1
|
Effect of Hydrogen on AM Pyroptosis Induced by Severe Burns in Rats. J Pers Med 2023; 13:jpm13030377. [PMID: 36983559 PMCID: PMC10053548 DOI: 10.3390/jpm13030377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 02/06/2023] [Accepted: 02/14/2023] [Indexed: 02/24/2023] Open
Abstract
Background: Hydrogen has anti-inflammatory and antioxidant effects and is beneficial to multiple organs. However, its effect on alveolar macrophage (AM) pyroptosis induced by burns is still unclear. The purpose of this research was to study the possible positive effects of hydrogen on burn-induced lung injury and the effects of hydrogen on AM pyroptosis during acute lung injury (ALI) induced by burns. Methods: In this study, histological changes in rat lungs in vivo were evaluated by micro-CT, and histological changes in isolated lungs were evaluated by hematoxylin and eosin (HE) staining. The expressions of leucine rich repeat (LRR) and pyrin domain (PYD) containing protein 3 (NLRP3), caspase-1 and Gasdermin-D (GSDMD) were analyzed by Western blotting. The expression of GSDMD was measured by immunofluorescence to evaluate the levels of lung inflammation and pyroptosis. The level of inflammation was assessed by enzyme-linked immunosorbent assay (ELISA). Pyroptosis was observed by transmission electron microscopy. Results: We observed that severe burn resulted in increased IL-1β and IL-18, overexpression of NLRP3 and caspase-1 proteins, and pyroptosis in rat lung tissues, as demonstrated by GSDMD overexpression and electron microscopy of AMs. We also observed that hydrogen treatment partially reversed the increase in lung tissue density and reduced pulmonary inflammation. Moreover, hydrogen reduced the HE pathological injury score in the lung tissues of severely burned rats. Hydrogen treatment significantly reduced the contents of IL-1β and IL-18 in the lung tissues and decreased the expression of NLRP3, caspase-1 and GSDMD proteins compared with the burn group. Transmission electron microscopy results also showed that the number of AM membrane pores was significantly reduced in the hydrogen treatment group. Conclusions: The results of this study suggest that hydrogen may protect against ALI induced by burn injury by inhibiting pyroptosis of macrophages via NLRP3.
Collapse
|
2
|
Prospective use of amniotic mesenchymal stem cell metabolite products for tissue regeneration. J Biol Eng 2023; 17:11. [PMID: 36759827 PMCID: PMC9912508 DOI: 10.1186/s13036-023-00331-1] [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: 09/09/2022] [Accepted: 02/06/2023] [Indexed: 02/11/2023] Open
Abstract
Chronic disease can cause tissue and organ damage constituting the largest obstacle to therapy which, in turn, reduces patients' quality-adjusted life-year. Degenerative diseases such as osteoporosis, Alzheimer's disease, Parkinson's disease, and infectious conditions such as hepatitis, cause physical injury to organs. Moreover, damage resulting from chronic conditions such as diabetes can also culminate in the loss of organ function. In these cases, organ transplantation constitutes the therapy of choice, despite the associated problems of immunological rejection, potential disease transmission, and high morbidity rates. Tissue regeneration has the potential to heal or replace tissues and organs damaged by age, disease, or trauma, as well as to treat disabilities. Stem cell use represents an unprecedented strategy for these therapies. However, product availability and mass production remain challenges. A novel therapeutic alternative involving amniotic mesenchymal stem cell metabolite products (AMSC-MP) has been developed using metabolites from stem cells which contain cytokines and growth factors. Its potential role in regenerative therapy has recently been explored, enabling broad pharmacological applications including various gastrointestinal, lung, bladder and renal conditions, as well as the treatment of bone wounds, regeneration and skin aging due to its low immunogenicity and anti-inflammatory effects. The various kinds of growth factors present in AMSC-MP, namely bFGF, VEGF, TGF-β, EGF and KGF, have their respective functions and activities. Each growth factor is formed by different proteins resulting in molecules with various physicochemical properties and levels of stability. This knowledge will assist in the manufacture and application of AMSC-MP as a therapeutic agent.
