1
|
Canet G, Da Gama Monteiro F, Rocaboy E, Diego-Diaz S, Khelaifia B, Godbout K, Lachhab A, Kim J, Valencia DI, Yin A, Wu HT, Howell J, Blank E, Laliberté F, Fortin N, Boscher E, Fereydouni-Forouzandeh P, Champagne S, Guisle I, Hébert SS, Pernet V, Liu H, Lu W, Debure L, Rapoport DM, Ayappa I, Varga AW, Parekh A, Osorio RS, Lacroix S, Burns MP, Lucey BP, Blessing EM, Planel E. Sleep-wake variation in body temperature regulates tau secretion and correlates with CSF and plasma tau. J Clin Invest 2025; 135:e182931. [PMID: 39903530 PMCID: PMC11957704 DOI: 10.1172/jci182931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 01/31/2025] [Indexed: 02/06/2025] Open
Abstract
Sleep disturbance is bidirectionally associated with an increased risk of Alzheimer's disease and other tauopathies. While the sleep-wake cycle regulates interstitial and cerebrospinal fluid (CSF) tau levels, the underlying mechanisms remain unknown. Understanding these mechanisms is crucial, given the evidence that tau pathology spreads through neuron-to-neuron transfer, involving the secretion and internalization of pathological tau forms. Here, we combined in vitro, in vivo, and clinical methods to reveal a pathway by which changes in body temperature (BT) over the sleep-wake cycle modulate extracellular tau levels. In mice, a higher BT during wakefulness and sleep deprivation increased CSF and plasma tau levels, while also upregulating unconventional protein secretion pathway I (UPS-I) events including (a) intracellular tau dephosphorylation, (b) caspase 3-mediated cleavage of tau (TauC3), and (c) membrane translocation of tau through binding to phosphatidylinositol 4,5-bisphosphate (PIP2) and syndecan 3. In humans, the increase in CSF and plasma tau levels observed after wakefulness correlated with BT increases during wakefulness. By demonstrating that sleep-wake variation in BT regulates extracellular tau levels, our findings highlight the importance of thermoregulation in linking sleep disturbances to tau-mediated neurodegeneration and the preventative potential of thermal interventions.
Collapse
Affiliation(s)
- Geoffrey Canet
- Centre de Recherche du CHU de Québec – Université Laval, Axe Neurosciences, Québec, Québec City, Canada
- Université Laval, Faculté de Médecine, Département de Psychiatrie et Neurosciences, Québec, Québec City, Canada
| | - Felipe Da Gama Monteiro
- Centre de Recherche du CHU de Québec – Université Laval, Axe Neurosciences, Québec, Québec City, Canada
- Université Laval, Faculté de Médecine, Département de Médecine Moléculaire, Québec, Québec City, Canada
| | - Emma Rocaboy
- Université Laval, Faculté de Médecine, Département de Psychiatrie et Neurosciences, Québec, Québec City, Canada
| | - Sofia Diego-Diaz
- Université Laval, Faculté de Médecine, Département de Psychiatrie et Neurosciences, Québec, Québec City, Canada
| | - Boutheyna Khelaifia
- Centre de Recherche du CHU de Québec – Université Laval, Axe Neurosciences, Québec, Québec City, Canada
- Université Laval, Faculté de Médecine, Département de Psychiatrie et Neurosciences, Québec, Québec City, Canada
| | - Kelly Godbout
- Université Laval, Faculté de Médecine, Département de Psychiatrie et Neurosciences, Québec, Québec City, Canada
| | - Aymane Lachhab
- Université Laval, Faculté de Médecine, Département de Psychiatrie et Neurosciences, Québec, Québec City, Canada
| | - Jessica Kim
- Department of Psychiatry, NYU Grossman School of Medicine, New York, New York, USA
| | - Daphne I. Valencia
- Mount Sinai Integrative Sleep Center, Division of Pulmonary, Critical Care, and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Audrey Yin
- Department of Psychiatry, NYU Grossman School of Medicine, New York, New York, USA
| | - Hau-Tieng Wu
- Department of Psychiatry, NYU Grossman School of Medicine, New York, New York, USA
| | - Jordan Howell
- Department of Psychiatry, NYU Grossman School of Medicine, New York, New York, USA
| | - Emily Blank
- Department of Psychiatry, NYU Grossman School of Medicine, New York, New York, USA
| | - Francis Laliberté
- Centre de Recherche du CHU de Québec – Université Laval, Axe Neurosciences, Québec, Québec City, Canada
| | - Nadia Fortin
- Centre de Recherche du CHU de Québec – Université Laval, Axe Neurosciences, Québec, Québec City, Canada
| | - Emmanuelle Boscher
- Centre de Recherche du CHU de Québec – Université Laval, Axe Neurosciences, Québec, Québec City, Canada
- Université Laval, Faculté de Médecine, Département de Psychiatrie et Neurosciences, Québec, Québec City, Canada
| | | | - Stéphanie Champagne
- Université Laval, Faculté de Médecine, Département de Psychiatrie et Neurosciences, Québec, Québec City, Canada
| | - Isabelle Guisle
- Centre de Recherche du CHU de Québec – Université Laval, Axe Neurosciences, Québec, Québec City, Canada
- Université Laval, Faculté de Médecine, Département de Psychiatrie et Neurosciences, Québec, Québec City, Canada
| | - Sébastien S. Hébert
- Centre de Recherche du CHU de Québec – Université Laval, Axe Neurosciences, Québec, Québec City, Canada
- Université Laval, Faculté de Médecine, Département de Psychiatrie et Neurosciences, Québec, Québec City, Canada
| | - Vincent Pernet
- Centre de Recherche du CHU de Québec – Université Laval, Axe Neurosciences, Québec, Québec City, Canada
- Université Laval, Faculté de Médecine, Département de Médecine Moléculaire, Québec, Québec City, Canada
- Department of Neurology, Inselspital, and
- Center for Experimental Neurology (ZEN), Bern University Hospital, University of Bern, Bern, Switzerland
- Department of Biomedical Research, University of Bern, Bern, Switzerland
| | - Haiyan Liu
