to hypotheses describing their organization in intact cells. These interactions occur within a larger protein complex or within a given nano-environment. To illustrate this complexity empirically, we developed a protocol to analyze FRET among more than two fluorescent proteins (multi-FRET). In multi-FRET, we model FRET among more than two fluorophores as the sum of all possible pairwise interactions within the complex. We validated our assumption by demonstrating that FRET among pairs within a fluorescent triplet resembled FRET between each pair measured in the absence of the third fluorophore. FRET between two receptor chains increases with increasing FRET between the ligand-binding chain (e.g., IFN-γR1, IL-10R1 and IFN-λR1) and an acylated fluorescent protein that preferentially resides within subsections of the plasma membrane. The interaction of IL-10R2 with IFN-λR1 or IL-10R1 results in decreased FRET between IL-10R2 and the acylated fluorescent protein. Finally, we analyzed FRET among four fluorescent proteins to demonstrate that as FRET between IFN-γR1 and IFN-γR2 or between IFN-αR1 and IFN-αR2c increases, FRET among other pairs of proteins changes within each complex.
Collapse
Affiliation(s)
- Christopher D Krause
- Department of Biochemistry and Molecular Biology, Robert Wood Johnson Medical School - The University of Medicine and Dentistry of New Jersey, 675 Hoes Lane West, Piscataway, NJ 08855, USA.
| | | | | |
Collapse
|
29
|
Krause CD, Digioia G, Izotova LS, Pestka S. Improving the spectral analysis of Fluorescence Resonance Energy Transfer in live cells: application to interferon receptors and Janus kinases. Cytokine 2013; 64:272-85. [PMID: 23796694 PMCID: PMC3868223 DOI: 10.1016/j.cyto.2013.05.026] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Revised: 04/22/2013] [Accepted: 05/29/2013] [Indexed: 01/21/2023]
Abstract
The observed Fluorescence Resonance Energy Transfer (FRET) between fluorescently labeled proteins varies in cells. To understand how this variation affects our interpretation of how proteins interact in cells, we developed a protocol that mathematically separates donor-independent and donor-dependent excitations of acceptor, determines the electromagnetic interaction of donors and acceptors, and quantifies the efficiency of the interaction of donors and acceptors. By analyzing large populations of cells, we found that misbalanced or insufficient expression of acceptor or donor as well as their inefficient or reversible interaction influenced FRET efficiency in vivo. Use of red-shifted donors and acceptors gave spectra with less endogenous fluorescence but produced lower FRET efficiency, possibly caused by reduced quenching of red-shifted fluorophores in cells. Additionally, cryptic interactions between jellyfish FPs artefactually increased the apparent FRET efficiency. Our protocol can distinguish specific and nonspecific protein interactions even within highly constrained environments as plasma membranes. Overall, accurate FRET estimations in cells or within complex environments can be obtained by a combination of proper data analysis, study of sufficient numbers of cells, and use of properly empirically developed fluorescent proteins.
Collapse
Affiliation(s)
- Christopher D Krause
- Department of Biochemistry and Molecular Biology, Robert Wood Johnson Medical School, The University of Medicine and Dentistry of New Jersey, 675 Hoes Lane West, Piscataway, NJ 08855, USA.
| | | | | | | |
Collapse
|
30
|
Krause CD, Digioia G, Izotova LS, Xie J, Kim Y, Schwartz BJ, Mirochnitchenko OV, Pestka S. Ligand-independent interaction of the type I interferon receptor complex is necessary to observe its biological activity. Cytokine 2013; 64:286-97. [PMID: 23830819 PMCID: PMC3770802 DOI: 10.1016/j.cyto.2013.06.309] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Revised: 04/23/2013] [Accepted: 06/10/2013] [Indexed: 10/26/2022]
Abstract
Ectopic coexpression of the two chains of the Type I and Type III interferon (IFN) receptor complexes (IFN-αR1 and IFN-αR2c, or IFN-λR1 and IL-10R2) yielded sensitivity to IFN-alpha or IFN-lambda in only some cells. We found that IFN-αR1 and IFN-αR2c exhibit FRET only when expressed at equivalent and low levels. Expanded clonal cell lines expressing both IFN-αR1 and IFN-αR2c were sensitive to IFN-alpha only when IFN-αR1 and IFN-αR2c exhibited FRET in the absence of human IFN-alpha. Coexpression of RACK-1 or Jak1 enhanced the affinity of the interaction between IFN-αR1 and IFN-αR2c. Both IFN-αR1 and IFN-αR2c exhibited FRET with Jak1 and Tyk2. Together with data showing that disruption of the preassociation between the IFN-gamma receptor chains inhibited its biological activity, we propose that biologically active IFN receptors require ligand-independent juxtaposition of IFN receptor chains assisted by their associated cytosolic proteins.