Collapse
|
3
|
Busanello-Costa M, Renno ACM, de Goes Santos CP, Quintana HT, Martignago CCS, Tim CR, Assis L. Red LED light therapy associated with epidermal growth factor on wound repair process in rats. Lasers Med Sci 2023; 38:36. [PMID: 36626000 DOI: 10.1007/s10103-022-03701-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 12/29/2022] [Indexed: 01/11/2023]
Abstract
Epidermal growth factor (EGF) and light-emitting diode (LED) are currently deployed as promissory treatments for skin repair; however, the mechanisms of their association are not yet evidenced. Thus, the present study aimed to evaluate the effects of combined treatment with EGF and red LED on the wound healing processes in rats. Adult Wistar rats were randomized in control group (CG) wounds without treatment; wounds submitted to EGF treatment (EGF); wounds submitted to LED treatment (LED); wounds submitted to EGF associated with LED treatments (EGF/LED). Treatments were performed immediately after the surgical procedure and each 24 h, totaling 8 sessions. Moreover, LED was applied before EGF treatment at a single point in the center of the wound. Morphological characteristics and the immunoexpression of COX-2, VEGF, and TGF-β were measured. The results demonstrated that EGF/LED group presented a higher wound healing index. Additionally, all experimental groups presented similar findings in the histological evaluation, the degree of inflammation, and the area of dermis-like tissue. However, for EGF-treated animals (with or without LED), neoepithelial length was higher. Furthermore, all the treated groups decreased COX-2 and increased VEGF immunoexpression, and only EGF/LED group enhanced the TGF-β protein expression when compared to the untreated group. This research shows that EGF and LED modulate inflammatory process and increase the vascularity. In addition, treatment of EGF associated with LED promoted a more evident positive effect for increasing TGF-β expression and may be promising resources in the clinical treatment of cutaneous wounds.
Collapse
Affiliation(s)
- Márcia Busanello-Costa
- Department of Biosciences, Federal University of São Paulo, 136, Silva Jardim Street, Santos, SP, 11015-020, Brazil
| | - Ana Claudia Muniz Renno
- Department of Biosciences, Federal University of São Paulo, 136, Silva Jardim Street, Santos, SP, 11015-020, Brazil
| | | | - Hananiah Tardivo Quintana
- Department of Biosciences, Federal University of São Paulo, 136, Silva Jardim Street, Santos, SP, 11015-020, Brazil
| | - Cintia Cristina Santi Martignago
- Department of Biosciences, Federal University of São Paulo, 136, Silva Jardim Street, Santos, SP, 11015-020, Brazil
- Department of Physiotherapy, Federal University of São Carlos, São Carlos, SP, Brazil
| | - Carla Roberta Tim
- Scientific Institute and Technological Department-University Brazil, São Paulo-Itaquera, SP, Brazil
| | - Lívia Assis
- Department of Biosciences, Federal University of São Paulo, 136, Silva Jardim Street, Santos, SP, 11015-020, Brazil.
- Scientific Institute and Technological Department-University Brazil, São Paulo-Itaquera, SP, Brazil.
| |
Collapse
|
4
|
Moghbeli K, Valenzi E, Naramore R, Sembrat JC, Chen K, Rojas MM, Wenzel SE, Lafyatis R, Modena B, Weathington NM. β-Agonist exposure preferentially impacts lung macrophage cyclic AMP-related gene expression in asthma and asthma COPD overlap syndrome. Am J Physiol Lung Cell Mol Physiol 2021; 321:L837-L843. [PMID: 34494468 DOI: 10.1152/ajplung.00260.2021] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Bronchoalveolar lavage (BAL) samples from Severe Asthma Research Program (SARP) patients display suppression of a module of genes involved in cAMP-signaling pathways (BALcAMP) correlating with severity, therapy, and macrophage constituency. We sought to establish if gene expression changes were specific to macrophages and compared gene expression trends from multiple sources. Datasets included single-cell RNA sequencing (scRNA-seq) from lung specimens including a fatal exacerbation of severe Asthma COPD Overlap Syndrome (ACOS) after intense therapy and controls without lung disease, bulk RNA sequencing from cultured macrophage (THP-1) cells after acute or prolonged β-agonist exposure, SARP datasets, and data from the Immune Modulators of Severe Asthma (IMSA) cohort. THP monocytes suppressed BALcAMP network gene expression after prolonged relative to acute β-agonist exposure, corroborating SARP observations. scRNA-seq from healthy and diseased lung tissue revealed 13 cell populations enriched for macrophages. In severe ACOS, BALcAMP gene network expression scores were decreased in many cell populations, most significantly for macrophage populations (P < 3.9e-111). Natural killer (NK) cells and type II alveolar epithelial cells displayed less robust network suppression (P < 9.2e-8). Alveolar macrophages displayed the most numerous individual genes affected and the highest amplitude of modulation. Key BALcAMP genes demonstrate significantly decreased expression in severe asthmatics in the IMSA cohort. We conclude that suppression of the BALcAMP gene module identified from SARP BAL samples is validated in the IMSA patient cohort with physiological parallels observed in a monocytic cell line and in a severe ACOS patient sample with effects preferentially localizing to macrophages.