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - William Lu
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Ludovic Debure
- Department of Psychiatry, NYU Grossman School of Medicine, New York, New York, USA
| | - David M. Rapoport
- Mount Sinai Integrative Sleep Center, Division of Pulmonary, Critical Care, and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Indu Ayappa
- Mount Sinai Integrative Sleep Center, Division of Pulmonary, Critical Care, and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Andrew W. Varga
- Mount Sinai Integrative Sleep Center, Division of Pulmonary, Critical Care, and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Ankit Parekh
- Mount Sinai Integrative Sleep Center, Division of Pulmonary, Critical Care, and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Ricardo S. Osorio
- Department of Psychiatry, NYU Grossman School of Medicine, New York, New York, USA
| | - Steve Lacroix
- Centre de Recherche du CHU de Québec – Université Laval, Axe Neurosciences, Québec, Québec City, Canada
- Université Laval, Faculté de Médecine, Département de Médecine Moléculaire, Québec, Québec City, Canada
| | - Mark P. Burns
- Laboratory for Brain Injury and Dementia, Department of Neuroscience, Georgetown University Medical Center, Washington, DC, USA
| | - Brendan P. Lucey
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Esther M. Blessing
- Department of Psychiatry, NYU Grossman School of Medicine, New York, New York, USA
| | - Emmanuel Planel
- Centre de Recherche du CHU de Québec – Université Laval, Axe Neurosciences, Québec, Québec City, Canada
- Université Laval, Faculté de Médecine, Département de Psychiatrie et Neurosciences, Québec, Québec City, Canada
| |
Collapse
|
2
|
Teuben MJ, Halvachizadeh S, Kalbas Y, Qiao Z, Cesarovic N, Weisskopf M, Teuber H, Kalbitz M, Cinelli P, Pfeifer R, Pape H, TREAT Research Group. Cellular activation status in femoral shaft fracture hematoma following different reaming techniques - A large animal model. J Orthop Res 2022; 40:2822-2830. [PMID: 35301740 PMCID: PMC9790649 DOI: 10.1002/jor.25309] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 08/20/2021] [Accepted: 02/14/2022] [Indexed: 02/04/2023]
Abstract
The local inflammatory impact of different reaming protocols in intramedullary nailing has been sparsely investigated. We examined the effect of different reaming protocols on fracture hematoma (FH) immunological characteristics in pigs. To do so, a standardized midshaft femur fracture was induced in adult male pigs. Fractures were treated with conventional reamed femoral nailing (group RFN, n = 6); unreamed femoral nailing (group UFN, n = 6); reaming with a Reamer Irrigator Aspirator device (group RIA, n = 12). Animals were observed for 6 h and FH was collected. FH-cell apoptosis and neutrophil receptor expression (Mac-1/CD11b and FcγRIII/CD16) were studied by flow cytometry and local temperature changes were analyzed. The study demonstrates that apoptosis-rates of FH-immune cells were significantly lower in group RIA (3.50 ± 0.53%) when compared with non-RIA groups: (group UFN 12.50 ± 5.22%, p = 0.028 UFN vs. RIA), (group RFN 13.30 ± 3.18%, p < 0.001, RFN vs. RIA). Further, RIA-FH showed lower neutrophil CD11b/CD16 expression when compared with RFN (mean difference of 43.0% median fluorescence intensity (MFI), p = 0.02; and mean difference of 35.3% MFI, p = 0.04, respectively). Finally, RIA induced a transient local hypothermia and hypothermia negatively correlated with both FH-immune cell apoptosis and neutrophil activation. In conclusion, immunologic changes observed in FH appear to be modified by certain reaming techniques. Irrigation during reaming was associated with transient local hypothermia, decreased apoptosis, and reduced neutrophil activation. Further study is warranted to examine whether the rinsing effect of RIA, specific tissue removal by reaming, or thermal effects predominantly determine local inflammatory changes during reaming.
Collapse
Affiliation(s)
- Michel Paul Johan Teuben
- Department of TraumatologyUniversity Hospital ZurichZurichSwitzerland,Harald Tscherne Laboratory for Orthopedic ResearchZurichSwitzerland
| | - Sascha Halvachizadeh
- Department of TraumatologyUniversity Hospital ZurichZurichSwitzerland,Harald Tscherne Laboratory for Orthopedic ResearchZurichSwitzerland
| | - Yannik Kalbas
- Department of TraumatologyUniversity Hospital ZurichZurichSwitzerland,Harald Tscherne Laboratory for Orthopedic ResearchZurichSwitzerland
| | - Zhi Qiao
- Department of Trauma and Reconstructive SurgeryUniversity Clinic RWTH AachenAachenGermany
| | - Nikola Cesarovic
- Division of Surgical ResearchUniversity of Zurich and University Hospital ZurichZurichSwitzerland,Department of Health Sciences, Translational Cardiovascular TechnologiesETH ZürichZürichSwitzerland,Department of Cardiothoracic and Vascular SurgeryGerman Heart Institute BerlinBerlinGermany
| | - Miriam Weisskopf
- Division of Surgical ResearchUniversity of Zurich and University Hospital ZurichZurichSwitzerland
| | - Henrik Teuber
- Department of TraumatologyUniversity Hospital ZurichZurichSwitzerland,Harald Tscherne Laboratory for Orthopedic ResearchZurichSwitzerland
| | - Miriam Kalbitz
- Department of Trauma and Orthopedic SurgeryUniversity Hospital Erlangen, Friedrich‐Alexander‐University NürnbergErlangenGermany
| | - Paolo Cinelli
- Department of TraumatologyUniversity Hospital ZurichZurichSwitzerland,Harald Tscherne Laboratory for Orthopedic ResearchZurichSwitzerland,Division of Surgical ResearchUniversity of Zurich and University Hospital ZurichZurichSwitzerland
| | - Roman Pfeifer
