1
|
Makrufardi F, Peng SW, Chung KF, Chadeau-Hyam M, Lee KY, Hsiao TC, Ho KF, Rusmawatiningtyas D, Murni IK, Arguni E, Wang YH, Ho SC, Yang FM, Chuang KJ, Lin SC, Chuang HC. Extreme temperatures modulate gene expression in the airway epithelium of the lungs in mice and asthma patients. Front Med (Lausanne) 2025; 12:1531154. [PMID: 40313552 PMCID: PMC12043461 DOI: 10.3389/fmed.2025.1531154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 03/28/2025] [Indexed: 05/03/2025] Open
Abstract
Background The objective of this study was to examine the effects of extreme temperatures on the gene signature and pathways of airway epithelial cells in mice and asthma patients. Methods We investigated the effects of temperature exposure at normal (22°C), and extreme low (10°C), high (40°C) and temperature fluctuation (40°C for 2 h followed by 10°C for next 2 h) in B6.Sftpc-CreER T2 ;Ai14(RCL-tdT)-D mice and pediatric and adult patient's airway epithelial exposed to extreme temperatures. Results We observed that Mmp8, Sftpb, Cxcl15 and Cd14 were significantly upregulated in airway epithelial cells in mice model. Cma1, Kit, Fdx1, Elf1a, Cdkn2aipnl, Htatsf1, Mfsd13a, Gtf2h5, Tiam2, and Trmt10c were significantly upregulated in 40°C exposure in airway epithelial cells. Sftpc, Gpr171, Sic34a2, Cox14, Lamp3, Luc7l, Nxnl, Tmub2, Tob1, and Cd3e genes were significantly upregulated in 10°C exposure group. Pediatric asthma subjects in the extreme high temperature group consistently showed decreased Wfdc21, Cib3, and Sftpc, at the same time increased Tiam2 and Cma1 expression, while in the extreme low temperature group exhibited consistently higher expression of Sftpc and Nxnl, at the same time decreased Wfdc21, Cib3, Cma1, and Dld expression. Notably, the mice in the extreme temperature fluctuation group showed decreased Wfdc21, Cib3, Gpr171, and Cttnbp2 expression, while increased Hbb-bs expression. Adult asthma subjects in the extreme temperature fluctuation group showed consistently decreased Wfdc21, Cib3, Gpr171, and Cttnbp2 expression, while increased Tiam2 and Cma1 expression. We observed that the mild, moderate, and severe asthma subject in the extreme low temperature group showed increased Tob1, Mub2, Sic34a2, Sftpc, Nxnl, Luc71, Lamp3, Gpr171, Cox14, and Cd3e expression, while in the severe asthma subjects showed increased expression in all temperature exposure group. Conclusion Our study highlights the effects of extreme temperatures on the gene signature of the airway epithelium in both mice and asthma patients. These findings suggest that extreme temperatures modulate gene expression in the airway epithelium, potentially serving as clinical indicators or biomarkers in response to climate change.
Collapse
Affiliation(s)
- Firdian Makrufardi
- International Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Child Health, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada – Dr. Sardjito Hospital, Yogyakarta, Indonesia
| | - Syue-Wei Peng
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Kian Fan Chung
- Imperial College London, National Heart and Lung Institute, London, United Kingdom
| | - Marc Chadeau-Hyam
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, United Kingdom
- MRC Centre for Environment and Health Imperial College London, London, United Kingdom
| | - Kang-Yun Lee
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Ta-Chih Hsiao
- Graduate Institute of Environmental Engineering, National Taiwan University, Taipei, Taiwan
| | - Kin-Fai Ho
- JC School of Public Health and Primary Care, The Chinese University of Hong Kong, Hong Kong, China
| | - Desy Rusmawatiningtyas
- Department of Child Health, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada – Dr. Sardjito Hospital, Yogyakarta, Indonesia
| | - Indah Kartika Murni
- Department of Child Health, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada – Dr. Sardjito Hospital, Yogyakarta, Indonesia
| | - Eggi Arguni
- Department of Child Health, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada – Dr. Sardjito Hospital, Yogyakarta, Indonesia
| | - Yuan-Hung Wang
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Medical Research, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Shu-Chuan Ho
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Feng-Ming Yang
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Kai-Jen Chuang
- School of Public Health, College of Public Health, Taipei Medical University, Taipei, Taiwan
- Department of Public Health, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Sheng-Chieh Lin
- Department of Pediatrics, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Division of Allergy, Asthma, and Immunology, Department of Pediatrics, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Hsiao-Chi Chuang
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Imperial College London, National Heart and Lung Institute, London, United Kingdom
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
- Cell Physiology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
2
|
Liu JF, Chen PC, Ling TY, Hou CH. Hyperthermia increases HSP production in human PDMCs by stimulating ROS formation, p38 MAPK and Akt signaling, and increasing HSF1 activity. Stem Cell Res Ther 2022; 13:236. [PMID: 35659731 PMCID: PMC9166587 DOI: 10.1186/s13287-022-02885-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 02/02/2022] [Indexed: 11/29/2022] Open
Abstract
Background Human placenta-derived multipotent cells (hPDMCs) are isolated from a source uncomplicated by ethical issues and are ideal for therapeutic applications because of their capacity for multilineage differentiation and proven immunosuppressive properties. It is known that heat shock preconditioning induces the upregulation of heat shock proteins (HSPs), which enhance survival and engraftment of embryonic stem cells (ESCs) during transplantation in live animal models, although whether heat shock preconditioning has the same effects in hPDMCs is unclear. Methods The hPDMCs were isolated from placenta of healthy donors. The cells were treated with heat shock (43 °C, 15 min), followed by evaluation of cell viability. Furthermore, the HSPs expression was assessed by Western blot, qPCR. The reactive oxygen species (ROS) production and signal pathway activation were determined by flow cytometry and Western blot, respectively. The regulatory pathways involved in HSPs expression were examined by pretreatment with chemical inhibitors, and siRNAs of MAPK, Akt, and heat shock factor 1 (HSF1), followed by determination of HSPs expression. Results This study demonstrates that heat shock treatment induced ROS generation and HPSs expression in hPDMCs. Heat shock stimulation also increased p38 MAPK and Akt phosphorylation. These effects were reduced by inhibitors of ROS, p38 MAPK and Akt. Moreover, we found that heat shock treatment enhanced nuclear translocation of the HSF1 in hPDMCs, representing activation of HSF1. Pretreatment of hPDMCs with ROS scavengers, SB203580 and Akt inhibitors also reduced the translocation of HSF1 induced by heat shock. Conclusions Our data indicate that heat shock acts via ROS to activate p38 MAPK and Akt signaling, which subsequently activates HSF1, leading to HSP activation and contributing to the protective role of hPDMCs.
Collapse
Affiliation(s)
- Ju-Fang Liu
- School of Oral Hygiene, College of Oral Medicine, Taipei Medical University, Taipei, 110, Taiwan.,Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, 404, Taiwan
| | - Po-Chun Chen
- Department of Life Science, National Taiwan Normal University, Taipei, 116, Taiwan.,Translational Medicine Center, Shin-Kong Wu Ho-Su Memorial Hospital, Taipei, 111, Taiwan.,Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, 404, Taiwan
| | - Thai-Yen Ling
- Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, 106, Taiwan
| | - Chun-Han Hou
- Department of Orthopedic Surgery, National Taiwan University Hospital, No. 1, Jen-Ai Road, Taipei, 100, Taiwan.
| |
Collapse
|
3
|
Lang BJ, Guerrero ME, Prince TL, Okusha Y, Bonorino C, Calderwood SK. The functions and regulation of heat shock proteins; key orchestrators of proteostasis and the heat shock response. Arch Toxicol 2021; 95:1943-1970. [PMID: 34003342 DOI: 10.1007/s00204-021-03070-8] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 05/03/2021] [Indexed: 12/14/2022]
Abstract
Cells respond to protein-damaging (proteotoxic) stress by activation of the Heat Shock Response (HSR). The HSR provides cells with an enhanced ability to endure proteotoxic insults and plays a crucial role in determining subsequent cell death or survival. The HSR is, therefore, a critical factor that influences the toxicity of protein stress. While named for its vital role in the cellular response to heat stress, various components of the HSR system and the molecular chaperone network execute essential physiological functions as well as responses to other diverse toxic insults. The effector molecules of the HSR, the Heat Shock Factors (HSFs) and Heat Shock Proteins (HSPs), are also important regulatory targets in the progression of neurodegenerative diseases and cancers. Modulation of the HSR and/or its extended network have, therefore, become attractive treatment strategies for these diseases. Development of effective therapies will, however, require a detailed understanding of the HSR, important features of which continue to be uncovered and are yet to be completely understood. We review recently described and hallmark mechanistic principles of the HSR, the regulation and functions of HSPs, and contexts in which the HSR is activated and influences cell fate in response to various toxic conditions.