Collapse
Affiliation(s)
- Christopher D. Krause
- Department of Biochemistry and Molecular Biology, Robert Wood Johnson Medical School - The University of Medicine and Dentistry of New Jersey, 675 Hoes Lane West, Piscataway, NJ 08855 USA
| | - Gina Digioia
- Department of Biochemistry and Molecular Biology, Robert Wood Johnson Medical School - The University of Medicine and Dentistry of New Jersey, 675 Hoes Lane West, Piscataway, NJ 08855 USA
- Pestka Biomedical Laboratories, 131 Ethel Road West, Suite 6, Piscataway, NJ 08854 USA
| | - Lara S. Izotova
- Department of Biochemistry and Molecular Biology, Robert Wood Johnson Medical School - The University of Medicine and Dentistry of New Jersey, 675 Hoes Lane West, Piscataway, NJ 08855 USA
| | - Junxia Xie
- Department of Biochemistry and Molecular Biology, Robert Wood Johnson Medical School - The University of Medicine and Dentistry of New Jersey, 675 Hoes Lane West, Piscataway, NJ 08855 USA
| | - Youngsun Kim
- Department of Biochemistry and Molecular Biology, Robert Wood Johnson Medical School - The University of Medicine and Dentistry of New Jersey, 675 Hoes Lane West, Piscataway, NJ 08855 USA
| | - Barbara J. Schwartz
- Department of Biochemistry and Molecular Biology, Robert Wood Johnson Medical School - The University of Medicine and Dentistry of New Jersey, 675 Hoes Lane West, Piscataway, NJ 08855 USA
| | - Olga V. Mirochnitchenko
- Department of Biochemistry and Molecular Biology, Robert Wood Johnson Medical School - The University of Medicine and Dentistry of New Jersey, 675 Hoes Lane West, Piscataway, NJ 08855 USA
| | - Sidney Pestka
- Department of Biochemistry and Molecular Biology, Robert Wood Johnson Medical School - The University of Medicine and Dentistry of New Jersey, 675 Hoes Lane West, Piscataway, NJ 08855 USA
- Pestka Biomedical Laboratories, 131 Ethel Road West, Suite 6, Piscataway, NJ 08854 USA
| |
Collapse
|
31
|
Abstract
Post-transcriptional mechanisms that modulate global and/or transcript-specific mRNA stability and translation contribute to the rapid and flexible control of gene expression in immune effector cells. These mechanisms rely on RNA-binding proteins (RBPs) that direct regulatory complexes (e.g. exosomes, deadenylases, decapping complexes, RNA-induced silencing complexes) to the 3'-untranslated regions of specific immune transcripts. Here, we review the surprising variety of post-transcriptional control mechanisms that contribute to gene expression in the immune system and discuss how defects in these pathways can contribute to autoimmune disease.
Collapse
Affiliation(s)
- Pavel Ivanov
- Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Boston, MA 02115, USA.
| | | |
Collapse
|
32
|
Almeida-Souza L, Asselbergh B, De Winter V, Goethals S, Timmerman V, Janssens S. HSPB1 facilitates the formation of non-centrosomal microtubules. PLoS One 2013; 8:e66541. [PMID: 23826100 PMCID: PMC3691211 DOI: 10.1371/journal.pone.0066541] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Accepted: 05/07/2013] [Indexed: 11/19/2022] Open
Abstract
The remodeling capacity of microtubules (MT) is essential for their proper function. In mammals, MTs are predominantly formed at the centrosome, but can also originate from non-centrosomal sites, a process that is still poorly understood. We here show that the small heat shock protein HSPB1 plays a role in the control of non-centrosomal MT formation. The HSPB1 expression level regulates the balance between centrosomal and non-centrosomal MTs. The HSPB1 protein can be detected specifically at sites of de novo forming non-centrosomal MTs, while it is absent from the centrosomes. In addition, we show that HSPB1 binds preferentially to the lattice of newly formed MTs in vitro, suggesting that its function occurs by stabilizing MT seeds. Our findings open new avenues for the understanding of the role of HSPB1 in the development, maintenance and protection of cells with specialized non-centrosomal MT arrays.