- Department of TraumatologyUniversity Hospital ZurichZurichSwitzerland,Harald Tscherne Laboratory for Orthopedic ResearchZurichSwitzerland
| | - Hans‐Christoph Pape
- Department of TraumatologyUniversity Hospital ZurichZurichSwitzerland,Harald Tscherne Laboratory for Orthopedic ResearchZurichSwitzerland
| | | |
Collapse
|
3
|
Deredge D, Wintrode PL, Tulapurkar ME, Nagarsekar A, Zhang Y, Weber DJ, Shapiro P, Hasday JD. A temperature-dependent conformational shift in p38α MAPK substrate-binding region associated with changes in substrate phosphorylation profile. J Biol Chem 2019; 294:12624-12637. [PMID: 31213525 DOI: 10.1074/jbc.ra119.007525] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2019] [Revised: 05/13/2019] [Indexed: 01/09/2023] Open
Abstract
Febrile-range hyperthermia worsens and hypothermia mitigates lung injury, and temperature dependence of lung injury is blunted by inhibitors of p38 mitogen-activated protein kinase (MAPK). Of the two predominant p38 isoforms, p38α is proinflammatory and p38β is cytoprotective. Here, we analyzed the temperature dependence of p38 MAPK activation, substrate interaction, and tertiary structure. Incubating HeLa cells at 39.5 °C stimulated modest p38 activation, but did not alter tumor necrosis factor-α (TNFα)-induced p38 activation. In in vitro kinase assays containing activated p38α and MAPK-activated kinase-2 (MK2), MK2 phosphorylation was 14.5-fold greater at 39.5 °C than at 33 °C. By comparison, we observed only 3.1- and 1.9-fold differences for activating transcription factor-2 (ATF2) and signal transducer and activator of transcription-1α (STAT1α) and a 7.7-fold difference for p38β phosphorylation of MK2. The temperature dependence of p38α:substrate binding affinity, as measured by surface plasmon resonance, paralleled substrate phosphorylation. Hydrogen-deuterium exchange MS (HDX-MS) of p38α performed at 33, 37, and 39.5 °C indicated temperature-dependent conformational changes in an α helix near the common docking and glutamate:aspartate substrate-binding domains at the known binding site for MK2. In contrast, HDX-MS analysis of p38β did not detect significant temperature-dependent conformational changes in this region. We observed no conformational changes in the catalytic domain of either isoform and no corresponding temperature dependence in the C-terminal p38α-interacting region of MK2. Because MK2 participates in the pathogenesis of lung injury, the observed changes in the structure and function of proinflammatory p38α may contribute to the temperature dependence of acute lung injury.
Collapse
Affiliation(s)
- Daniel Deredge
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland 21201
| | - Patrick L Wintrode
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland 21201
| | - Mohan E Tulapurkar
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Ashish Nagarsekar
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Yinghua Zhang
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - David J Weber
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Paul Shapiro
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland 21201
| | - Jeffrey D Hasday
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland 21201 .,Medicine and Research Services, Baltimore Veterans Affairs Medical Center, Baltimore, Maryland 21201
| |
Collapse
|
4
|
Keitelman IA, Sabbione F, Shiromizu CM, Giai C, Fuentes F, Rosso D, Ledo C, Miglio Rodriguez M, Guzman M, Geffner JR, Galletti J, Jancic C, Gómez MI, Trevani AS. Short-Term Fever-Range Hyperthermia Accelerates NETosis and Reduces Pro-inflammatory Cytokine Secretion by Human Neutrophils. Front Immunol 2019; 10:2374. [PMID: 31681277 PMCID: PMC6813732 DOI: 10.3389/fimmu.2019.02374] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 09/23/2019] [Indexed: 01/18/2023] Open
Abstract
Fever is a hallmark of infections and inflammatory diseases, represented by an increase of 1-4°C in core body temperature. Fever-range hyperthermia (FRH) has been shown to increase neutrophil recruitment to local sites of infection. Here, we evaluated the impact of a short period (1 h) of FRH (STFRH) on pro-inflammatory and bactericidal human neutrophil functions. STFRH did not affect neutrophil spontaneous apoptosis but reverted the lipopolysaccharide (LPS)-induced anti-apoptotic effect compared with that under normothermic conditions. Furthermore, STFRH accelerated phorbol myristate acetate (PMA)-induced NETosis evaluated either by the nuclear DNA decondensation at 2 h post-stimulation or by the increase in extracellular DNA that colocalized with myeloperoxidase (MPO) at 4 h post-stimulation. Increased NETosis upon STFRH was associated with an increase in reactive oxygen species (ROS) production but not in autophagy levels. STFRH also increased NETosis in response to Pseudomonas aeruginosa challenge but moderately reduced its phagocytosis. However, these STFRH-induced effects did not influence the ability of neutrophils to kill bacteria after 4 h of co-culture. STFRH also significantly reduced neutrophil capacity to release the pro-inflammatory cytokines chemokine (C-X-C motif) ligand 8/interleukin 8 (CXCL8/IL-8) and IL-1β in response to LPS and P. aeruginosa challenge. Altogether, these results indicate that a short and mild hyperthermal period is enough to modulate neutrophil responses to bacterial encounter. They also suggest that fever spikes during bacterial infections might lead neutrophils to trigger an emergency response promoting neutrophil extracellular trap (NET) formation to ensnare bacteria in order to wall off the infection and to reduce their release of pro-inflammatory cytokines in order to limit the inflammatory response.