Collapse
Affiliation(s)
- Kaveh Moghbeli
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Eleanor Valenzi
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Rachel Naramore
- Division of General Internal Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - John Charles Sembrat
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Kong Chen
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Mauricio M Rojas
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Ohio State University, Columbus, Ohio
| | - Sally E Wenzel
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania.,Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Robert Lafyatis
- Division of Rheumatology and Clinical Immunology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Brian Modena
- Allergy & Rheumatology Medical Clinic, Scripps Research Institute, La Jolla, California
| | - Nathaniel M Weathington
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
5
|
Adiliaghdam F, Cavallaro P, Mohad V, Almpani M, Kühn F, Gharedaghi MH, Najibi M, Rahme LG, Hodin RA. Targeting the gut to prevent sepsis from a cutaneous burn. JCI Insight 2020; 5:137128. [PMID: 33004693 PMCID: PMC7566703 DOI: 10.1172/jci.insight.137128] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 08/26/2020] [Indexed: 12/13/2022] Open
Abstract
Severe burn injury induces gut barrier dysfunction and subsequently a profound systemic inflammatory response. In the present study, we examined the role of the small intestinal brush border enzyme, intestinal alkaline phosphatase (IAP), in preserving gut barrier function and preventing systemic inflammation after burn wound infection in mice. Mice were subjected to a 30% total body surface area dorsal burn with or without intradermal injection of Pseudomonas aeruginosa. Mice were gavaged with 2000 units of IAP or vehicle at 3 and 12 hours after the insult. We found that both endogenously produced and exogenously supplemented IAP significantly reduced gut barrier damage, decreased bacterial translocation to the systemic organs, attenuated systemic inflammation, and improved survival in this burn wound infection model. IAP attenuated liver inflammation and reduced the proinflammatory characteristics of portal serum. Furthermore, we found that intestinal luminal contents of burn wound-infected mice negatively impacted the intestinal epithelial integrity compared with luminal contents of control mice and that IAP supplementation preserved monolayer integrity. These results indicate that oral IAP therapy may represent an approach to preserving gut barrier function, blocking proinflammatory triggers from entering the portal system, preventing gut-induced systemic inflammation, and improving survival after severe burn injuries.
Collapse
Affiliation(s)
- Fatemeh Adiliaghdam
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Paul Cavallaro
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Vidisha Mohad
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Marianna Almpani
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Shriners Hospital for Children, Boston, Massachusetts, USA
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts, USA
| | - Florian Kühn
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of General, Visceral and Transplant Surgery, Hospital of the University of Munich, Munich, Germany
| | - Mohammad Hadi Gharedaghi
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Mehran Najibi
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Laurence G. Rahme
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Shriners Hospital for Children, Boston, Massachusetts, USA
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts, USA
| | - Richard A. Hodin
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
6
|
Liu JS, Du J, Cheng X, Zhang XZ, Li Y, Chen XL. Exosomal miR-451 from human umbilical cord mesenchymal stem cells attenuates burn-induced acute lung injury. J Chin Med Assoc 2019; 82:895-901. [PMID: 31800531 DOI: 10.1097/jcma.0000000000000189] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The aim of this study was to investigate the molecular mechanism of human umbilical cord mesenchymal stem cells (MSCs)-derived exosomes (hUCMSC-Exos) in regulating burn-induced acute lung injury (ALI). METHODS In this study, we initially isolated exosomes from hUCMSCs and identified them by transmission electron microscopy. The expression of the protein markers CD9 and CD63 in the exosomes was determined by western blot analysis. The expression of miR-451 in the hUCMSC-Exos was determined by qRT-PCR. The levels of TNF-α, IL-1β, and IL-6 in lung tissues and serum as well as the levels of malondialdehyde, myeloperoxidase, superoxide dismutase in lung tissues were detected by ELISA. Hematoxylin-eosin stain was used to observe the morphological changes of lung tissues after burn. Terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling assays were performed to detect apoptosis in lung tissues after burn. The expression of proteins related to the Toll-like receptor 4 (TLR4)/NF-κB signaling pathway in lung tissues after burn was analyzed by western blotting. RESULTS Our results showed that hUCMSC-Exos successfully decreased TNF-α, IL-1β, and IL-6 levels in rats after burn, and this reduction was reversed when the miR-451 expression in the hUCMSC-Exo group was inhibited. HUCMSC-Exo-derived miR-451 improves ALI via the TLR4/NF-κB pathway. CONCLUSION We demonstrated that exosomes derived from hUCMSCs mediate miR-451 to attenuate burn-induced ALI.