Collapse
Affiliation(s)
- Leonardo Almeida-Souza
- Department of Molecular Genetics, VIB and University of Antwerp, Antwerpen, Belgium
- Neurogenetics Laboratory, Institute Born Bunge, University of Antwerp, Antwerpen, Belgium
| | - Bob Asselbergh
- Department of Molecular Genetics, VIB and University of Antwerp, Antwerpen, Belgium
- Neurogenetics Laboratory, Institute Born Bunge, University of Antwerp, Antwerpen, Belgium
| | - Vicky De Winter
- Department of Molecular Genetics, VIB and University of Antwerp, Antwerpen, Belgium
- Neurogenetics Laboratory, Institute Born Bunge, University of Antwerp, Antwerpen, Belgium
| | - Sofie Goethals
- Department of Molecular Genetics, VIB and University of Antwerp, Antwerpen, Belgium
- Neurogenetics Laboratory, Institute Born Bunge, University of Antwerp, Antwerpen, Belgium
| | - Vincent Timmerman
- Department of Molecular Genetics, VIB and University of Antwerp, Antwerpen, Belgium
- Neurogenetics Laboratory, Institute Born Bunge, University of Antwerp, Antwerpen, Belgium
- * E-mail: (VT); (SJ)
| | - Sophie Janssens
- Department of Molecular Genetics, VIB and University of Antwerp, Antwerpen, Belgium
- GROUP-ID Consortium, Laboratory for Immunoregulation and Mucosal Immunology, University of Ghent, Ghent, Belgium
- Department of Molecular Biomedical Research, VIB, Ghent, Belgium
- * E-mail: (VT); (SJ)
| |
Collapse
|
33
|
Arrigo AP, Gibert B. Protein interactomes of three stress inducible small heat shock proteins: HspB1, HspB5 and HspB8. Int J Hyperthermia 2013; 29:409-22. [DOI: 10.3109/02656736.2013.792956] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
|
34
|
Arrigo AP. Human small heat shock proteins: Protein interactomes of homo- and hetero-oligomeric complexes: An update. FEBS Lett 2013; 587:1959-69. [DOI: 10.1016/j.febslet.2013.05.011] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2013] [Revised: 05/02/2013] [Accepted: 05/02/2013] [Indexed: 10/26/2022]
|
35
|
Stockert J, Wolf A, Kaddatz K, Schnitzer E, Finkernagel F, Meissner W, Müller-Brüsselbach S, Kracht M, Müller R. Regulation of TAK1/TAB1-mediated IL-1β signaling by cytoplasmic PPARβ/δ. PLoS One 2013; 8:e63011. [PMID: 23646170 PMCID: PMC3639976 DOI: 10.1371/journal.pone.0063011] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2012] [Accepted: 03/27/2013] [Indexed: 11/19/2022] Open
Abstract
The peroxisome proliferator-activated receptor subtypes PPARα, PPARβ/δ, PPARγ are members of the steroid hormone receptor superfamily with well-established functions in transcriptional regulation. Here, we describe an unexpected cytoplasmic function of PPARβ/δ. Silencing of PPARβ/δ expression interferes with the expression of a large subset of interleukin-1β (IL-1β)-induced target genes in HeLa cells, which is preceded by an inhibition of the IL-1β-induced phosphorylation of TAK1 and its downstream effectors, including the NFκBα inhibitor IκBα (NFKBIA) and the NFκBα subunit p65 (RELA). PPARβ/δ enhances the interaction between TAK1 and the small heat-shock protein HSP27, a known positive modulator of TAK1-mediated IL-1β signaling. Consistent with these findings, PPARβ/δ physically interacts with both the endogenous cytoplasmic TAK1/TAB1 complex and HSP27, and PPARβ/δ overexpression increases the TAK1-induced transcriptional activity of NFκB. These observations suggest that PPARβ/δ plays a role in the assembly of a cytoplasmic multi-protein complex containing TAK1, TAB1, HSP27 and PPARβ/δ, and thereby participates in the NFκB response to IL-1β.
Collapse
Affiliation(s)
- Josefine Stockert
- Institute of Molecular Biology and Tumor Research (IMT), Philipps University, Marburg, Germany
| | - Alexander Wolf
- Rudolf Buchheim Institute for Pharmacology, Giessen, Germany
| | - Kerstin Kaddatz
- Institute of Molecular Biology and Tumor Research (IMT), Philipps University, Marburg, Germany
| | - Evelyn Schnitzer
- Institute of Molecular Biology and Tumor Research (IMT), Philipps University, Marburg, Germany
| | - Florian Finkernagel
- Institute of Molecular Biology and Tumor Research (IMT), Philipps University, Marburg, Germany
| | - Wolfgang Meissner
- Institute of Molecular Biology and Tumor Research (IMT), Philipps University, Marburg, Germany
| | | | - Michael Kracht
- Rudolf Buchheim Institute for Pharmacology, Giessen, Germany
| | - Rolf Müller
- Institute of Molecular Biology and Tumor Research (IMT), Philipps University, Marburg, Germany
- * E-mail:
| |
Collapse
|
36
|
Amirouche A, Tadesse H, Lunde JA, Bélanger G, Côté J, Jasmin BJ. Activation of p38 signaling increases utrophin A expression in skeletal muscle via the RNA-binding protein KSRP and inhibition of AU-rich element-mediated mRNA decay: implications for novel DMD therapeutics. Hum Mol Genet 2013; 22:3093-111. [PMID: 23575223 DOI: 10.1093/hmg/ddt165] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Several therapeutic approaches are currently being developed for Duchenne muscular dystrophy (DMD) including upregulating the levels of endogenous utrophin A in dystrophic fibers. Here, we examined the role of post-transcriptional mechanisms in controlling utrophin A expression in skeletal muscle. We show that activation of p38 leads to an increase in utrophin A independently of a transcriptional induction. Rather, p38 controls the levels of utrophin A mRNA by extending the half-life of transcripts via AU-rich elements (AREs). This mechanism critically depends on a decrease in the functional availability of KSRP, an RNA-binding protein known to promote decay of ARE-containing transcripts. In vitro and in vivo binding studies revealed that KSRP interacts with specific AREs located within the utrophin A 3' UTR. Electroporation experiments to knockdown KSRP led to an increase in utrophin A in wild-type and mdx mouse muscles. In pre-clinical studies, treatment of mdx mice with heparin, an activator of p38, causes a pronounced increase in utrophin A in diaphragm muscle fibers. Together, these studies identify a pathway that culminates in the post-transcriptional regulation of utrophin A through increases in mRNA stability. Furthermore, our results constitute proof-of-principle showing that pharmacological activation of p38 may prove beneficial as a novel therapeutic approach for DMD.