Collapse
Affiliation(s)
- Irene A. Keitelman
- Laboratorio de Inmunidad Innata, Instituto de Medicina Experimental (IMEX)-CONICET, Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Florencia Sabbione
- Laboratorio de Inmunidad Innata, Instituto de Medicina Experimental (IMEX)-CONICET, Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Carolina M. Shiromizu
- Laboratorio de Inmunidad Innata, Instituto de Medicina Experimental (IMEX)-CONICET, Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Constanza Giai
- Instituto de Investigaciones en Microbiología y Parasitología Médica (IMPaM) UBA-CONICET, Buenos Aires, Argentina
| | - Federico Fuentes
- Laboratorio de Microscopía, Instituto de Medicina Experimental (IMEX)-CONICET, Academia Nacional de Medicina, Buenos Aires, Argentina
| | - David Rosso
- Laboratorio de Inmunidad Innata, Instituto de Medicina Experimental (IMEX)-CONICET, Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Camila Ledo
- Instituto de Investigaciones en Microbiología y Parasitología Médica (IMPaM) UBA-CONICET, Buenos Aires, Argentina
- Departamento de Investigaciones Biomédicas y Biotecnológicas, Centro de Estudios Biomédicos, Biotecnológicos, Ambientales y de Diagnóstico (CEBBAD), Universidad Maimónides, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Maximiliano Miglio Rodriguez
- Laboratorio de Inmunidad Innata, Instituto de Medicina Experimental (IMEX)-CONICET, Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Mauricio Guzman
- Laboratorio de Inmunidad Innata, Instituto de Medicina Experimental (IMEX)-CONICET, Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Jorge R. Geffner
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), CONICET, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Jeremías Galletti
- Laboratorio de Inmunidad Innata, Instituto de Medicina Experimental (IMEX)-CONICET, Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Carolina Jancic
- Laboratorio de Inmunidad Innata, Instituto de Medicina Experimental (IMEX)-CONICET, Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Marisa I. Gómez
- Departamento de Investigaciones Biomédicas y Biotecnológicas, Centro de Estudios Biomédicos, Biotecnológicos, Ambientales y de Diagnóstico (CEBBAD), Universidad Maimónides, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
- Departamento de Microbiología, Parasitología e Inmunología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Analía S. Trevani
- Laboratorio de Inmunidad Innata, Instituto de Medicina Experimental (IMEX)-CONICET, Academia Nacional de Medicina, Buenos Aires, Argentina
- Departamento de Microbiología, Parasitología e Inmunología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
- *Correspondence: Analía S. Trevani
| |
Collapse
|
5
|
Díaz FE, Dantas E, Cabrera M, Benítez CA, Delpino MV, Duette G, Rubione J, Sanjuan N, Trevani AS, Geffner J. Fever-range hyperthermia improves the anti-apoptotic effect induced by low pH on human neutrophils promoting a proangiogenic profile. Cell Death Dis 2016; 7:e2437. [PMID: 27787523 PMCID: PMC5133997 DOI: 10.1038/cddis.2016.337] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 09/03/2016] [Accepted: 09/20/2016] [Indexed: 01/18/2023]
Abstract
Neutrophils have the shortest lifespan among leukocytes and usually die via apoptosis, limiting their deleterious potential. However, this tightly regulated cell death program can be modulated by pathogen-associated molecular patterns (PAMPs), danger-associated molecular pattern (DAMPs), and inflammatory cytokines. We have previously reported that low pH, a hallmark of inflammatory processes and solid tumors, moderately delays neutrophil apoptosis. Here we show that fever-range hyperthermia accelerates the rate of neutrophil apoptosis at neutral pH but markedly increases neutrophil survival induced by low pH. Interestingly, an opposite effect was observed in lymphocytes; hyperthermia plus low pH prevents lymphocyte activation and promotes the death of lymphocytes and lymphoid cell lines. Analysis of the mechanisms through which hyperthermia plus low pH increased neutrophil survival revealed that hyperthermia further decreases cytosolic pH induced by extracellular acidosis. The fact that two Na+/H+ exchanger inhibitors, 5-(N-ethyl-N-isopropyl) amiloride (EIPA) and amiloride, reproduced the effects induced by hyperthermia suggested that it prolongs neutrophil survival by inhibiting the Na+/H+ antiporter. The neutrophil anti-apoptotic effect induced by PAMPs, DAMPs, and inflammatory cytokines usually leads to the preservation of the major neutrophil effector functions such as phagocytosis and reactive oxygen species (ROS) production. In contrast, our data revealed that the anti-apoptotic effect induced by low pH and hyperthermia induced a functional profile characterized by a low phagocytic activity, an impairment in ROS production and a high ability to suppress T-cell activation and to produce the angiogenic factors VEGF, IL-8, and the matrix metallopeptidase 9 (MMP-9). These results suggest that acting together fever and local acidosis might drive the differentiation of neutrophils into a profile able to promote both cancer progression and tissue repair during the late phase of inflammation, two processes that are strongly dependent on the local production of angiogenic factors by infiltrating immune cells.
Collapse
Affiliation(s)
- Fernando Erra Díaz
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), CONICET, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Ezequiel Dantas
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), CONICET, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Maia Cabrera
- Instituto de Investigaciones Farmacológicas (ININFA), CONICET, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Constanza A Benítez
- Instituto de Inmunología, Genética y Metabolismo (INIGEM), CONICET, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - María V Delpino
- Instituto de Inmunología, Genética y Metabolismo (INIGEM), CONICET, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Gabriel Duette
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), CONICET, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Julia Rubione
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), CONICET, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Norberto Sanjuan
- Instituto de Investigaciones en Microbiología y Parasitología Médica (IMPAM), CONICET, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Analía S Trevani
- Instituto de Medicina Experimental (IMEX), CONICET, Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Jorge Geffner
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), CONICET, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
6
|
Lecchi C, Rota N, Vitali A, Ceciliani F, Lacetera N. In vitro assessment of the effects of temperature on phagocytosis, reactive oxygen species production and apoptosis in bovine polymorphonuclear cells. Vet Immunol Immunopathol 2016; 182:89-94. [PMID: 27863557 DOI: 10.1016/j.vetimm.2016.10.007] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 09/02/2016] [Accepted: 10/19/2016] [Indexed: 01/02/2023]
Abstract
Heat stress exerts a direct negative effect on farm animal health, triggering physiological responses. Environmental high temperature induces immunosuppression in dairy cows, increasing the risk of mastitis and milk somatic cell counts. The influence of heat stress on leukocytes activities has not been fully elucidated. The present in vitro study was aimed at assessing whether the exposure to temperature simulating conditions of severe whole body hyperthermia affects defensive functions of bovine blood polymorphonuclear cells. Blood was collected from seven clinically healthy, multiparous, late lactating Holstein cows. After isolation, PMN were incubated at either 39 or 41°C. Phagocytosis, respiratory burst and apoptosis were then investigated. The selected temperatures of 39°C or 41°C mimicked conditions of normothermia or severe heat stress, respectively. Phagocytosis assay was carried out by measuring the fluorescence of phagocyted fluorescein-labelled E. coli bioparticles. The modulation of oxidative burst activity was studied by the cytochrome C reduction method. Apoptosis was determined by measuring the activities of two enzymes that play an effector role in the process, namely Caspase-3 and Caspase-7. Statistical analyses were performed using SPSS 22.0. A Student t-test for paired samples and a Generalised Estimating Equation were used based on data distribution. The phagocytosis rate was reduced (-37%, P<0.01) when PMN were incubated for 2h at 41°C, when compared to phagocytosis rate measured at 39°C. The oxidative burst, as determined by extracellular production of reactive oxygen species (ROS), was also reduced by the exposure of cells to 41°C compared to 39°C. Such reduction ranged between -2 and -21% (P<0.05). Apoptosis rate was not affected by different temperatures. The results reported in this study suggest that phagocytosis and ROS production in PMN exposed to severe high temperature are impaired, partially explaining the higher occurrence of infections during periods of hot weather.