Collapse
Affiliation(s)
- Benjamin J Lang
- Department of Radiation Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Martin E Guerrero
- Laboratory of Oncology, Institute of Medicine and Experimental Biology of Cuyo (IMBECU), National Scientific and Technical Research Council (CONICET), 5500, Mendoza, Argentina
| | - Thomas L Prince
- Department of Radiation Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Yuka Okusha
- Department of Radiation Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Cristina Bonorino
- Departamento de Ciências Básicas da Saúde, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, RS, Brasil.,Department of Surgery, School of Medicine, University of California, La Jolla, San Diego, CA, 92093, USA
| | - Stuart K Calderwood
- Department of Radiation Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA.
| |
Collapse
|
4
|
Hyperthermia induced disruption of mechanical balance leads to G1 arrest and senescence in cells. Biochem J 2021; 478:179-196. [PMID: 33346336 DOI: 10.1042/bcj20200705] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 12/16/2020] [Accepted: 12/21/2020] [Indexed: 12/14/2022]
Abstract
Human body temperature limits below 40°C during heat stroke or fever. The implications of prolonged exposure to the physiologically relevant temperature (40°C) on cellular mechanobiology is poorly understood. Here, we have examined the effects of heat stress (40°C for 72 h incubation) in human lung adenocarcinoma (A549), mouse melanoma (B16F10), and non-cancerous mouse origin adipose tissue cells (L929). Hyperthermia increased the level of ROS, γ-H2AX and HSP70 and decreased mitochondrial membrane potential in the cells. Heat stress impaired cell division, caused G1 arrest, induced cellular senescence, and apoptosis in all the tested cell lines. The cells incubated at 40°C for 72 h displayed a significant decrease in the f-actin level and cellular traction as compared with cells incubated at 37°C. Also, the cells showed a larger focal adhesion area and stronger adhesion at 40°C than at 37°C. The mitotic cells at 40°C were unable to round up properly and displayed retracting actin stress fibers. Hyperthermia down-regulated HDAC6, increased the acetylation level of microtubules, and perturbed the chromosome alignment in the mitotic cells at 40°C. Overexpression of HDAC6 rescued the cells from the G1 arrest and reduced the delay in cell rounding at 40°C suggesting a crucial role of HDAC6 in hyperthermia mediated responses. This study elucidates the significant role of cellular traction, focal adhesions, and cytoskeletal networks in mitotic cell rounding and chromosomal misalignment. It also highlights the significance of HDAC6 in heat-evoked senile cellular responses.
Collapse
|
5
|
Mancilla-Galindo J, Galindo-Sevilla N. Exploring the rationale for thermotherapy in COVID-19. Int J Hyperthermia 2021; 38:202-212. [PMID: 33682604 DOI: 10.1080/02656736.2021.1883127] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Increased transmissibility of the pandemic severe acute respiratory coronavirus 2 (SARS-CoV-2) has been noted to occur at lower ambient temperatures. This is seemingly related to a better replication of most respiratory viruses, including SARS-CoV-2, at lower-than-core body temperatures (i.e., 33 °C vs 37 °C). Also, intrinsic characteristics of SARS-CoV-2 make it a heat-susceptible pathogen. Thermotherapy has successfully been used to combat viral infections in plants which could otherwise result in great economic losses; 90% of viruses causing infections in plants are positive-sense single-stranded ribonucleic acid (+ssRNA) viruses, a characteristic shared by SARS-CoV-2. Thus, it is possible to envision the use of heat-based interventions (thermotherapy or mild-temperature hyperthermia) in patients with COVID-19 for which moderate cycles (every 8-12 h) of mild-temperature hyperthermia (1-2 h) have been proposed. However, there are potential safety and mechanistic concerns which could limit the use of thermotherapy only to patients with mild-to-moderate COVID-19 to prevent disease progression rather than to treat patients who have already progressed to severe-to-critical COVID-19. Here, we review the characteristics of SARS-CoV-2 which make it a heat-susceptible virus, potential host mechanisms which could be enhanced at higher temperatures to aid viral clearance, and how thermotherapy could be investigated as a modality of treatment in patients with COVID-19 while taking into consideration potential risks.
Collapse
Affiliation(s)
- Javier Mancilla-Galindo
- Facultad de Medicina, División de Investigación, Unidad de Investigación UNAM-INC, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Norma Galindo-Sevilla
- Departamento de Infectología e Inmunología, Instituto Nacional de Perinatología, Mexico City, Mexico
| |
Collapse
|
6
|
Chauhan A, Kumar R, Singh P, Jha SK, Kuanr BK. RF hyperthermia by encapsulated Fe3O4 nanoparticles induces cancer cell death via time-dependent caspase-3 activation. Nanomedicine (Lond) 2020; 15:355-379. [DOI: 10.2217/nnm-2019-0187] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Aim: To explore the optimum temperature for cancer cell death using magnetic hyperthermia (MH), which in turn will affect the mode of cell death. Method: The focus of this study is to improve upon the existing methodology for the synthesis of chitosan encapsulated Fe3O4. MH was done at different temperatures. The cell death pathway was explored using flow cytometry and western blot. Results: Coated Fe3O4 exhibited low cytotoxicity, high stability and heating efficiency. MH at 43°C was the optimum temperature for robust cell death. Cell death pathway suggested that during the initial stages of recovery, apoptosis was the main mode of cell death. While at later stages, major apoptosis and minor necrosis were observed. Conclusion: It is important to find out the long-term effect of hyperthermia treatment on cancer cells and their consequences on surrounding healthy cells.
Collapse
Affiliation(s)
- Anjali Chauhan
- Special Centre for Nanoscience, Jawaharlal Nehru University, New Delhi, 110067, India
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Ravi Kumar
- Special Centre for Nanoscience, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Pooja Singh
- National Institute of Plant Genome Research, New Delhi, 110067, India
| | - Sushil K Jha
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Bijoy Kumar Kuanr
- Special Centre for Nanoscience, Jawaharlal Nehru University, New Delhi, 110067, India
| |
Collapse
|
7
|
Girard PM, Peynot N, Lelièvre JM. Differential correlations between changes to glutathione redox state, protein ubiquitination, and stress-inducible HSPA chaperone expression after different types of oxidative stress. Cell Stress Chaperones 2018; 23:985-1002. [PMID: 29754332 PMCID: PMC6111089 DOI: 10.1007/s12192-018-0909-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 04/04/2018] [Accepted: 05/01/2018] [Indexed: 01/03/2023] Open
Abstract
In primary bovine fibroblasts with an hspa1b/luciferase transgene, we examined the intensity of heat-shock response (HSR) following four types of oxidative stress or heat stress (HS), and its putative relationship with changes to different cell parameters, including reactive oxygen species (ROS), the redox status of the key molecules glutathione (GSH), NADP(H) NAD(H), and the post-translational protein modifications carbonylation, S-glutathionylation, and ubiquitination. We determined the sub-lethal condition generating the maximal luciferase activity and inducible HSPA protein level for treatments with hydrogen peroxide (H2O2), UVA-induced oxygen photo-activation, the superoxide-generating agent menadione (MN), and diamide (DA), an electrophilic and sulfhydryl reagent. The level of HSR induced by oxidative stress was the highest after DA and MN, followed by UVA and H2O2 treatments, and was not correlated to the level of ROS production nor to the extent of protein S-glutathionylation or carbonylation observed immediately after stress. We found a correlation following oxidative treatments between HSR and the level of GSH/GSSG immediately after stress, and the increase in protein ubiquitination during the recovery period. Conversely, HS treatment, which led to the highest HSR level, did not generate ROS nor modified or depended on GSH redox state. Furthermore, the level of protein ubiquitination was maximum immediately after HS and lower than after MN and DA treatments thereafter. In these cells, heat-induced HSR was therefore clearly different from oxidative stress-induced HSR, in which conversely early redox changes of the major cellular thiol predicted the level of HSR and polyubiquinated proteins.
Collapse
Affiliation(s)
- Pierre-Marie Girard
- Institut Curie, PSL Research University, CNRS UMR3347, INSERM U1021, 91405, Orsay, France
- Université Paris-Sud, Université Paris-Saclay, Rue Georges Clémenceau, 91405, Orsay, France
| | - Nathalie Peynot
- UMR BDR, INRA, ENVA, Université Paris Saclay, 78350, Jouy-en-Josas, France
| | - Jean-Marc Lelièvre
- UMR BDR, INRA, ENVA, Université Paris Saclay, 78350, Jouy-en-Josas, France.
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350, Jouy-en-Josas, France.
| |
Collapse
|
8
|
Shepard AM, Bharwani A, Durisko Z, Andrews PW. Reverse Engineering the Febrile System. QUARTERLY REVIEW OF BIOLOGY 2018; 91:419-57. [PMID: 29562118 DOI: 10.1086/689482] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Fever, the elevation of core body temperature by behavioral or physiological means, is one of the most salient aspects of human sickness, yet there is debate regarding its functional role. In this paper, we demonstrate that the febrile system is an evolved adaptation shaped by natural selection to coordinate the immune system to fight pathogens. First, we show that previous arguments in favor of fever being an adaptation are epistemologically inadequate, and we describe how an adaptationist strategy addresses this issue more effectively. Second, we argue that the mechanisms producing fever provide clear indications of adaptation. Third, we demonstrate that there are many beneficial immune system responses activated during fever and that these responses are not mere byproducts of heat on chemical reactions. Rather, we show that natural selection appears to have modified several immune system effects to be coordinated by fever. Fourth, we argue that there are some adaptations that coordinate the febrile system with other important fitness components, particularly growth and reproduction. Finally, we discuss evidence that the febrile system may also have evolved an antitumor function, providing suggestions for future research into this area. This research informs the debate on the functional value of fever and antipyretic use.