Collapse
Affiliation(s)
- Adel Amirouche
- Faculty of Medicine, Department of Cellular and Molecular Medicine and Centre for Neuromuscular Disease, University of Ottawa, ON, Canada K1H 8M5
| | | | | | | | | | | |
Collapse
|
37
|
Wong J, Si X, Angeles A, Zhang J, Shi J, Fung G, Jagdeo J, Wang T, Zhong Z, Jan E, Luo H. Cytoplasmic redistribution and cleavage of AUF1 during coxsackievirus infection enhance the stability of its viral genome. FASEB J 2013; 27:2777-87. [PMID: 23572232 DOI: 10.1096/fj.12-226498] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Coxsackievirus B3 (CVB3) is a causative agent of viral myocarditis, hepatitis, pancreatitis, and meningitis in humans. The adenosine-uridine (AU)-rich element RNA binding factor 1 (AUF1) is an integral component in the regulation of gene expression. AUF1 destabilizes mRNAs and targets them for degradation by binding to AU-rich elements in the 3' untranslated region (UTR) of mRNAs. The 3'-UTR of the CVB3 genome contains canonical AU-rich sequences, raising the possibility that CVB3 RNA may also be subjected to AUF1-mediated degradation. Here, we reported that CVB3 infection led to cytoplasmic redistribution and cleavage of AUF1. These events are independent of CVB3-induced caspase activation but require viral protein production. Overexpression of viral protease 2A reproduced CVB3-induced cytoplasmic redistribution of AUF1, while in vitro cleavage assay revealed that viral protease 3C contributed to AUF1 cleavage. Furthermore, we showed that knockdown of AUF1 facilitated viral RNA, protein, and progeny production, suggesting an antiviral property for AUF1 against CVB3 infection. Finally, an immunoprecipitation study demonstrated the physical interaction between AUF1 and the 3'-UTR of CVB3, potentially targeting CVB3 genome toward degradation. Together, our results suggest that cleavage of AUF1 may be a strategy employed by CVB3 to enhance the stability of its viral genome.
Collapse
Affiliation(s)
- Jerry Wong
- James Hogg Research Center, Providence Heart and Lung Institute, St. Paul's Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Hsp27 and F-box protein β-TrCP promote degradation of mRNA decay factor AUF1. Mol Cell Biol 2013; 33:2315-26. [PMID: 23530064 DOI: 10.1128/mcb.00931-12] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Activation of the mitogen-activated protein (MAP) pathway kinases p38 and MK2 induces phosphorylation of the chaperone Hsp27 and stabilization of mRNAs containing AU-rich elements (AREs) (ARE-mRNAs). Likewise, expression of phosphomimetic mutant forms of Hsp27 also stabilizes ARE-mRNAs. It appears to perform this function by promoting degradation of the ARE-mRNA decay factor AUF1 by proteasomes. In this study, we examined the molecular mechanism linking Hsp27 phosphorylation to AUF1 degradation by proteasomes. AUF1 is a target of β-TrCP, the substrate recognition subunit of the E3 ubiquitin ligase Skp1-cullin-F-box protein complex, SCF(β-TrCP). Depletion of β-TrCP stabilized AUF1. In contrast, overexpression of β-TrCP enhanced ubiquitination and degradation of AUF1 and led to stabilization of reporter mRNAs containing cytokine AREs. Enhanced AUF1 degradation required expression of phosphomimetic mutant forms of both Hsp27 and AUF1. Our results suggest that a signaling axis composed of p38 MAP kinase-MK2-Hsp27-β-TrCP may promote AUF1 degradation by proteasomes and stabilization of cytokine ARE-mRNAs.