Collapse
Affiliation(s)
- Cristina Lecchi
- Dipartimento di Medicina Veterinaria, Università di Milano, Milano, Italy.
| | - Nicola Rota
- Dipartimento di Scienze Veterinarie per la Salute, la Produzione Animale e la Sicurezza Alimentare, Università di Milano, Milano, Italy
| | - Andrea Vitali
- Dipartimento di Scienze Agrarie e Forestali, Università della Tuscia, Viterbo, Italy
| | - Fabrizio Ceciliani
- Dipartimento di Medicina Veterinaria, Università di Milano, Milano, Italy
| | - Nicola Lacetera
- Dipartimento di Scienze Agrarie e Forestali, Università della Tuscia, Viterbo, Italy
| |
Collapse
|
7
|
Andocs G, Rehman MU, Zhao QL, Tabuchi Y, Kanamori M, Kondo T. Comparison of biological effects of modulated electro-hyperthermia and conventional heat treatment in human lymphoma U937 cells. Cell Death Discov 2016; 2:16039. [PMID: 27551529 PMCID: PMC4979466 DOI: 10.1038/cddiscovery.2016.39] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 04/28/2016] [Accepted: 05/01/2016] [Indexed: 11/10/2022] Open
Abstract
Loco-regional hyperthermia treatment has long history in oncology. Modulated electro-hyperthermia (mEHT, trade name: oncothermia) is an emerging curative treatment method in this field due to its highly selective actions. The impedance-matched, capacitive-coupled modulated radiofrequency (RF) current is selectively focused in the malignant cell membrane of the cancer cells. Our objective is studying the cell-death process and comparing the cellular effects of conventional water-bath hyperthermia treatment to mEHT. The U937 human histiocytic lymphoma cell line was used for the experiments. In the case of conventional hyperthermia treatment, cells were immersed in a thermoregulated water bath, whereas in the case of mEHT, the cells were treated using a special RF generator (LabEHY, Oncotherm) and an applicator. The heating dynamics, the maximum temperature reached (42 °C) and the treatment duration (30 min) were exactly the same in both cases. Cell samples were analysed using different flow cytometric methods as well as microarray gene expression assay and western blot analysis was also used to reveal the molecular basis of the induced effects. Definite difference was observed in the biological response to different heat treatments. At 42 °C, only mEHT induced significant apoptotic cell death. The GeneChip analysis revealed a whole cluster of genes, which are highly up-regulated in case of only RF heating, but not in conventional heating. The Fas, c-Jun N-terminal kinases (JNK) and ERK signalling pathway was the dominant factor to induce apoptotic cell death in mEHT, whereas the cell-protective mechanisms dominated in case of conventional heating. This study has clearly shown that conventional hyperthermia and RF mEHT can result in different biological responses at the same temperature. The reason for the difference is the distinct, non-homogenous energy distribution on the cell membrane, which activates cell death-related signalling pathways in mEHT treatment but not in conventional heat treatment.
Collapse
Affiliation(s)
- G Andocs
- Department of Radiological Sciences, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama , Toyama, Japan
| | - M U Rehman
- Department of Radiological Sciences, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama , Toyama, Japan
| | - Q-L Zhao
- Department of Radiological Sciences, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama , Toyama, Japan
| | - Y Tabuchi
- Division of Molecular Genetics, Life Science Research Center, University of Toyama , Toyama, Japan
| | - M Kanamori
- Department of Human Science, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama , Toyama, Japan
| | - T Kondo
- Department of Radiological Sciences, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama , Toyama, Japan
| |
Collapse
|
8
|
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
|
9
|
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
|
10
|
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
|
11
|
Sun J, Guo M, Pang H, Qi J, Zhang J, Ge Y. Treatment of malignant glioma using hyperthermia. Neural Regen Res 2014; 8:2775-82. [PMID: 25206588 PMCID: PMC4145998 DOI: 10.3969/j.issn.1673-5374.2013.29.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2013] [Accepted: 07/20/2013] [Indexed: 12/28/2022] Open
Abstract
Thirty pathologically diagnosed patients with grade III–IV primary or recurrent malignant glioma (tumor diameter 3–7 cm) were randomly divided into two groups. The control group underwent conventional radiotherapy and chemotherapy. In the hyperthermia group, primary cases received hyperthermia treatment, and patients with recurrent tumors were treated with hyperthermia in com-bination with radiotherapy and chemotherapy. Hyperthermia treatment was administered using a 13.56-MHz radio frequency hyperthermia device. Electrodes were inserted into the tumor with the aid of a CT-guided stereotactic apparatus and heat was applied for 1 hour. During 3 months after hyperthermia, patients were evaluated with head CT or MRI every month. Gliomas in the hyper-thermia group exhibited growth retardation or growth termination. Necrosis was evident in 80% of the heated tumor tissue and there was a decrease in tumor diameter. Our findings indicate that ra-dio frequency hyperthermia has a beneficial effect in the treatment of malignant glioma.