Collapse
|
9
|
Potla R, Tulapurkar ME, Luzina IG, Atamas SP, Singh IS, Hasday JD. Exposure to febrile-range hyperthermia potentiates Wnt signalling and epithelial-mesenchymal transition gene expression in lung epithelium. Int J Hyperthermia 2017; 34:1-10. [PMID: 28540808 DOI: 10.1080/02656736.2017.1316875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
BACKGROUND As environmental and body temperatures vary, lung epithelial cells experience temperatures significantly different from normal core temperature. Our previous studies in human lung epithelium showed that: (i) heat shock accelerates wound healing and activates profibrotic gene expression through heat shock factor-1 (HSF1); (ii) HSF1 is activated at febrile temperatures (38-41 °C) and (iii) hypothermia (32 °C) activates and hyperthermia (39.5 °C) reduces expression of a subset of miRNAs that target protein kinase-Cα (PKCα) and enhance proliferation. METHODS We analysed the effect of hypo- and hyperthermia exposure on Wnt signalling by exposing human small airway epithelial cells (SAECs) and HEK293T cells to 32, 37 or 39.5 °C for 24 h, then analysing Wnt-3a-induced epithelial-mesenchymal transition (EMT) gene expression by qRT-PCR and TOPFlash reporter plasmid activity. Effects of miRNA mimics and inhibitors and the HSF1 inhibitor, KNK437, were evaluated. RESULTS Exposure to 39.5 °C for 24 h increased subsequent Wnt-3a-induced EMT gene expression in SAECs and Wnt-3a-induced TOPFlash activity in HEK293T cells. Increased Wnt responsiveness was associated with HSF1 activation and blocked by KNK437. Overexpressing temperature-responsive miRNA mimics reduced Wnt responsiveness in 39.5 °C-exposed HEK293T cells, but inhibitors of the same miRNAs failed to restore Wnt responsiveness in 32 °C-exposed HEK293T cells. CONCLUSIONS Wnt responsiveness, including expression of genes associated with EMT, increases after exposure to febrile-range temperature through an HSF1-dependent mechanism that is independent of previously identified temperature-dependent miRNAs. This process may be relevant to febrile fibrosing lung diseases, including the fibroproliferative phase of acute respiratory distress syndrome (ARDS) and exacerbations of idiopathic pulmonary fibrosis (IPF).
Collapse
Affiliation(s)
- Ratnakar Potla
- a Department of Medicine , University of Maryland School of Medicine , Baltimore , MD , USA
| | - Mohan E Tulapurkar
- a Department of Medicine , University of Maryland School of Medicine , Baltimore , MD , USA
| | - Irina G Luzina
- a Department of Medicine , University of Maryland School of Medicine , Baltimore , MD , USA.,b Medicine and Research Services, Baltimore Veterans Affairs Medical Care System , Baltimore , MD , USA
| | - Sergei P Atamas
- a Department of Medicine , University of Maryland School of Medicine , Baltimore , MD , USA.,b Medicine and Research Services, Baltimore Veterans Affairs Medical Care System , Baltimore , MD , USA
| | - Ishwar S Singh
- a Department of Medicine , University of Maryland School of Medicine , Baltimore , MD , USA
| | - Jeffrey D Hasday
- a Department of Medicine , University of Maryland School of Medicine , Baltimore , MD , USA.,b Medicine and Research Services, Baltimore Veterans Affairs Medical Care System , Baltimore , MD , USA
| |
Collapse
|
10
|
LeGrand EK, Day JD. Self-harm to preferentially harm the pathogens within: non-specific stressors in innate immunity. Proc Biol Sci 2016; 283:rspb.2016.0266. [PMID: 27075254 PMCID: PMC4843660 DOI: 10.1098/rspb.2016.0266] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 03/22/2016] [Indexed: 12/12/2022] Open
Abstract
Therapies with increasing specificity against pathogens follow the immune system's evolutionary course in maximizing host defence while minimizing self-harm. Nevertheless, even completely non-specific stressors, such as reactive molecular species, heat, nutrient and oxygen deprivation, and acidity can be used to preferentially harm pathogens. Strategic use of non-specific stressors requires exploiting differences in stress vulnerability between pathogens and hosts. Two basic vulnerabilities of pathogens are: (i) the inherent vulnerability to stress of growth and replication (more immediately crucial for pathogens than for host cells) and (ii) the degree of pathogen localization, permitting the host's use of locally and regionally intense stress. Each of the various types of non-specific stressors is present during severe infections at all levels of localization: (i) ultra-locally within phagolysosomes, (ii) locally at the infected site, (iii) regionally around the infected site and (iv) systemically as part of the acute-phase response. We propose that hosts strategically use a coordinated system of non-specific stressors at local, regional and systemic levels to preferentially harm the pathogens within. With the rising concern over emergence of resistance to specific therapies, we suggest more scrutiny of strategies using less specific therapies in pathogen control. Hosts' active use of multiple non-specific stressors is likely an evolutionarily basic defence whose retention underlies and supplements the well-recognized immune defences that directly target pathogens.
Collapse
Affiliation(s)
- Edmund K LeGrand
- Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN, USA
| | - Judy D Day
- Department of Mathematics and National Institute for Mathematical and Biological Synthesis, University of Tennessee, Knoxville, TN, USA
| |
Collapse
|
11
|
Scheraga RG, Thompson C, Tulapurkar ME, Nagarsekar AC, Cowan M, Potla R, Sun J, Cai R, Logun C, Shelhamer J, Todd NW, Singh IS, Luzina IG, Atamas SP, Hasday JD. Activation of heat shock response augments fibroblast growth factor-1 expression in wounded lung epithelium. Am J Physiol Lung Cell Mol Physiol 2016; 311:L941-L955. [PMID: 27638903 DOI: 10.1152/ajplung.00262.2016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 09/08/2016] [Indexed: 12/29/2022] Open
Abstract
We previously showed that coincident exposure to heat shock (HS; 42°C for 2 h) and TNF-α synergistically induces apoptosis in mouse lung epithelium. We extended this work by analyzing HS effects on human lung epithelial responses to clinically relevant injury. Cotreatment with TNF-α and HS induced little caspase-3 and poly(ADP-ribose) polymerase cleavage in human small airway epithelial cells, A549 cells, and BEAS2B cells. Scratch wound closure rates almost doubled when A549 and BEAS2B cells and air-liquid interface cultures of human bronchial epithelial cells were heat shocked immediately after wounding. Microarray, qRT-PCR, and immunoblotting showed fibroblast growth factor 1 (FGF1) to be synergistically induced by HS and wounding. Enhanced FGF1 expression in HS/wounded A549 was blocked by inhibitors of p38 MAPK (SB203580) or HS factor (HSF)-1 (KNK-437) and in HSF1 knockout BEAS2B cells. PCR demonstrated FGF1 to be expressed from the two most distal promoters in wounded/HS cells. Wound closure in HS A549 and BEAS2B cells was reduced by FGF receptor-1/3 inhibition (SU-5402) or FGF1 depletion. Exogenous FGF1 accelerated A549 wound closure in the absence but not presence of HS. In the presence of exogenous FGF1, HS slowed wound closure, suggesting that it increases FGF1 expression but impairs FGF1-stimulated wound closure. Frozen sections from normal and idiopathic pulmonary fibrosis (IPF) lung were analyzed for FGF1 and HSP70 by immunofluorescence confocal microscopy and qRT-PCR. FGF1 and HSP70 mRNA levels were 7.5- and 5.9-fold higher in IPF than normal lung, and the proteins colocalized to fibroblastic foci in IPF lung. We conclude that HS signaling may have an important impact on gene expression contributing to lung injury, healing, and fibrosis.