Collapse
|
39
|
Damgaard CK, Lykke-Andersen J. Regulation of ARE-mRNA Stability by Cellular Signaling: Implications for Human Cancer. Cancer Treat Res 2013; 158:153-80. [PMID: 24222358 DOI: 10.1007/978-3-642-31659-3_7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
During recent years, it has become clear that regulation of mRNA stability is an important event in the control of gene expression. The stability of a large class of mammalian mRNAs is regulated by AU-rich elements (AREs) located in the mRNA 3' UTRs. mRNAs with AREs are inherently labile but as a response to different cellular cues they can become either stabilized, allowing expression of a given gene, or further destabilized to silence their expression. These tightly regulated mRNAs include many that encode growth factors, proto-oncogenes, cytokines, and cell cycle regulators. Failure to properly regulate their stability can therefore lead to uncontrolled expression of factors associated with cell proliferation and has been implicated in several human cancers. A number of transfactors that recognize AREs and regulate the translation and degradation of ARE-mRNAs have been identified. These transfactors are regulated by signal transduction pathways, which are often misregulated in cancers. This chapter focuses on the function of ARE-binding proteins with an emphasis on their regulation by signaling pathways and the implications for human cancer.
Collapse
|
40
|
Post-transcriptional control of gene expression by AUF1: mechanisms, physiological targets, and regulation. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2012; 1829:680-8. [PMID: 23246978 DOI: 10.1016/j.bbagrm.2012.12.002] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2012] [Revised: 11/30/2012] [Accepted: 12/06/2012] [Indexed: 12/30/2022]
Abstract
AUF1 is a family of four proteins generated by alternative pre-mRNA splicing that form high affinity complexes with AU-rich, mRNA-destabilizing sequences located within the 3' untranslated regions of many labile mRNAs. While AUF1 binding is most frequently associated with accelerated mRNA decay, emerging examples have demonstrated roles as a mRNA stabilizer or even translational regulator for specific transcripts. In this review, we summarize recent advances in our understanding of mRNA recognition by AUF1 and the biochemical and functional consequences of these interactions. In addition, unique properties of individual AUF1 isoforms and the roles of these proteins in modulating expression of genes associated with inflammatory, neoplastic, and cardiac diseases are discussed. Finally, we describe mechanisms that regulate AUF1 expression in cells, and current knowledge of regulatory switches that modulate the cellular levels and/or activities of AUF1 isoforms through distinct protein post-translational modifications. This article is part of a Special Issue entitled: RNA Decay mechanisms.
Collapse
|
41
|
Barrett LW, Fletcher S, Wilton SD. Regulation of eukaryotic gene expression by the untranslated gene regions and other non-coding elements. Cell Mol Life Sci 2012; 69:3613-34. [PMID: 22538991 PMCID: PMC3474909 DOI: 10.1007/s00018-012-0990-9] [Citation(s) in RCA: 390] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2011] [Revised: 02/28/2012] [Accepted: 04/02/2012] [Indexed: 02/07/2023]
Abstract
There is now compelling evidence that the complexity of higher organisms correlates with the relative amount of non-coding RNA rather than the number of protein-coding genes. Previously dismissed as "junk DNA", it is the non-coding regions of the genome that are responsible for regulation, facilitating complex temporal and spatial gene expression through the combinatorial effect of numerous mechanisms and interactions working together to fine-tune gene expression. The major regions involved in regulation of a particular gene are the 5' and 3' untranslated regions and introns. In addition, pervasive transcription of complex genomes produces a variety of non-coding transcripts that interact with these regions and contribute to regulation. This review discusses recent insights into the regulatory roles of the untranslated gene regions and non-coding RNAs in the control of complex gene expression, as well as the implications of this in terms of organism complexity and evolution.
Collapse
Affiliation(s)
- Lucy W Barrett
- Centre for Neuromuscular and Neurological Disorders (CNND), The University of Western Australia (M518), 35 Stirling Highway, Crawley, WA 6009, Australia.
| | | | | |
Collapse
|
42
|
The mRNA-bound proteome and its global occupancy profile on protein-coding transcripts. Mol Cell 2012; 46:674-90. [PMID: 22681889 DOI: 10.1016/j.molcel.2012.05.021] [Citation(s) in RCA: 928] [Impact Index Per Article: 71.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2012] [Revised: 05/14/2012] [Accepted: 05/17/2012] [Indexed: 01/17/2023]
Abstract
Protein-RNA interactions are fundamental to core biological processes, such as mRNA splicing, localization, degradation, and translation. We developed a photoreactive nucleotide-enhanced UV crosslinking and oligo(dT) purification approach to identify the mRNA-bound proteome using quantitative proteomics and to display the protein occupancy on mRNA transcripts by next-generation sequencing. Application to a human embryonic kidney cell line identified close to 800 proteins. To our knowledge, nearly one-third were not previously annotated as RNA binding, and about 15% were not predictable by computational methods to interact with RNA. Protein occupancy profiling provides a transcriptome-wide catalog of potential cis-regulatory regions on mammalian mRNAs and showed that large stretches in 3' UTRs can be contacted by the mRNA-bound proteome, with numerous putative binding sites in regions harboring disease-associated nucleotide polymorphisms. Our observations indicate the presence of a large number of mRNA binders with diverse molecular functions participating in combinatorial posttranscriptional gene-expression networks.