Collapse
Affiliation(s)
- Jiahang Sun
- Department of Neurosurgery, the Second Affiliated Hospital of Harbin Medical University, Harbin 150000, Heilongjiang Province, China
| | - Mian Guo
- Department of Neurosurgery, the Second Affiliated Hospital of Harbin Medical University, Harbin 150000, Heilongjiang Province, China
| | - Hengyuan Pang
- Department of Neurosurgery, the Second Affiliated Hospital of Harbin Medical University, Harbin 150000, Heilongjiang Province, China
| | - Jingtao Qi
- Department of Neurosurgery, the Second Affiliated Hospital of Harbin Medical University, Harbin 150000, Heilongjiang Province, China
| | - Jinwei Zhang
- Department of Neurosurgery, the Second Affiliated Hospital of Harbin Medical University, Harbin 150000, Heilongjiang Province, China
| | - Yunlong Ge
- Department of Neurosurgery, the Second Affiliated Hospital of Harbin Medical University, Harbin 150000, Heilongjiang Province, China
| |
Collapse
|
12
|
Yang ZY, Kan JT, Cheng ZY, Wang XL, Zhu YZ, Guo W. Daphnoretin-induced apoptosis in HeLa cells: a possible mitochondria-dependent pathway. Cytotechnology 2013; 66:51-61. [PMID: 24091880 DOI: 10.1007/s10616-013-9536-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2012] [Accepted: 01/04/2013] [Indexed: 11/27/2022] Open
Abstract
Daphnoretin is a bicoumarin compound isolated from a natural product, Wikstroemia indica, which has been used to treat many diseases. It has strong antiviral and anti-tumor activities. Taking the anti-tumor activity of daphnoretin as a starting point, the present study aimed to test the pro-apoptotic effect of daphnoretin and its underlying mechanism in HeLa cells. The inhibitory effects of daphnoretin on viability and proliferation of HeLa cells were determined by the MTT assay. Daphnoretin-induced apoptotic morphological changes were analyzed by mitochondrial membrane potential and Hoechst staining. The number and stage of apoptotic HeLa cells were determined by flow cytometry. Gene expression was determined by reverse-transcription polymerase chain reaction. Protein expression was determined by western blot. The caspase activity of HeLa cells was detected by a caspase-3 and caspase-9 colorimetric assay kit. We found that daphnoretin significantly inhibited HeLa cells' viability by the MTT assay and flow cytometry. The nuclei of the apoptotic cells exhibited strong, blue fluorescence in Hoechst staining. Bax mRNA and protein levels were increased while bcl-2 mRNA levels were decreased after daphnoretin treatment. Daphnoretin also activated both caspase-3 and caspase-9. These findings suggest that daphnoretin promotes apoptosis of HeLa cells in a mitochondria-mediated way. Daphnoretin therefore has potential to be a promising drug to treat uterine cervix cancer.
Collapse
Affiliation(s)
- Zhen-Yu Yang
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200011, China
| | | | | | | | | | | |
Collapse
|
13
|
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
|
14
|
Mastrorilli C, Welles EG, Hux B, Christopherson PW. Botryoid nuclei in the peripheral blood of a dog with heatstroke. Vet Clin Pathol 2013; 42:145-9. [PMID: 23731001 DOI: 10.1111/vcp.12041] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
An EDTA-anticoagulated blood sample collected from a 1.5-year-old, intact male, English Bulldog was submitted for a CBC. The CBC data and blood smear evaluation revealed borderline high hematocrit (54%, reference interval 37-55%), inappropriate rubricytosis, moderate leukopenia due to both mature neutropenia and lymphopenia, and mild thrombocytopenia. Numerous leukocytes showed evidence of karyolysis, pyknosis, and karyorhexis, and apoptotic bodies were frequent in the background. Many neutrophils had botryoid nuclei characterized by increased numbers of nuclear segments radially arranged with spoke-like, delicate chromatin filaments connecting the segments centrally. The finding of botryoid nuclei and inappropriate rubricytosis was indicative of severe hyperthermia, such as heatstroke. The dog had been exercised a long time during conditions of high temperature and humidity until he collapsed. The dog was diagnosed with severe heatstroke, hypovolemic shock, disseminated intravascular coagulation, and multiorgan dysfunction syndrome. Despite aggressive treatment, the patient died of cardiopulmonary arrest. Botryoid nuclei are frequent in people with heatstroke. In the authors' experience, botryoid nuclei are seen commonly in dogs with heatstroke, but they have never been reported in veterinary medicine. The presence of petechiation with only mild thrombocytopenia and inappropriate rubricytosis also is suggestive of heatstroke and manifests ongoing life-threatening vascular derangement.
Collapse
Affiliation(s)
- Cinzia Mastrorilli
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, USA.
| | | | | | | |
Collapse
|
15
|
Nagarsekar A, Tulapurkar ME, Singh IS, Atamas SP, Shah NG, Hasday JD. Hyperthermia promotes and prevents respiratory epithelial apoptosis through distinct mechanisms. Am J Respir Cell Mol Biol 2012; 47:824-33. [PMID: 22962066 DOI: 10.1165/rcmb.2012-0105oc] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Hyperthermia has been shown to confer cytoprotection and to augment apoptosis in different experimental models. We analyzed the mechanisms of both effects in the same mouse lung epithelial (MLE) cell line (MLE15). Exposing MLE15 cells to heat shock (HS; 42°C, 2 h) or febrile-range hyperthermia (39.5°C) concurrent with activation of the death receptors, TNF receptor 1 or Fas, greatly accelerated apoptosis, which was detectable within 30 minutes and was associated with accelerated activation of caspase-2, -8, and -10, and the proapoptotic protein, Bcl2-interacting domain (Bid). Caspase-3 activation and cell death were partially blocked by inhibitors targeting all three initiator caspases. Cells expressing the IκB superrepessor were more susceptible than wild-type cells to TNF-α-induced apoptosis at 37°C, but HS and febrile-range hyperthermia still increased apoptosis in these cells. Delaying HS for 3 hours after TNF-α treatment abrogated its proapoptotic effect in wild-type cells, but not in IκB superrepressor-expression cells, suggesting that TNF-α stimulates delayed resistance to the proapoptotic effects of HS through an NF-κB-dependent mechanism. Pre-exposure to 2-hour HS beginning 6 to16 hours before TNF-α treatment or Fas activation reduced apoptosis in MLE15 cells. The antiapoptotic effects of HS pretreatment were reduced in TNF-α-treated embryonic fibroblasts from heat shock factor-1 (HSF1)-deficient mice, but the proapoptotic effects of concurrent HS were preserved. Thus, depending on the temperature and timing relative to death receptor activation, hyperthermia can exert pro- and antiapoptotic effects through distinct mechanisms.