Collapse
Affiliation(s)
- Ji-Song Liu
- Department of Burns, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Juan Du
- Department of Minimally Invasive Surgery, Thirds Hospital of Bengbu, Bengbu, Anhui, China
| | - Xiu Cheng
- School of Pharmacy, BengBu Medical College, Bengbu, Anhui, China
| | - Xiang-Zhou Zhang
- Department of Burn and Plastic Surgery, Thirds Hospital of Bengbu, Bengbu, Anhui, China
| | - Yong Li
- Department of Burn and Plastic Surgery, Thirds Hospital of Bengbu, Bengbu, Anhui, China
| | - Xu-Lin Chen
- Department of Burns, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
7
|
Weathington N, O’Brien ME, Radder J, Whisenant TC, Bleecker ER, Busse WW, Erzurum SC, Gaston B, Hastie AT, Jarjour NN, Meyers DA, Milosevic J, Moore WC, Tedrow JR, Trudeau JB, Wong HP, Wu W, Kaminski N, Wenzel SE, Modena BD. BAL Cell Gene Expression in Severe Asthma Reveals Mechanisms of Severe Disease and Influences of Medications. Am J Respir Crit Care Med 2019; 200:837-856. [PMID: 31161938 PMCID: PMC6812436 DOI: 10.1164/rccm.201811-2221oc] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 06/03/2019] [Indexed: 01/16/2023] Open
Abstract
Rationale: Gene expression of BAL cells, which samples the cellular milieu within the lower respiratory tract, has not been well studied in severe asthma.Objectives: To identify new biomolecular mechanisms underlying severe asthma by an unbiased, detailed interrogation of global gene expression.Methods: BAL cell expression was profiled in 154 asthma and control subjects. Of these participants, 100 had accompanying airway epithelial cell gene expression. BAL cell expression profiles were related to participant (age, sex, race, and medication) and sample traits (cell proportions), and then severity-related gene expression determined by correlating transcripts and coexpression networks to lung function, emergency department visits or hospitalizations in the last year, medication use, and quality-of-life scores.Measurements and Main Results: Age, sex, race, cell proportions, and medications strongly influenced BAL cell gene expression, but leading severity-related genes could be determined by carefully identifying and accounting for these influences. A BAL cell expression network enriched for cAMP signaling components most differentiated subjects with severe asthma from other subjects. Subsequently, an in vitro cellular model showed this phenomenon was likely caused by a robust upregulation in cAMP-related expression in nonsevere and β-agonist-naive subjects given a β-agonist before cell collection. Interestingly, ELISAs performed on BAL lysates showed protein levels may partly disagree with expression changes.Conclusions: Gene expression in BAL cells is influenced by factors seldomly considered. Notably, β-agonist exposure likely had a strong and immediate impact on cellular gene expression, which may not translate to important disease mechanisms or necessarily match protein levels. Leading severity-related genes were discovered in an unbiased, system-wide analysis, revealing new targets that map to asthma susceptibility loci.