Collapse
|
43
|
Ciocca DR, Arrigo AP, Calderwood SK. Heat shock proteins and heat shock factor 1 in carcinogenesis and tumor development: an update. Arch Toxicol 2012; 87:19-48. [PMID: 22885793 DOI: 10.1007/s00204-012-0918-z] [Citation(s) in RCA: 194] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2012] [Accepted: 07/25/2012] [Indexed: 12/20/2022]
Abstract
Heat shock proteins (HSP) are a subset of the molecular chaperones, best known for their rapid and abundant induction by stress. HSP genes are activated at the transcriptional level by heat shock transcription factor 1 (HSF1). During the progression of many types of cancer, this heat shock transcriptional regulon becomes co-opted by mechanisms that are currently unclear, although evidently triggered in the emerging tumor cell. Concerted activation of HSF1 and the accumulation of HSPs then participate in many of the traits that permit the malignant phenotype. Thus, cancers of many histologies exhibit activated HSF1 and increased HSP levels that may help to deter tumor suppression and evade therapy in the clinic. We review here the extensive work that has been carried out and is still in progress aimed at (1) understanding the oncogenic mechanisms by which HSP genes are switched on, (2) determining the roles of HSF1/HSP in malignant transformation and (3) discovering approaches to therapy based on disrupting the influence of the HSF1-controlled transcriptome in cancer.
Collapse
Affiliation(s)
- Daniel R Ciocca
- Oncology Laboratory, Institute of Experimental Medicine and Biology of Cuyo (IMBECU), Scientific and Technological Center (CCT), CONICET, 5500 Mendoza, Argentina.
| | - Andre Patrick Arrigo
- Apoptosis Cancer and Development, Cancer Research Center of Lyon (CRCL), UMR INSERM 1052-CNRS 5286, Claude Bernard University, Lyon-1, Cheney A Building, Centre Regional Léon Bérard, 28, rue Laennec 69008 LYON, France. ;
| | - Stuart K Calderwood
- Molecular and Cellular Radiation Oncology, Department of Radiation Oncology, Beth Israel Deaconess Medical Center, Boston, MA02215
| |
Collapse
|
44
|
Roggli E, Gattesco S, Pautz A, Regazzi R. Involvement of the RNA-binding protein ARE/poly(U)-binding factor 1 (AUF1) in the cytotoxic effects of proinflammatory cytokines on pancreatic beta cells. Diabetologia 2012; 55:1699-708. [PMID: 22159912 DOI: 10.1007/s00125-011-2399-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2011] [Accepted: 11/07/2011] [Indexed: 01/01/2023]
Abstract
AIMS/HYPOTHESIS Chronic exposure of pancreatic beta cells to proinflammatory cytokines leads to impaired insulin secretion and apoptosis. ARE/poly(U)-binding factor 1 (AUF1) belongs to a protein family that controls mRNA stability and translation by associating with adenosine- and uridine-rich regions of target messengers. We investigated the involvement of AUF1 in cytokine-induced beta cell dysfunction. METHODS Production and subcellular distribution of AUF1 isoforms were analysed by western blotting. To test for their role in the control of beta cell functions, each isoform was overproduced individually in insulin-secreting cells. The contribution to cytokine-mediated beta cell dysfunction was evaluated by preventing the production of AUF1 isoforms by RNA interference. The effect of AUF1 on the production of potential targets was assessed by western blotting. RESULTS MIN6 cells and human pancreatic islets were found to produce four AUF1 isoforms (p42>p45>p37>p40). AUF1 isoforms were mainly localised in the nucleus but were partially translocated to the cytoplasm upon exposure of beta cells to cytokines and activation of the ERK pathway. Overproduction of AUF1 did not affect glucose-induced insulin secretion but promoted apoptosis. This effect was associated with a decrease in the production of the anti-apoptotic proteins, B cell leukaemia/lymphoma 2 (BCL2) and myeloid cell leukaemia sequence 1 (MCL1). Silencing of AUF1 isoforms restored the levels of the anti-apoptotic proteins, attenuated the activation of the nuclear factor-κB (NFκB) pathway, and protected the beta cells from cytokine-induced apoptosis. CONCLUSIONS/INTERPRETATION Our findings point to a contribution of AUF1 to the deleterious effects of cytokines on beta cell functions and suggest a role for this RNA-binding protein in the early phases of type 1 diabetes.