Collapse
Affiliation(s)
- Ashish Nagarsekar
- Pulmonary and Critical Care Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | | | | | | | | | | |
Collapse
|
16
|
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
|
17
|
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
|
18
|
Zgair AK. The effect of high temperature on the kinetics of lipopolysaccharide (LPS)-induced human monocytes activity in vitro. Cell Immunol 2012; 275:55-60. [DOI: 10.1016/j.cellimm.2012.03.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2011] [Revised: 03/09/2012] [Accepted: 03/13/2012] [Indexed: 11/25/2022]
|
19
|
Beachy SH, Repasky EA. Toward establishment of temperature thresholds for immunological impact of heat exposure in humans. Int J Hyperthermia 2011; 27:344-52. [PMID: 21591898 DOI: 10.3109/02656736.2011.562873] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
There is interest in understanding the health impact of thermal effects as a result of exposure of humans to radiofrequency/microwave (RF/MW) fields. Immune cells and responses are affected by modest changes in temperature and it is important to quantify these effects and establish safety thresholds similar to what has been done with other tissue targets. Since previous summaries of thresholds for thermal damage to normal tissues have not focused much attention to cells of the immune system, this summary highlights recent studies which demonstrate positive and some negative effects of temperature shifts on human immune cells. We emphasise literature reporting adverse immunological endpoints (such as cell damage, death and altered function) and provide the temperature at which these effects were noted. Whereas there have been many in vitro studies of adverse temperature effects on immune cells, there has been limited validation of these temperature effects in vivo. However, data from heat stress/stroke patients do provide some information regarding core temperatures (40°C) at which thermal damage to immunological processes can begin to occur. We conclude that there is considerable need for more quantitative time temperature assessments using relevant animal models, more complete kinetic analyses to determine how long immunological effects persist, and for analysis of whether frequency of exposure has impact on immune function. To date, no attempt to categorise effects by using cumulative thermal dose measurements (e.g. cumulative equivalent minutes at a given temperature) has been conducted for cells or tissues of the immune system, representing a major gap in this field.
Collapse
Affiliation(s)
- Sarah H Beachy
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | | |
Collapse
|
20
|
Aref S, Abdullah D, Fouda M, El Menshawy N, Azmy E, Bassam A, Menessy A, El Refaei M. Neutrophil Apoptosis in Neutropenic Patients With Hepatitis C Infection: Role of Caspases 3, 10, and GM-CSF. Indian J Hematol Blood Transfus 2011; 27:81-7. [PMID: 22654297 PMCID: PMC3136665 DOI: 10.1007/s12288-011-0067-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2010] [Accepted: 04/10/2011] [Indexed: 10/18/2022] Open
Abstract
Patients with chronic HCV infection are prone to increased susceptibility bacterial infection due to neutropenia complicating the course of this disease. Neutropenia in those patients may stem from enhanced neutrophil apoptosis. However, the molecular mechanism of neutrophil apoptosis has not been clearly defined. Neutrophils harvested from 26 neutropenic patients with hepatitis C infection and nine age and sex-matched healthy control subjects were examined for the degree of apoptosis. Neutrophil apoptosis was quantified by flow cytometry through determination of annexin-V expression at 0 time (fresh neutrophil), and 24 h culture. Neutrophils from healthy subjects were also incubated with either 10% heterologous normal or neutropenic sera, with and without 10 µg GM-CSF. Caspases 3, 10 were assessed colormetrically in neutrophils at 0 times and after 24 h culture. At 0 time culture the neutrophil apoptosis of the HCV patients was in significantly higher as compared to that of normal control (P = 0.059). At 24 h culture patients neutrophils cultured with neutropenic patients own sera showed neutrophil apoptosis significantly increased as compared to that at 0 time culture and this effect was significantly attenuated in similar culture with addition of GM-CSF (P < 0.001). On the other hand patient's neutrophil cultured with normal sera showed insignificantly increased neutrophil apoptosis at 24 h culture as compared to that at 0 time culture. Caspases 3 and 10 activities were significantly higher in patients neutrophil after 24 h cultured with patients own sera as compared to 0 time culture (P < 0.001 for both). Addition of GM-CSF to the neutrophil culture down regulates the caspases 3 and 10 activities. The correlation study between annexin-V expression and caspases activities revealed a borderline positive correlation between annexin-V and caspase 3 (r = 0.376, P = 0.058), and significant positive correlation with caspase 10 activity (r = 0.494, P = 0.01). In conclusion, these findings suggest that enhanced neutrophil apoptosis demonstrated in neutropenic patients with HCV infection might be induced through activation of caspase 10 and is attenuated by GM-CSF.
Collapse
Affiliation(s)
- Salah Aref
- Hematology Unit, Department of Clinical Pathology, Mansoura Faculty of Medicine, Mansoura, Egypt
| | - Doaa Abdullah
- Hematology Unit, Department of Clinical Pathology, Mansoura Faculty of Medicine, Mansoura, Egypt
| | - Manal Fouda
- Hematology Unit, Department of Clinical Pathology, Mansoura Faculty of Medicine, Mansoura, Egypt
| | - Nadia El Menshawy
- Hematology Unit, Department of Clinical Pathology, Mansoura Faculty of Medicine, Mansoura, Egypt
| | - Emaad Azmy
- Department of Internal Medicine, Mansoura Faculty of Medicine, Mansoura, Egypt
| | - Ansaf Bassam
- Department of Internal Medicine, Mansoura Faculty of Medicine, Mansoura, Egypt
| | - Aymen Menessy
- Department of Internal Medicine, Mansoura Faculty of Medicine, Mansoura, Egypt
| | | |
Collapse
|
21
|
Rapid decrease of CD16 (FcγRIII) expression on heat-shocked neutrophils and their recognition by macrophages. J Biomed Biotechnol 2011; 2011:284759. [PMID: 21541219 PMCID: PMC3085332 DOI: 10.1155/2011/284759] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2010] [Accepted: 02/22/2011] [Indexed: 12/19/2022] Open
Abstract
Accumulation of neutrophils in the site of inflammation is a
typical mechanism of innate immunity. The accumulated neutrophils
are exposed to stressogenic factors usually associated with
inflammation. Here, we studied response of human peripheral blood
neutrophils subjected to short, febrile-range heat stress. We
show that 90 min heat stress slowed down the spontaneous apoptosis
of neutrophils. In the absence of typical markers of apoptosis the
heat-shocked neutrophils induced antiinflammatory effect in human
monocyte-derived macrophages (hMDMs), yet without being engulfed.