Collapse
Affiliation(s)
- Rachel G Scheraga
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland.,Critical Care Section, National Heart, Lung, Blood Institute, Bethesda, Maryland
| | | | - Mohan E Tulapurkar
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Ashish C Nagarsekar
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Mark Cowan
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland.,Medicine and Research Services, Baltimore Veterans Affairs Medical Care System, Baltimore, Maryland
| | - Ratnakar Potla
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Junfeng Sun
- Critical Care Section, National Heart, Lung, Blood Institute, Bethesda, Maryland
| | - Rongman Cai
- Critical Care Section, National Heart, Lung, Blood Institute, Bethesda, Maryland
| | - Carolea Logun
- Critical Care Section, National Heart, Lung, Blood Institute, Bethesda, Maryland
| | - James Shelhamer
- Critical Care Section, National Heart, Lung, Blood Institute, Bethesda, Maryland
| | - Nevins W Todd
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland.,Medicine and Research Services, Baltimore Veterans Affairs Medical Care System, Baltimore, Maryland
| | - Ishwar S Singh
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland.,Medicine and Research Services, Baltimore Veterans Affairs Medical Care System, Baltimore, Maryland
| | - Irina G Luzina
- Division of Rheumatology, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland.,Medicine and Research Services, Baltimore Veterans Affairs Medical Care System, Baltimore, Maryland
| | - Sergei P Atamas
- Division of Rheumatology, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland.,Medicine and Research Services, Baltimore Veterans Affairs Medical Care System, Baltimore, Maryland
| | - Jeffrey D Hasday
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland; .,Medicine and Research Services, Baltimore Veterans Affairs Medical Care System, Baltimore, Maryland
| |
Collapse
|
12
|
TANG SHU, CHEN HONGBO, CHENG YANFEN, NASIR MOHAMMADABDEL, KEMPER NICOLE, BAO ENDONG. The interactive association between heat shock factor 1 and heat shock proteins in primary myocardial cells subjected to heat stress. Int J Mol Med 2016; 37:56-62. [PMID: 26719858 PMCID: PMC4687434 DOI: 10.3892/ijmm.2015.2414] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Accepted: 10/15/2015] [Indexed: 12/21/2022] Open
Abstract
Heat shock factor 1 (HSF1) is a heat shock transcription factor that rapidly induces heat shock gene transcription following thermal stress. In this study, we subjected primary neonatal rat myocardial cells to heat stress in vitro to create a model system for investigating the trends in expression and association between various heat shock proteins (HSPs) and HSF1 under adverse environmental conditions. After the cells were subjected to heat stress at 42˚C for different periods of time, HSP and HSF1 mRNA and protein levels were detected by qPCR and western blot analysis in the heat-stressed cells. The HSF1 expression levels significantly increased in the cells following 120 min of exposure to heat stess compared to the levels observed at the beginning of heat stress exposure. HSP90 followed a similar trend in expression to HSF1, whereas HSP70 followed an opposite trend. However, no significant changes were observed in the crystallin, alpha B (CRYAB, also known as HSP beta-5) expression levels during the 480‑min period of exposure to heat stress. The interaction between the HSPs and HSF1 was analyzed by STRING 9.1, and it was found that HSF1 interacted with HSP90 and HSP70, and that it did not play a role in regulating CRYAB expression. Based on our findings, HSP70 may suppress HSF1 in rat myocardial cells under conditions of heat stress. Furthermore, our data demonstrate that HSF1 is not the key factor for all HSPs, and this was particularly the case for CRYAB.
Collapse
Affiliation(s)
- SHU TANG
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, P.R. China
| | - HONGBO CHEN
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, P.R. China
| | - YANFEN CHENG
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, P.R. China
| | - MOHAMMAD ABDEL NASIR
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, P.R. China
| | - NICOLE KEMPER
- Institute for Animal Hygiene, Animal Welfare and Farm Animal Behaviour, University of Veterinary Medicine Hannover, Foundation, D-30559 Hannover, Germany
| | - ENDONG BAO
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, P.R. China
| |
Collapse
|
13
|
Potla R, Singh IS, Atamas SP, Hasday JD. Shifts in temperature within the physiologic range modify strand-specific expression of select human microRNAs. RNA (NEW YORK, N.Y.) 2015; 21:1261-1273. [PMID: 26018549 PMCID: PMC4478345 DOI: 10.1261/rna.049122.114] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Accepted: 03/15/2015] [Indexed: 06/04/2023]
Abstract
Previous studies have revealed that clinically relevant changes in temperature modify clinically relevant gene expression profiles through transcriptional regulation. Temperature dependence of post-transcriptional regulation, specifically, through expression of miRNAs has been less studied. We comprehensively analyzed the effect of 24 h exposure to 32°C or 39.5°C on miRNA expression profile in primary cultured human small airway epithelial cells (hSAECs) and its impact on expression of a targeted protein, protein kinase C α (PKCα). Using microarray, and solution hybridization-based nCounter assays, with confirmation by quantitative RT-PCR, we found significant temperature-dependent changes in expression level of only five mature human miRNAs, representing only 1% of detected miRNAs. Four of these five miRNAs are the less abundant passenger (star) strands. They exhibited a similar pattern of increased expression at 32°C and reduced expression at 39.5°C relative to 37°C. As PKCα mRNA has multiple potential binding sites for three of these miRNAs, we analyzed PKCα protein expression in HEK 293T cells and hSAECs. PKCα protein levels were lowest at 32°C and highest at 39.5°C and specific miRNA inhibitors reduced these effects. Finally, we analyzed cell-cycle progression in hSAECs and found 32°C cells exhibited the greatest G1 to S transition, a process known to be inhibited by PKCα, and the effect was mitigated by specific miRNA inhibitors. These results demonstrate that exposure to clinically relevant hypothermia or hyperthermia modifies expression of a narrow subset of miRNAs and impacts expression of at least one signaling protein involved in multiple important cellular processes.
Collapse
Affiliation(s)
- Ratnakar Potla
- Pulmonary and Critical Care Medicine Division, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA
| | - Ishwar S Singh
- Pulmonary and Critical Care Medicine Division, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA Medicine and Research Services, Baltimore VA Medical Center, Baltimore, Maryland 21201, USA
| | - Sergei P Atamas
- Medicine and Research Services, Baltimore VA Medical Center, Baltimore, Maryland 21201, USA Division of Rheumatology and Clinical Immunology, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA
| | - Jeffrey D Hasday
- Pulmonary and Critical Care Medicine Division, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA Medicine and Research Services, Baltimore VA Medical Center, Baltimore, Maryland 21201, USA
| |
Collapse
|
14
|
Evans SS, Repasky EA, Fisher DT. Fever and the thermal regulation of immunity: the immune system feels the heat. Nat Rev Immunol 2015; 15:335-49. [PMID: 25976513 PMCID: PMC4786079 DOI: 10.1038/nri3843] [Citation(s) in RCA: 714] [Impact Index Per Article: 71.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Fever is a cardinal response to infection that has been conserved in warm-blooded and cold-blooded vertebrates for more than 600 million years of evolution. The fever response is executed by integrated physiological and neuronal circuitry and confers a survival benefit during infection. In this Review, we discuss our current understanding of how the inflammatory cues delivered by the thermal element of fever stimulate innate and adaptive immune responses. We further highlight the unexpected multiplicity of roles of the pyrogenic cytokine interleukin-6 (IL-6), both during fever induction and during the mobilization of lymphocytes to the lymphoid organs that are the staging ground for immune defence. We also discuss the emerging evidence suggesting that the adrenergic signalling pathways associated with thermogenesis shape immune cell function.
Collapse
Affiliation(s)
- Sharon S Evans
- Department of Immunology, Roswell Park Cancer Institute, Elm &Carlton Streets, Buffalo, New York 14263, USA
| | - Elizabeth A Repasky
- Department of Immunology, Roswell Park Cancer Institute, Elm &Carlton Streets, Buffalo, New York 14263, USA
| | - Daniel T Fisher
- Department of Immunology, Roswell Park Cancer Institute, Elm &Carlton Streets, Buffalo, New York 14263, USA
| |
Collapse
|
15
|
Zhang Z, Chen L, Ni H. Antipyretic therapy in critically ill patients with sepsis: an interaction with body temperature. PLoS One 2015; 10:e0121919. [PMID: 25822614 PMCID: PMC4378844 DOI: 10.1371/journal.pone.0121919] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2014] [Accepted: 02/06/2015] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND AND OBJECTIVE The effect of antipyretic therapy on mortality in patients with sepsis remains undetermined. The present study aimed to investigate the role of antipyretic therapy in ICU patients with sepsis by using a large clinical database. METHODS The multiparameter intelligent monitoring in intensive care II (MIMIC- II) database was employed for the study. Adult patients with sepsis were included for analysis. Antipyretic therapy included antipyretic medication and external cooling. Multivariable model with interaction terms were employed to explore the association of antipyretic therapy and mortality risk. MAIN RESULTS A total of 15,268 patients fulfilled inclusion criteria and were included in the study. In multivariable model by treating temperature as a continuous variable, there was significant interaction between antipyretic therapy and the maximum temperature (Tmax). While antipyretic therapy had no significant effect on mortality in low temperature quintiles, antipyretic therapy was associated with increased risk of death in the quintile with body temperature >39°C (OR: 1.29, 95% CI: 1.04-1.61). CONCLUSION Our study shows that there is no beneficial effect on reducing mortality risk with the use of antipyretic therapy in ICU patients with sepsis. External cooling may even be harmful in patients with sepsis.