Collapse
Affiliation(s)
- E Roggli
- Department of Cell Biology and Morphology, Faculty of Biology and Medicine, University of Lausanne, Rue du Bugnon 9, CH-1005 Lausanne, Switzerland
| | | | | | | |
Collapse
|
45
|
Ito M, Imai M, Muraki M, Miyado K, Qin J, Kyuwa S, Yoshikawa Y, Hosoi Y, Saito H, Takahashi Y. GSTT1 is upregulated by oxidative stress through p38-MK2 signaling pathway in human granulosa cells: possible association with mitochondrial activity. Aging (Albany NY) 2012; 3:1213-23. [PMID: 22207314 PMCID: PMC3273902 DOI: 10.18632/aging.100418] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
We previously reported that GSTT1 was upregulated in human granulosa cells during aging and that activation and localization of p38 MAPK was changed in parallel. Although oxidative stress is responsible for these changes, the age-associated expression of GSTT1 regulated by MAPKs and the role of GSTT1 in aged granulosa cells remain unclear. Therefore, we examined the relationship between the expression of GSTT1 and MAPK signaling pathways using human granulosa-like KGN cells stimulated with H2O2 in the presence or absence of various MAPK inhibitors. Interestingly, H2O2-induced GSTT1 was only inhibited by a p38 inhibitor. An inhibitor of MK2, a downstream regulator of p38, also diminished H2O2-induced GSTT1 upregulation. Notably, both p38 and MK2 were significantly inactivated in cells carrying an shRNA construct of GSTT1 (ΔGSTT1 cells), suggesting that the p38-MK2 pathway is essential for age-associated upregulation of GSTT1. The relevance of GSTT1 in mitochondrial activity was then determined. ΔGSTT1 cells displayed enhanced polarization of mitochondrial membrane potential without increasing the apoptosis, suggesting that the age-associated upregulation of GSTT1 may influence the mitochondrial activity of granulosa cells. Collectively, it appears that the age-associated expression of GSTT1 is induced through the p38 signaling pathway and GSTT1 influences homeostatic activities in granulosa cells.
Collapse
Affiliation(s)
- Megumu Ito
- Division of Reproductive Medicine, Department of Perinatal Medicine and Maternal Care, National Center for Child Health and Development, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Wu X, Brewer G. The regulation of mRNA stability in mammalian cells: 2.0. Gene 2012; 500:10-21. [PMID: 22452843 DOI: 10.1016/j.gene.2012.03.021] [Citation(s) in RCA: 182] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2012] [Revised: 02/20/2012] [Accepted: 03/04/2012] [Indexed: 12/24/2022]
Abstract
Messenger RNA decay is an essential step in gene expression to set mRNA abundance in the cytoplasm. The binding of proteins and/or noncoding RNAs to specific recognition sequences or secondary structures within mRNAs dictates mRNA decay rates by recruiting specific enzyme complexes that perform the destruction processes. Often, the cell coordinates the degradation or stabilization of functional subsets of mRNAs encoding proteins collectively required for a biological process. As well, extrinsic or intrinsic stimuli activate signal transduction pathways that modify the mRNA decay machinery with consequent effects on decay rates and mRNA abundance. This review is an update to our 2001 Gene review on mRNA stability in mammalian cells, and we survey the enormous progress made over the past decade.
Collapse
Affiliation(s)
- Xiangyue Wu
- Department of Molecular Genetics, UMDNJ-Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA
| | | |
Collapse
|
47
|
Bode JG, Ehlting C, Häussinger D. The macrophage response towards LPS and its control through the p38(MAPK)-STAT3 axis. Cell Signal 2012; 24:1185-94. [PMID: 22330073 DOI: 10.1016/j.cellsig.2012.01.018] [Citation(s) in RCA: 310] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2012] [Accepted: 01/27/2012] [Indexed: 12/19/2022]
Abstract
In macrophages detection of gram-negative bacteria particularly involves binding of the outer-wall component lipopolysaccharide (LPS) to its cognate receptor complex, comprising Toll like receptor 4 (TLR4), CD14 and MD2. LPS-induced formation of the LPS receptor complex elicits a signaling network, including intra-cellular signal-transduction directly activated by the TLR4 receptor complex as well as successional induction of indirect autocrine and paracrine signaling events. All these different pathways are integrated into the macrophage response towards an inflammatory stimulus by a highly complex cross-talk of the pathways engaged. This also includes a tight control by several intra- and inter-cellular feedback loops warranting an inflammatory response sufficient to battle invading pathogens and to avoid non-essential tissue damage caused by an overwhelming inflammatory response. Several evidences indicate that the reciprocal cross-talk between the p38(MAPK)-pathway and signal transducer and activator of transcription (STAT)3-mediated signal-transduction forms a critical axis successively activated by LPS. The balanced activation of this axis is essential for both induction and propagation of the inflammatory macrophage response as well as for the control of the resolution phase, which is largely driven by IL-10 and sustained STAT3 activation. In this context regulation of suppressor of cytokine signaling (SOCS)3 expression and the recently described divergent regulatory roles of the two p38(MAPK)-activated protein kinases MK2 and MK3 for the regulation of LPS-induced NF-κB- and IRF3-mediated signal-transduction and gene expression, which includes the regulation of IFNβ, IL-10 and DUSP1, appears to play an important role.