Importantly, the expression of FcγRIII (CD16) was sharply reduced.
Surprisingly, concentration of the soluble CD16 did not change in
heat-shocked neutrophil supernates indicating that the reduction
of the cell surface CD16 was achieved mainly by inhibition of
fresh CD16 delivery. Inhibitors of 90 kDa heat shock protein
(HSP90), a molecular chaperone found in membrane platforms
together with CD16 and CD11b, significantly increased the observed
effects caused by heat shock. The presented data suggest a novel
systemic aspect of increased temperature which relies on immediate
modification by heat of a neutrophil molecular pattern. This
effect precedes cell death and may be beneficial in the initial
phase of inflammation providing a nonphlogistic signal to
macrophages before it comes from apoptotic cells.
Collapse
|
22
|
Lipke AB, Matute-Bello G, Herrero R, Wong VA, Mongovin SM, Martin TR. Death receptors mediate the adverse effects of febrile-range hyperthermia on the outcome of lipopolysaccharide-induced lung injury. Am J Physiol Lung Cell Mol Physiol 2011; 301:L60-70. [PMID: 21515659 DOI: 10.1152/ajplung.00314.2010] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
We have shown that febrile-range hyperthermia enhances lung injury and mortality in mice exposed to inhaled LPS and is associated with increased TNF-α receptor activity, suppression of NF-κB activity in vitro, and increased apoptosis of alveolar epithelial cells in vivo. We hypothesized that hyperthermia enhances lung injury and mortality in vivo by a mechanism dependent on TNF receptor signaling. To test this, we exposed mice lacking the TNF-receptor family members TNFR1/R2 or Fas (TNFR1/R2(-/-) and lpr) to inhaled LPS with or without febrile-range hyperthermia. For comparison, we studied mice lacking IL-1 receptor activity (IL-1R(-/-)) to determine the role of inflammation on the effect of hyperthermia in vivo. TNFR1/R2(-/-) and lpr mice were protected from augmented alveolar permeability and mortality associated with hyperthermia, whereas IL-1R(-/-) mice were susceptible to augmented alveolar permeability but protected from mortality associated with hyperthermia. Hyperthermia decreased pulmonary concentrations of TNF-α and keratinocyte-derived chemokine after LPS in C57BL/6 mice and did not affect pulmonary inflammation but enhanced circulating markers of oxidative injury and nitric oxide metabolites. The data suggest that hyperthermia enhances lung injury by a mechanism that requires death receptor activity and is not directly associated with changes in inflammation mediated by hyperthermia. In addition, hyperthermia appears to enhance mortality by generating a systemic inflammatory response and not by a mechanism directly associated with respiratory failure. Finally, we observed that exposure to febrile-range hyperthermia converts a modest, survivable model of lung injury into a fatal syndrome associated with oxidative and nitrosative stress, similar to the systemic inflammatory response syndrome.
Collapse
Affiliation(s)
- Anne B Lipke
- Division of Pulmonary and Critical Care Medicine, Puget Sound Medical Center, University of Washington, Seattle, Washington 98108, USA.
| | | | | | | | | | | |
Collapse
|
23
|
Abstract
Species differences in life span have been attributed to cellular survival during various stressors, designated here as 'cell resilience'. In primary fibroblast cultures, cell resilience during exposure to free radicals, hypoglycemia, hyperthermia, and various toxins has shown generally consistent correlations with the species characteristic life spans of birds and mammals. However, the mechanistic links of cell resilience in fibroblast cultures to different species life spans are poorly understood. We propose that certain experimental stressors are relevant to somatic damage in vivo during inflammatory responses of innate immunity, particularly, resistance to reactive oxygen species (ROS), low glucose, and hyperthermia. According to this hypothesis, somatic cell resilience determines species differences in longevity during repeated infections and traumatic injuries in the natural environment. Infections and injury expose local fibroblasts and other cells to ROS generated by macrophages and to local temperature elevations. Systemically, acute phase immune reactions cause hypoglycemia and hyperthermia. We propose that cell resilience to somatic stressors incurred in inflammation is important in the evolution of longevity and that longer-lived species are specifically more resistant to immune-related stressors. This hypothesis further specifies Kirkwood's disposable soma theory. We suggest expanding the battery of stressors and markers used for comparative studies to additional cell types and additional parameters relevant to host defense and to their ecological specificities.
Collapse
Affiliation(s)
- Caleb E Finch
- University of Southern California, Los Angeles, CA 90089-0191, USA.
| | | | | | | |
Collapse
|
24
|
Using in vivo zebrafish models to understand the biochemical basis of neutrophilic respiratory disease. Biochem Soc Trans 2009; 37:830-7. [PMID: 19614603 DOI: 10.1042/bst0370830] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Neutrophilic inflammation in the lung protects against infectious disease, and usually resolves spontaneously after removal of the inflammatory stimulus. However, much lung disease is caused by a failure of resolution of neutrophilic inflammation. Our laboratory is seeking an understanding of the biochemical basis of inflammation resolution, using the zebrafish model system. Zebrafish larvae are transparent, allowing visualization of GFP (green fluorescent protein)-labelled leucocytes during inflammation in vivo, and they can be readily manipulated by a range of forward and reverse genetic techniques. This combination of advantages makes zebrafish a powerful tool for the study of in vivo inflammatory processes. Using this model, we have visualized the process of inflammation resolution in vivo, and identified a role for apoptosis in this process. In addition, we have performed a forward genetic screen for mutants with defective resolution of inflammation, and reverse genetic experiments examining the influence of candidate genes on inflammation resolution. We have established a platform for screening for compounds with anti-inflammatory activity, which has yielded a number of interesting leads. Looking forward to succeed in the future, we are working at combining mutants, transgenes and pharmacological agents to dissect the biochemical basis of inflammation resolution, and to identify compounds that might be used to treat patients with respiratory disease.
Collapse
|