Collapse
Affiliation(s)
- Zhongheng Zhang
- Department of Critical Care Medicine, Jinhua Municipal Central Hospital, Jinhua Hospital of Zhejiang University, Zhejiang, P. R. China
| | - Lin Chen
- Department of Critical Care Medicine, Jinhua Municipal Central Hospital, Jinhua Hospital of Zhejiang University, Zhejiang, P. R. China
| | - Hongying Ni
- Department of Critical Care Medicine, Jinhua Municipal Central Hospital, Jinhua Hospital of Zhejiang University, Zhejiang, P. R. China
| |
Collapse
|
16
|
Tulapurkar ME, Ramarathnam A, Hasday JD, Singh IS. Bacterial lipopolysaccharide augments febrile-range hyperthermia-induced heat shock protein 70 expression and extracellular release in human THP1 cells. PLoS One 2015; 10:e0118010. [PMID: 25659128 PMCID: PMC4320107 DOI: 10.1371/journal.pone.0118010] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2014] [Accepted: 01/06/2015] [Indexed: 01/17/2023] Open
Abstract
Sepsis, a devastating and often lethal complication of severe infection, is characterized by fever and dysregulated inflammation. While infections activate the inflammatory response in part through Toll-like receptors (TLRs), fever can partially activate the heat shock response with generation of heat shock proteins (HSPs). Since extracellular HSPs, especially HSP70 (eHSP70), are proinflammatory TLR agonists, we investigated how exposure to the TLR4 agonist, bacterial lipopolysaccharide (LPS) and febrile range hyperthermia (FRH; 39.5°C) modify HSP70 expression and extracellular release. Using differentiated THP1 cells, we found that concurrent exposure to FRH and LPS as well as TLR2 and TLR3 agonists synergized to activate expression of inducible HSP72 (HSPA1A) mRNA and protein via a p38 MAP kinase-requiring mechanism. Treatment with LPS for 6 h stimulated eHSP70 release; levels of eHSP70 released at 39.5°C were higher than at 37°C roughly paralleling the increase in intracellular HSP72 in the 39.5°C cells. By contrast, 6 h exposure to FRH in the absence of LPS failed to promote eHSP70 release. Release of eHSP70 by LPS-treated THP1 cells was inhibited by glibenclamide, but not brefeldin, indicating that eHSP70 secretion occurred via a non-classical protein secretory mechanism. Analysis of eHSP70 levels in exosomes and exosome-depleted culture supernatants from LPS-treated THP1 cells using ELISA demonstrated similar eHSP70 levels in unfractionated and exosome-depleted culture supernatants, indicating that LPS-stimulated eHSP70 release did not occur via the exosome pathway. Immunoblot analysis of the exosome fraction of culture supernatants from these cells showed constitutive HSC70 (HSPA8) to be the predominant HSP70 family member present in exosomes. In summary, we have shown that LPS stimulates macrophages to secrete inducible HSP72 via a non-classical non-exosomal pathway while synergizing with FRH exposure to increase both intracellular and secreted levels of inducible HSP72. The impact of increased macrophage intracellular HSP70 levels and augmented secretion of proinflammatory eHSP70 in the febrile, infected patient remains to be elucidated.
Collapse
Affiliation(s)
- Mohan E. Tulapurkar
- Division of Pulmonary and Critical Care, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Aparna Ramarathnam
- Division of Pulmonary and Critical Care, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Jeffrey D. Hasday
- Division of Pulmonary and Critical Care, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
- Cytokine Core Laboratory, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
- Research Services of the Baltimore Veteran Affairs Medical Center, Baltimore, Maryland, United States of America
| | - Ishwar S. Singh
- Division of Pulmonary and Critical Care, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
- Research Services of the Baltimore Veteran Affairs Medical Center, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
17
|
Abstract
The heat shock response (HSR) is an ancient and highly conserved process that is essential for coping with environmental stresses, including extremes of temperature. Fever is a more recently evolved response, during which organisms temporarily subject themselves to thermal stress in the face of infections. We review the phylogenetically conserved mechanisms that regulate fever and discuss the effects that febrile-range temperatures have on multiple biological processes involved in host defense and cell death and survival, including the HSR and its implications for patients with severe sepsis, trauma, and other acute systemic inflammatory states. Heat shock factor-1, a heat-induced transcriptional enhancer is not only the central regulator of the HSR but also regulates expression of pivotal cytokines and early response genes. Febrile-range temperatures exert additional immunomodulatory effects by activating mitogen-activated protein kinase cascades and accelerating apoptosis in some cell types. This results in accelerated pathogen clearance, but increased collateral tissue injury, thus the net effect of exposure to febrile range temperature depends in part on the site and nature of the pathologic process and the specific treatment provided.
Collapse
Affiliation(s)
- Jeffrey D Hasday
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Maryland School of Medicine and the Baltimore V.A. Medical Center, Baltimore, Maryland
| | | | | |
Collapse
|
18
|
Rohde MC, Corydon TJ, Hansen J, Pedersen CB, Schmidt SP, Gregersen N, Banner J. Heat stress and sudden infant death syndrome--stress gene expression after exposure to moderate heat stress. Forensic Sci Int 2013; 232:16-24. [PMID: 24053860 DOI: 10.1016/j.forsciint.2013.06.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2012] [Revised: 04/21/2013] [Accepted: 06/03/2013] [Indexed: 10/26/2022]
Abstract
The aim of the present study was to investigate stress gene expression in cultured primary fibroblasts established from Achilles tendons collected during autopsies from sudden infant death syndrome (SIDS) cases, and age-matched controls (infants dying in a traumatic event). Expression of 4 stress responsive genes, HSPA1B, HSPD1, HMOX1, and SOD2, was studied by quantitative reverse transcriptase PCR analysis of RNA purified from cells cultured under standard or various thermal stress conditions. The expression of all 4 genes was highly influenced by thermal stress in both SIDS and control cells. High interpersonal variance found in the SIDS group indicated that they represented a more heterogeneous group than controls. The SIDS group responded to thermal stress with a higher expression of the HSPA1B and HSPD1 genes compared to the control group, whereas no significant difference was observed in the expression of SOD2 and HMOX1 between the two groups. The differences were related to the heat shock treatment as none of the genes were expressed significantly different in SIDS at base levels at 37 °C. SOD2 and HMOX1 were up regulated in both groups, for SOD2 though the expression was lower in SIDS at all time points measured, and may be less related to heat stress. Being found dead in the prone position (a known risk factor for SIDS) was related to a lower HSPA1B up-regulation in SIDS compared to SIDS found on their side or back. The study demonstrates the potential usefulness of gene expression studies using cultured fibroblasts established from deceased individuals as a tool for molecular and pathological investigations in forensic and biomedical sciences.
Collapse
Affiliation(s)
- Marianne Cathrine Rohde
- Institute of Forensic Medicine, Department of Forensic Pathology, Aarhus University, Denmark.
| | | | | | | | | | | | | |
Collapse
|
19
|
Abstract
The heat shock response is a highly conserved primitive response that is essential for survival against a wide range of stresses, including extremes of temperature. Fever is a more recently evolved response, during which organisms raise their core body temperature and temporarily subject themselves to thermal stress in the face of infections. The present review documents studies showing the potential overlap between the febrile response and the heat shock response and how both activate the same common transcriptional programme (although with different magnitudes) including the stress-activated transcription factor, heat shock factor-1, to modify host defences in the context of infection, inflammation and injury. The review focuses primarily on how hyperthermia within the febrile range that often accompanies infections and inflammation acts as a biological response modifier and modifies innate immune responses. The characteristic 2-3 °C increase in core body temperature during fever activates and utilises elements of the heat shock response pathway to modify cytokine and chemokine gene expression, cellular signalling and immune cell mobilisation to sites of inflammation, infection and injury. Interestingly, typical proinflammatory agonists such as Toll-like receptor agonists modify the heat shock-induced transcriptional programme and expression of HSP genes following co-exposure to febrile range hyperthermia or heat shock, suggesting a complex reciprocal regulation between the inflammatory pathway and the heat shock response pathway.
Collapse
Affiliation(s)
- Ishwar S Singh
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA.
| | | |
Collapse
|
20
|
Jajarmi V, Bandehpour M, Kazemi B. Regulation of insulin biosynthesis in non-beta cells by a heat shock promoter. J Biosci Bioeng 2013; 116:147-51. [PMID: 23541501 DOI: 10.1016/j.jbiosc.2013.02.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Revised: 01/26/2013] [Accepted: 02/21/2013] [Indexed: 12/23/2022]
Abstract
Insulin production under the stringent control is the main issue in gene-based therapeutic strategies directed to type 1 diabetes. As a novel approach, inducible promoters may provide a promising tool for this purpose. In this study, we hypothesize that this control may be achieved via a promoter derived from the heat shock multigene family, Hsp70 A1A, which is inducible at 42°C. To yield mature insulin in transfected fibroblasts (3T3/NIH), a recombinant human insulin gene consisting of sequences corresponding to furin cleavable sites was fused to the promoter. Heat-stimulated cells initiated to release biologically active insulin within 30 min with a ten-fold increase after 24 h. The role of upstream regulatory elements of the promoter on its activity in heat stress conditions was examined. No significant difference between the activity of the minimal and full-length promoters was observed. This promoter exhibited low basal expression in non-inducing conditions. Results indicate that this promoter is responsive to a heat induction after approximately 30 min which causes an efficient insulin production over a relatively short period of time. These potential features of this promoter may provide an insight to control the insulin production in vivo upon an external and physical stimulation.