Collapse
Affiliation(s)
- Johannes G Bode
- Department of Gastroenterology, Hepatology and Infectious Disease, University Hospital, Heinrich Heine University of Düsseldorf, Moorenstrasse 5, 40225 Düsseldorf, Germany.
| | | | | |
Collapse
|
48
|
Johnsen IB, Nguyen TT, Bergstrøm B, Lien E, Anthonsen MW. Toll-like receptor 3-elicited MAPK activation induces stabilization of interferon-β mRNA. Cytokine 2011; 57:337-46. [PMID: 22200507 DOI: 10.1016/j.cyto.2011.11.024] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2011] [Revised: 11/29/2011] [Accepted: 11/30/2011] [Indexed: 01/18/2023]
Abstract
Prolonged release of cytokines after activation of the innate immune system may lead to systemic infection and inflammatory diseases. Many cytokines with short half-lives contain adenine- and uridine-rich elements (AREs) in their 3'-untranslated region (UTR), which mediate mRNA destabilization. The Toll-like receptors (TLRs) TLR3 and TLR4 induce immune responses via the adaptor proteins TRIF or TRIF and MyD88, respectively, leading to IFN-β production. The 3'-UTR of IFN-β mRNA contains an ARE sequence. We demonstrate that the TLR3 ligand dsRNA and the TLR4 ligand LPS induce stabilization of IFN-β mRNA transcripts in monocyte-derived dendritic cells. In cells from TRIF(-/-) and MyD88(-/-) mice we found that dsRNA-induced stabilization of IFN-β mRNA is TRIF-dependent. MAPK-activated protein 2 (MK2) has previously been found to regulate mRNA stabilization. We show that dsRNA elicits increased MK2 activation, mediated by TRIF and p38 MAPK. Chemical inhibition of p38 and MK2, and siRNA knockdown of MK2 relieved dsRNA-triggered prolongation of IFN-β mRNA half-life. Taken together, these results suggest that TLR3 induces signaling mechanisms involving TRIF, p38 MAPK and MK2 to enhance stabilization of IFN-β mRNA contributing to enhanced IFN-β levels during pathogen infections.
Collapse
Affiliation(s)
- Ingvild Bjellmo Johnsen
- Department of Laboratory Medicine, Children's and Women's Health, Norwegian University of Science and Technology, Trondheim N-7006, Norway.
| | | | | | | | | |
Collapse
|
49
|
Nagaraja GM, Kaur P, Neumann W, Asea EE, Bausero MA, Multhoff G, Asea A. Silencing Hsp25/Hsp27 gene expression augments proteasome activity and increases CD8+ T-cell-mediated tumor killing and memory responses. Cancer Prev Res (Phila) 2011; 5:122-37. [PMID: 22185976 DOI: 10.1158/1940-6207.capr-11-0121] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Relatively high expression of Hsp27 in breast and prostate cancer is a predictor of poor clinical outcome. This study elucidates a hitherto unknown mechanism by which Hsp27 regulates proteasome function and modulates tumor-specific T-cell responses. Here, we showed that short-term silencing of Hsp25 or Hsp27 using siRNA or permanent silencing of Hsp25 using lentivirus RNA interference technology enhanced PA28α mRNA expression, PA28α protein expression, and proteasome activity; abrogated metastatic potential; induced the regression of established breast tumors by tumor-specific CD8(+) T cells; and stimulated long-lasting memory responses. The adoptive transfer of reactive CD8(+) T cells from mice bearing Hsp25-silenced tumors efficiently induced the regression of established tumors in nontreated mice which normally succumb to tumor burden. The overexpression of Hsp25 and Hsp27 resulted in the repression of normal proteasome function, induced poor antigen presentation, and resulted in increased tumor burden. Taken together, this study establishes a paradigm shift in our understanding of the role of Hsp27 in the regulation of proteasome function and tumor-specific T-cell responses and paves the way for the development of molecular targets to enhance proteasome function and concomitantly inhibit Hsp27 expression in tumors for therapeutic gain.
Collapse
Affiliation(s)
- Ganachari M Nagaraja
- Division of Investigative Pathology, Scott & White Healthcare and Texas A&M Health Science Center, College of Medicine, Temple, TX 76504, USA.
| | | | | | | | | | | | | |
Collapse
|