Collapse
Affiliation(s)
- Vahid Jajarmi
- Department of Medical Biotechnology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | | |
Collapse
|
21
|
Gupta A, Cooper ZA, Tulapurkar ME, Potla R, Maity T, Hasday JD, Singh IS. Toll-like receptor agonists and febrile range hyperthermia synergize to induce heat shock protein 70 expression and extracellular release. J Biol Chem 2013; 288:2756-66. [PMID: 23212905 PMCID: PMC3554941 DOI: 10.1074/jbc.m112.427336] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2012] [Indexed: 01/06/2023] Open
Abstract
Heat shock protein (Hsp) 70 expression can be stimulated by febrile range temperature (FRT). Hsp70 has been shown to be elevated in serum of patients with sepsis, and when released from cells, extracellular Hsp70 exerts endotoxin-like effects through Toll-like receptor 4 (TLR4) receptors. Circulating TLR agonists and fever both persist for the first several days of sepsis, and each can activate Hsp70 expression; however, the effect of combined exposure to FRT and TLR agonists on Hsp70 expression is unknown. We found that concurrent exposure to FRT (39.5 °C) and agonists for TLR4 (LPS), TLR2 (Pam3Cys), or TLR3 (poly(IC)) synergized to increase Hsp70 expression and extracellular release in RAW264.7 macrophages. The increase in Hsp70 expression was associated with activation of p38 and ERK MAP kinases, phosphorylation of histone H3, and increased recruitment of HSF1 to the Hsp70 promoter. Pretreatment with the p38 MAPK inhibitor SB283580 but not the ERK pathway inhibitor UO126 significantly reduced Hsp70 gene modification and Hsp70 expression in RAW cells co-exposed to LPS and FRT. In mice challenged with intratracheal LPS and then exposed to febrile range hyperthermia (core temperature, ∼39.5 °C), Hsp70 levels in lung tissue and in cell-free lung lavage were increased compared with mice exposed to either hyperthermia or LPS alone. We propose a model of how enhanced Hsp70 expression and extracellular release in patients concurrently exposed to fever and TLR agonists may contribute to the pathogenesis of sepsis.
Collapse
Affiliation(s)
- Aditi Gupta
- From the Division of Pulmonary and Critical Care, Department of Medicine
| | - Zachary A. Cooper
- From the Division of Pulmonary and Critical Care, Department of Medicine
| | | | - Ratnakar Potla
- From the Division of Pulmonary and Critical Care, Department of Medicine
| | - Tapan Maity
- From the Division of Pulmonary and Critical Care, Department of Medicine
| | - Jeffrey D. Hasday
- From the Division of Pulmonary and Critical Care, Department of Medicine
- the Mucosal Biology Research Center, and
- the Cytokine Core Laboratory, University of Maryland School of Medicine and
- Research Services, Baltimore Veteran Affairs Medical Center, Baltimore, Maryland 21201
| | - Ishwar S. Singh
- From the Division of Pulmonary and Critical Care, Department of Medicine
- the Mucosal Biology Research Center, and
- Research Services, Baltimore Veteran Affairs Medical Center, Baltimore, Maryland 21201
| |
Collapse
|
22
|
Estimates of exposure to cold before death from immunohistochemical expression patterns of HSP70 in glomerular podocytes. Int J Legal Med 2012; 127:783-90. [DOI: 10.1007/s00414-012-0806-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Accepted: 12/03/2012] [Indexed: 12/16/2022]
|
23
|
Wang DC, Zhang Y, Chen HY, Li XL, Qin LJ, Li YJ, Zhang HY, Wang S. Hyperthermia Promotes Apoptosis and Suppresses Invasion in C6 Rat Glioma Cells. Asian Pac J Cancer Prev 2012; 13:3239-45. [DOI: 10.7314/apjcp.2012.13.7.3239] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
24
|
LeGrand EK, Alcock J. Turning up the heat: immune brinksmanship in the acute-phase response. QUARTERLY REVIEW OF BIOLOGY 2012; 87:3-18. [PMID: 22518930 DOI: 10.1086/663946] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
The acutephase response (APR) is a systemic response to severe trauma, infection, and cancer, although many of the numerous cytokine-mediated components of the APR are incompletely understood. Some of these components, such as fever, reduced availability of iron and zinc, and nutritional restriction due to anorexia, appear to be stressors capable of causing harm to both the pathogen and the host. We review how the host benefits from differences in susceptibility to stress between pathogens and the host. Pathogens, infected host cells, and neoplastic cells are generally more stressed or vulnerable to additional stress than the host because: (a) targeted local inflammation works in synergy with APR stressors; (b) proliferation/growth increases vulnerability to stress; (c) altered pathogen physiology results in pathogen stress or vulnerability; and (d) protective heat shock responses are partially abrogated in pathogens since their responses are utilized by the host to enhance immune responses. Therefore, the host utilizes a coordinated system of endogenous stressors to provide additional levels of defense against pathogens. This model of immune brinksmanship can explain the evolutionary basis for the mutually stressful components of the APR.
Collapse
Affiliation(s)
- Edmund Kenwood LeGrand
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee Knoxville, Tennessee 37996, USA.
| | | |
Collapse
|
25
|
Moreau A, Keller DI, Huang H, Fressart V, Schmied C, Timour Q, Chahine M. Mexiletine differentially restores the trafficking defects caused by two brugada syndrome mutations. Front Pharmacol 2012; 3:62. [PMID: 22529811 PMCID: PMC3330751 DOI: 10.3389/fphar.2012.00062] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2012] [Accepted: 03/27/2012] [Indexed: 11/13/2022] Open
Abstract
The human cardiac sodium channel Na(v)1.5 encoded by the SCN5A gene plays a critical role in cardiac excitability and the propagation of action potentials. Na(v)1.5 dysfunctions due to mutations cause cardiac diseases such as the LQT3 form of long QT syndrome, conduction disorders, and Brugada syndrome (BrS). They have also recently been associated with dilated cardiomyopathy. BrS is characterized by coved ST-segment elevation on surface ECGs and lethal ventricular arrhythmias in an apparently structurally normal heart. Na(v)1.5 mutations that cause BrS result in a loss of channel function. Our aim was to functionally characterize two novel Na(v)1.5 mutations (A124D and V1378M) in BrS patients. Wild-type (WT) and mutant Na(v)1.5 channels were expressed in tsA201 cells in the presence of the β(1)-auxiliary subunit. The patch-clamp technique and immunocytochemistry approaches were used to study the mutant channels and their cellular localization. The two mutant channels displayed a dramatic reduction in current density but had normal gating properties. The reduction in current density was caused by the retention of channel proteins in the endoplasmic reticulum (ER). Mutant channel retention could be partially reversed by incubating transfected cells at 25°C and by treating them with mexiletine (for V1378M but not A124D), or with curcumin or thapsigargin, two drugs that target ER resident proteins. It is likely that the clinical phenotypes observed in these two BrS patients were related to a surface expression defect caused by ER retention.
Collapse
Affiliation(s)
- Adrien Moreau
- Centre de Recherche, Institut Universitaire en Santé Mentale de QuébecQuebec City, QC, Canada
| | - Dagmar I. Keller
- Department of Internal Medicine and Cardiology, University Hospital ZurichZurich, Switzerland
| | - Hai Huang
- Centre de Recherche, Institut Universitaire en Santé Mentale de QuébecQuebec City, QC, Canada
| | - Véronique Fressart
- UF Cardiogénétique et Myogénétique, Groupe Hospitalier Pitié-SalpêtrièreParis, France
| | - Christian Schmied
- Department of Cardiology, University Hospital ZurichZurich, Switzerland
| | - Quadiri Timour
- INSERM ERI22, Université Claude Bernard de LyonLyon, France
| | - Mohamed Chahine
- Centre de Recherche, Institut Universitaire en Santé Mentale de QuébecQuebec City, QC, Canada
- Department of Medicine, Université LavalQuebec City, QC, Canada
| |
Collapse
|
26
|
Tulapurkar ME, Almutairy EA, Shah NG, He JR, Puche AC, Shapiro P, Singh IS, Hasday JD. Febrile-range hyperthermia modifies endothelial and neutrophilic functions to promote extravasation. Am J Respir Cell Mol Biol 2012; 46:807-14. [PMID: 22281986 DOI: 10.1165/rcmb.2011-0378oc] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a neutrophil (polymorphonuclear leukocyte; PMN)-driven lung injury that is associated with fever and heat-stroke, and involves approximately 40% mortality. In murine models of acute lung injury (ALI), febrile-range hyperthermia (FRH) enhanced PMN accumulation, vascular permeability, and epithelial injury, in part by augmenting pulmonary cysteine-x-cysteine (CXC) chemokine expression. To determine whether FRH increases chemokine responsiveness within the lung, we used in vivo and in vitro models that bypass the endogenous generation of chemokines. We measured PMN transalveolar migration (TAM) in mice after intratracheal instillations of the human CXC chemokine IL-8 in vivo, and of IL-8-directed PMN transendothelial migration (TEM) through human lung microvascular endothelial cell (HMVEC-L) monolayers in vitro. Pre-exposure to FRH increased in vivo IL-8-directed PMN TAM by 23.5-fold and in vitro TEM by 7-fold. Adoptive PMN transfer demonstrated that enhanced PMN TAM required both PMN donors and recipients to be exposed to FRH, suggesting interdependent effects on PMNs and endothelium. FRH exposure caused the activation of extracellular signal-regulated kinase (ERK) and p38 mitogen-activated protein kinase in lung homogenates and circulating PMNs, with an associated increase in HSP27 phosphorylation and stress-fiber formation. The inhibition of these signaling pathways with U0126 and SB203580 blocked the effects of FRH on PMN extravasation in vivo and in vitro. Collectively, these results (1) demonstrate that FRH augments chemokine-directed PMN extravasation through direct effects on endothelium and PMNs, (2) identify ERK and p38 signaling pathways in the effect, and (3) underscore the complex effects of physiologic temperature change on innate immune function and its potential consequences for lung injury.
Collapse
Affiliation(s)
- Mohan E Tulapurkar
- Division of Pulmonary and Critical Care, Department of Medicine, School of Medicine, University of Maryland, 20 Penn St., Baltimore, MD 21201, USA
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Weil A, Luce K, Dröse S, Wittig I, Brandt U, Osiewacz HD. Unmasking a temperature-dependent effect of the P. anserina i-AAA protease on aging and development. Cell Cycle 2011; 10:4280-90. [PMID: 22134244 PMCID: PMC3272260 DOI: 10.4161/cc.10.24.18560] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2011] [Accepted: 10/27/2011] [Indexed: 12/15/2022] Open
Abstract
Different molecular pathways involved in maintaining mitochondrial function are of fundamental importance to control cellular homeostasis. Mitochondrial i-AAA protease is part of such a surveillance system and PaIAP is the putative ortholog in the fungal aging model Podospora anserina. Here we investigated the role of PaIAP in aging and development. Deletion of the gene encoding PaIAP resulted in a specific phenotype. When incubated at 27°C, spore germination and fruiting body formation are not different from that of the corresponding wild-type strain. Unexpectedly, the lifespan of the deletion strain is strongly increased. In contrast, cultivation at an elevated temperature of 37°C leads to impairments in spore germination and fruiting body formation, and to a reduced lifespan. The higher PaIAP abundance in wild-type strains of the fungus grown at elevated temperature and the phenotype of the deletion strain unmasks a temperature-related role of the protein. The protease appears to be part of a molecular system that has evolved to allow survival under changing temperatures as they characteristically occur in nature.
Collapse
Affiliation(s)
- Andrea Weil
- Goethe University, Faculty for Biosciences & Cluster of Excellence Macromolecular Complexes Frankfurt, Institute of Molecular Biosciences, Frankfurt, Germany
| | | | | | | | | | | |
Collapse
|
28
|
Sawka MN, Leon LR, Montain SJ, Sonna LA. Integrated Physiological Mechanisms of Exercise Performance, Adaptation, and Maladaptation to Heat Stress. Compr Physiol 2011; 1:1883-928. [DOI: 10.1002/cphy.c100082] [Citation(s) in RCA: 299] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
29
|
Maity TK, Henry MM, Tulapurkar ME, Shah NG, Hasday JD, Singh IS. Distinct, gene-specific effect of heat shock on heat shock factor-1 recruitment and gene expression of CXC chemokine genes. Cytokine 2011; 54:61-7. [PMID: 21266308 PMCID: PMC3048923 DOI: 10.1016/j.cyto.2010.12.017] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2010] [Revised: 12/19/2010] [Accepted: 12/22/2010] [Indexed: 11/26/2022]
Abstract
The heat shock (HS) response, a phylogenetically conserved ubiquitous response to stress, is generally characterized by the induced expression of heat shock protein (HSP) genes. Our earlier studies showed that the stress-activated transcription factor, heat shock factor-1 (HSF1), activated at febrile range or HS temperatures also modified expression of non-HSP genes including cytokine and chemokine genes. We also showed by in silico analysis that 28 among 29 human and mouse CXC chemokine genes had multiple putative heat shock response elements (HSEs) present in their gene promoters. To further determine whether these potential HSEs were functional and bound HSF1, we analyzed the recruitment of HSF1 to promoters of 5 human CXC chemokine genes (CXCL-1, 2, 3, 5 and 8) by chromatin immunoprecipitation (ChIP) assay and analyzed the effect of HS exposure on tumor necrosis factor-α (TNFα)-induced expression of these genes in human lung epithelial-like A549 cells. HSF1 ChIP analysis showed that HSF1 was recruited to all but one of these CXC chemokine genes (CXCL-3) and HS caused a significant increase in recruitment of HSF1 to one or multiple HSEs present in the promoters of CXCL-1, 2, 5 and 8 genes. However, the effect of HS exposure on expression of these genes showed a variable gene-specific effect. For example, CXCL8 expression was markedly enhanced (p<0.05) whereas CXCL5 expression was significantly repressed (p<0.05) in cells exposed to HS coincident with TNFα stimulation. In contrast, expression of CXCL1 and CXCL2, despite HSF1 recruitment to their promoters, was not affected by HS exposure. Our results indicate that some, if not all, putative HSEs present in the CXC chemokine gene promoters are functional and recruit HSF1 in vivo but the effects on gene expression are variable and gene specific. We speculate, the physical proximity and interactions of other transcription factors and co-regulators with HSF1 could be critical to determining the effects of HS on the expression of these genes.
Collapse
Affiliation(s)
- Tapan K. Maity
- Division of Pulmonary and Critical Care, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Michael M. Henry
- Division of Pulmonary and Critical Care, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Mohan E. Tulapurkar
- Division of Pulmonary and Critical Care, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Nirav G. Shah
- Division of Pulmonary and Critical Care, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Jeffrey D. Hasday
- Division of Pulmonary and Critical Care, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Mucosal Biology Research Center, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Cytokine Core Laboratory, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Research Services of the Baltimore VA Medical Center, Baltimore, Maryland, USA 21201
| | - Ishwar S. Singh
- Division of Pulmonary and Critical Care, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Mucosal Biology Research Center, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Research Services of the Baltimore VA Medical Center, Baltimore, Maryland, USA 21201
| |
Collapse
|
30
|
Rowe RW, Strebel FR, Proett JM, Deng W, Chan D, He G, Siddik Z, Bull JMC. Fever-range whole body thermotherapy combined with oxaliplatin: a curative regimen in a pre-clinical breast cancer model. Int J Hyperthermia 2011; 26:565-76. [PMID: 20707651 DOI: 10.3109/02656736.2010.483635] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
PURPOSE Studies were conducted to test whether fever-range whole body thermal therapy would boost the efficacy of oxaliplatin chemotherapy without substantial toxicity. MATERIALS AND METHODS The effect of mild heat (40 degrees C) on oxaliplatin cytotoxicity, cellular uptake, and platinum-DNA adduct formation was studied in vitro using the MTLn3 tumour cell line. In vivo oxaliplatin was administered at various doses and times before, during and after fever-range thermal therapy (6 h at 40 degrees C) to rats bearing an MTLn3 mammary adenocarcinoma. Tumour growth, survival, and toxicity were measured to determine treatment outcome. RESULTS Heating halved the oxaliplatin IC-50 dose for MTLn3 cells. Cellular uptake of platinum and platinum adducts increased by 34% and 36%, respectively, with heat. In vivo, 50% of all rats given 10 mg/kg oxaliplatin 24 h before thermal therapy were completely immunologically cured, while a further 11% regressed their primary tumour but ultimately succumbed to metastases, and 17% experienced a limited response with increased survival. The curative response occurred only in a narrow range of doses, with most cures at 10 mg/kg. Thermochemotherapy-treated, but uncured, animals had delayed incidence and slowed growth of metastases. Anti-tumour efficacy was greatest, and toxicity was least, when oxaliplatin was administered 12 or 24 h before fever-range whole body thermal therapy. CONCLUSIONS When properly dosed and scheduled, oxaliplatin thermochemotherapy achieved permanent eradication of all primary and metastatic tumours in 50% of animals, seemingly through an immune response. Successful clinical translation of this protocol would yield hitherto unseen cures and substantial improvement in quality of life.
Collapse
Affiliation(s)
- R Wanda Rowe
- University of Texas Medical School, Division of Oncology, Houston, Texas 77030, USA
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Pinsino A, Turturici G, Sconzo G, Geraci F. Rapid changes in heat-shock cognate 70 levels, heat-shock cognate phosphorylation state, heat-shock transcription factor, and metal transcription factor activity levels in response to heavy metal exposure during sea urchin embryonic development. ECOTOXICOLOGY (LONDON, ENGLAND) 2011; 20:246-254. [PMID: 21082241 DOI: 10.1007/s10646-010-0576-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 11/04/2010] [Indexed: 05/30/2023]
Abstract
The aim of the present study was to analyze and compare the effects of several metals on the embryos of the sea urchin Paracentrotus lividus, a key species within the Mediterranean Sea ecosystem. Embryos were continuously exposed from fertilization to the following metals: 0.6 mg/l copper, 3 mg/l lead, and 6 mg/l nickel. The embryos were then monitored for metal responses at the gastrula stage, which occurred 24 h after exposure. A biochemical multi-experimental approach was taken and involved the investigation of the levels of HSC70 expression and the involvement of heat shock factor (HSF) and/or metal transcription factor (MTF) in the response. Immunoblotting assays and electrophoretic mobility shift assays (EMSA) were used to detect stress protein levels and to study the interaction between DNA and specific transcription factors, respectively. In the 1 h during exposure to heavy metals, changes in HSC70 levels and HSC70 a phosphorylation state were observed. Rapid changes in HSF and MTF DNA-binding activity also occurred during the early stages of heavy metal exposure. In contrast, few developmental abnormalities were observed at the gastrula stage but more abnormalities were observed 48 h after metal exposure. These data demonstrate that changes in HSC70 levels and phosphorylation state as well as in HSF and MTF binding activities may be used to rapidly detect responses to heavy metal exposure. Detection of biochemical and molecular changes in response to metal exposure before manifestation of morpho-pathological effects are important for the prediction of morbidity, and these markers will be useful for determining the response to exposure as part of a toxicological exposure-response experiment and for determining responses for an impact assessment.
Collapse
Affiliation(s)
- Annalisa Pinsino
- Department of Cellular and Developmental Biology, University of Palermo, Viale delle Scienze, 90128, Palermo, Italy
| | | | | | | |
Collapse
|
32
|
Shah NG, Tulapurkar ME, Singh IS, Shelhamer JH, Cowan MJ, Hasday JD. Prostaglandin E2 potentiates heat shock-induced heat shock protein 72 expression in A549 cells. Prostaglandins Other Lipid Mediat 2010; 93:1-7. [PMID: 20382255 PMCID: PMC2919605 DOI: 10.1016/j.prostaglandins.2010.03.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2009] [Revised: 03/23/2010] [Accepted: 03/31/2010] [Indexed: 12/11/2022]
Abstract
The heat shock (HS) response is an important cytoprotective response comprising the expression of heat shock proteins (HSPs) and orchestrated by the heat/stress-induced transcription factor, heat shock factor-1 (HSF-1). Previous studies suggest that the activation threshold and magnitude of the HS response may be modified by treatment with arachidonic acid (AA). We analyzed the effect of exogenous AA and its metabolites, PGE(2), LTD(4), and 15-HETE on HSF-1-dependent gene expression in A549 human respiratory epithelial-like cells. When added at 1microM, PGE(2) much more than LTD(4), but not 15-HETE increased activity of a synthetic HSF-1-dependent reporter after HS exposure (42 degrees C for 2h), but had no effect in the absence of HS. Exposing A549 cells to HS stimulated the release of PGE(2) and treatment with the cyclooxygenase inhibitor, ibuprofen, reduced HS-induced HSF-1-dependent transcription. PGE(2) increased HS-induced HSP72 mRNA and protein expression but EMSA and Western blot analysis failed to show an effect on HSF-1 DNA binding activity or post-translational modification. In summary, we showed that HS stimulates the generation of PGE(2), which augments the generation of HSPs. The clinical consequences of this pathway have yet to be determined.
Collapse
Affiliation(s)
- Nirav G. Shah
- Division of Pulmonary and Critical Care, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Mohan E. Tulapurkar
- Division of Pulmonary and Critical Care, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Ishwar S. Singh
- Division of Pulmonary and Critical Care, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Mucosal Biology Research Center, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Research Services of the Baltimore VA Medical Center, Baltimore, Maryland, USA 21201
| | - James H. Shelhamer
- Critical Care Medicine Department, National Institutes of Health, Bethesda, Maryland, USA 20892
| | - Mark J. Cowan
- Division of Pulmonary and Critical Care, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Research Services of the Baltimore VA Medical Center, Baltimore, Maryland, USA 21201
| | - Jeffrey D. Hasday
- Division of Pulmonary and Critical Care, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Mucosal Biology Research Center, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Research Services of the Baltimore VA Medical Center, Baltimore, Maryland, USA 21201
| |
Collapse
|
33
|
Raggam RB, Salzer HJF, Marth E, Heiling B, Paulitsch AH, Buzina W. Molecular detection and characterisation of fungal heat shock protein 60. Mycoses 2010; 54:e394-9. [PMID: 20667000 DOI: 10.1111/j.1439-0507.2010.01933.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Heat shock proteins (Hsp) are highly conserved molecules, which are both constitutively expressed and up-regulated in response to various stress conditions. In particular, fungal Hsp60 can act as immunodominant antigens and facilitate powerful immunological properties. A possible cellular heat shock response was investigated in eight fungi (Aspergillus fumigatus, Aspergillus terreus, Penicillium chrysogenum, Cladosporium cladosporioides, Scedosporium apiospermum, Trichophyton mentagrophytes, Candida albicans and Saccharomyces cerevisiae). Fully automated RNA extraction was followed by quantitative real-time RT-PCR targeting fungus-specific Hsp60 mRNA and sequencing of the amplicon. Levels of temperature-dependent gene expression were evaluated and rates of similarity and identity were compared. While Hsp60 mRNA was constitutively expressed in all the samples tested, a temperature-dependent induction was not shown in C. cladosporioides. In the 80-amino acid fragment from the hypothetical protein, 66% of the amino acids were identical, 20% showed a conserved and 8% a semi-conserved substitution. Our findings should contribute to a better understanding of host-pathogen relationship and suggest that fungal Hsp60 under temperature-related stress conditions might act as an immunogenic trigger in orchestrating fungi-related diseases.
Collapse
Affiliation(s)
- Reinhard B Raggam
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Austria
| | | | | | | | | | | |
Collapse
|
34
|
do Amaral BC, Connor EE, Tao S, Hayen J, Bubolz J, Dahl GE. Heat stress abatement during the dry period influences prolactin signaling in lymphocytes. Domest Anim Endocrinol 2010; 38:38-45. [PMID: 19733997 DOI: 10.1016/j.domaniend.2009.07.005] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2009] [Revised: 07/23/2009] [Accepted: 07/23/2009] [Indexed: 11/19/2022]
Abstract
Heat stress perturbs prolactin (PRL) release and affects dairy cow lactational performance and immune cell function. We hypothesized that greater PRL concentration in plasma of heat-stressed cows relative to cooled cows would decrease expression of prolactin receptor (PRL-R) mRNA and increase mRNA expression of suppressors of cytokine signaling (SOCS) in lymphocytes, altering their cytokine production. To test this hypothesis, multiparous Holstein cows were dried off 46 d before their expected calving date and assigned randomly to heat stress (HT; n=9) or cooling (CL; n=7) during the entire dry period. A second study was conducted the following year with an additional 21 cows (12 HT; 9 CL). Lymphocytes were isolated from cows at -46, -20, +2, and +20 d relative to expected calving date and mRNA expression of PRL-R, SOCS-1, SOCS-2, SOCS-3, cytokine-inducible SH2-containing protein (CIS), and heat shock protein 70 KDa A5 (HSPA5), and housekeeping genes hydroxymethylbilane synthase (HMBS), ATP synthase, H+ transporting mitochondrial F1 complex, beta subunit (ATP5B), and ribosomal protein S9 (RPS9) was analyzed by quantitative real-time reverse transcriptase polymerase chain reaction (RT-PCR). Cows exposed to HT had greater PRL concentration in plasma compared with CL cows. Measurement of lymphocyte proliferation indicated that lymphocytes of CL cows proliferated more than those from HT cows and exressed more PRL-R mRNA and less SOCS-1 and SOCS-3 mRNA relative to HT cows. Further, lymphocytes from CL cows produced more tumor necrosis factor-alpha (TNF-alpha) than those from HT cows. These results suggest that changes in PRL-signaling pathway genes during heat stress are associated with differential cytokine secretion by lymphocytes and may regulate lymphocyte proliferation in dairy cows.
Collapse
Affiliation(s)
- B C do Amaral
- Department of Animal Sciences, University of Florida, Gainesville, FL, USA 32611, United States
| | | | | | | | | | | |
Collapse
|
35
|
Cooper ZA, Ghosh A, Gupta A, Maity T, Benjamin IJ, Vogel SN, Hasday JD, Singh IS. Febrile-range temperature modifies cytokine gene expression in LPS-stimulated macrophages by differentially modifying NF-{kappa}B recruitment to cytokine gene promoters. Am J Physiol Cell Physiol 2009; 298:C171-81. [PMID: 19846753 DOI: 10.1152/ajpcell.00346.2009] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We previously showed that exposure to febrile-range temperatures (FRT, 39.5-40 degrees C) reduces LPS-induced TNF-alpha expression, in part through the direct interaction of heat shock factor-1 (HSF1) with the TNF-alpha gene promoter. However, it is not known whether exposure to FRT also modifies more proximal LPS-induced signaling events. Using HSF1-null mice, we confirmed that HSF1 is required for FRT-induced repression of TNF-alpha in vitro by LPS-stimulated bone marrow-derived macrophages and in vivo in mice challenged intratracheally with LPS. Exposing LPS-stimulated RAW 264.7 mouse macrophages to FRT reduced TNF-alpha expression while increasing IL-1beta expression despite the two genes sharing a common myeloid differentiation protein-88 (MyD88)-dependent pathway. Global activation of the three LPS-induced signaling intermediates that lead to cytokine gene expression, ERK and p38 MAPKs and NF-kappaB, was not affected by exposing RAW 264.7 cells to FRT as assessed by ERK and p38 phosphorylation and NF-kappaB in vitro DNA-binding activity and activation of a NF-kappaB-dependent synthetic promoter. However, chromatin immunoprecipitation (ChIP) analysis demonstrated that exposure to FRT reduced LPS-induced recruitment of NF-kappaB p65 to the TNF-alpha promoter while simultaneously increasing its recruitment to the IL-1beta promoter. These data suggest that FRT exerts its effects on cytokine gene expression in a gene-specific manner through distal effects on promoter activation rather than proximal receptor activation and signal transduction.
Collapse
Affiliation(s)
- Zachary A Cooper
- Univ. of Maryland School of Medicine, Health Science Facility-II, Rm. S311, 20 Penn St., Baltimore, MD 21201,USA
| | | | | | | | | | | | | | | |
Collapse
|