Collapse
Affiliation(s)
- Nathaniel Weathington
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Michael E. O’Brien
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Josiah Radder
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Thomas C. Whisenant
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California
| | - Eugene R. Bleecker
- Division of Genetics, Genomics and Precision Medicine, University of Arizona, Tucson, Arizona
| | - William W. Busse
- Division of Allergy, Pulmonary, and Critical Care Medicine, University of Wisconsin, Madison, Wisconsin
| | - Serpil C. Erzurum
- Lerner Research Institute, Respiratory Institute, Cleveland Clinic, Cleveland, Ohio
| | - Benjamin Gaston
- Division of Pediatric Pulmonary, Allergy and Immunology, Case Western Reserve University and Rainbow Babies Children’s Hospital, Cleveland, Ohio
| | - Annette T. Hastie
- Section on Pulmonary, Critical Care, Allergy and Immunologic Diseases, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Nizar N. Jarjour
- Division of Allergy, Pulmonary, and Critical Care Medicine, University of Wisconsin, Madison, Wisconsin
| | - Deborah A. Meyers
- Division of Genetics, Genomics and Precision Medicine, University of Arizona, Tucson, Arizona
| | - Jadranka Milosevic
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Wendy C. Moore
- Section on Pulmonary, Critical Care, Allergy and Immunologic Diseases, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - John R. Tedrow
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - John B. Trudeau
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Hesper P. Wong
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Wei Wu
- Computational Biology Department, Carnegie Mellon University, Pittsburgh, Pennsylvania
| | - Naftali Kaminski
- Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Sally E. Wenzel
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Department of Environmental and Occupational Health, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania; and
| | - Brian D. Modena
- Division of Allergy, National Jewish Hospital, Denver, Colorado
| |
Collapse
|
8
|
Su Y, Luo H, Yang J. Heparin-binding EGF-like growth factor attenuates lung inflammation and injury in a murine model of pulmonary emphysema. Growth Factors 2018; 36:246-262. [PMID: 30600734 DOI: 10.1080/08977194.2018.1552270] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Pulmonary inflammation and progressive lung destruction are the major causes of chronic obstructive pulmonary disease (COPD), resulting in emphysema and irreversible pulmonary dysfunction. Heparin-binding EGF-like growth factor (HB-EGF), is known to play a protective role in the process of various inflammatory diseases. However, its effect on COPD is poorly understood. This study was designed to determine the effect of HB-EGF on lung inflammation and injury in a murine model of pulmonary emphysema. HB-EGF promoted percent survival and body weight, attenuated lung injury, inflammatory cells, and cytokines infiltration, and prevented lung function decline. Additionally, treatment of rHB-EGF suppressed the nuclear translocation of nuclear factor κB (NF-κB)/p65, decreased TUNEL-positive cells and the expression of caspase 3, and increased the expression of PCNA, HB-EGF, and EGF receptor (EGFR). We conclude that HB-EGF attenuates lung inflammation and injury, probably through the activation of EGFR, followed by suppression of NF-ΚB signalling, promotion of cell proliferation, and inhibition of apoptosis.
Collapse
Affiliation(s)
- Yanwei Su
- a School of Nursing, Tongji Medical College , Huazhong University of Science and Technology , Wuhan , China
| | - Heng Luo
- b Department of Pathology, Union Hospital, Tongji Medical College , Huazhong University of Science and Technology , Wuhan , China
| | - Jixin Yang
- c Department of Pediatric Surgery, Tongji Hospital, Tongji Medical College , Huazhong University of Science and Technology , Wuhan , China
| |
Collapse
|
9
|
Bai RX, Xu YY, Qin G, Chen YM, Wang HF, Wang M, Du SY. Repression of TXNIP-NLRP3 axis restores intestinal barrier function via inhibition of myeloperoxidase activity and oxidative stress in nonalcoholic steatohepatitis. J Cell Physiol 2018; 234:7524-7538. [PMID: 30387131 DOI: 10.1002/jcp.27513] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2018] [Accepted: 09/10/2018] [Indexed: 12/13/2022]
Abstract
Dysfunction of the intestinal barrier function occurs in hepatic injury, but the specific mechanisms responsible are largely unknown. Recently, NOD-like receptor 3 (NLRP3) inflammasome functions in impairing endothelial barrier function. In this study, we test the hypothesis that TXNIP-NLRP3 axis repression prevents against intestinal barrier function disruption in nonalcoholic steatohepatitis (NASH). First, lipopolysaccharide (LPS)-induced alterations in expression of ZO-1 and occludin, myeloperoxidase (MPO) activity, reactive oxygen species (ROS) level, and transepithelial electric resistance (TEER) in intestinal epithelial cells (IECs) isolated from C57BL/6 wild-type (WT) and TXNIP-/- mice were evaluated. The underlying regulatory mechanisms of TXNIP knockout in vivo were investigated with the detection of expressions of TXNIP, NLRP3 and ZO-1, and occludin, the interaction of TXNIP-NLRP3, MPO activity, ROS level, permeability of intestinal mucosa, levels of inflammatory factors in serum, and LPS concentration. We identified that TXNIP knockout promoted ZO-1 and occludin expression, yet reduced MPO activity, ROS level, and cell permeability in IECs, indicating restored the intestinal barrier function. However, LPS upregulated TXNIP and NLRP3 expression, as well as contributed to the interaction between TXNIP and NLRP3 in vitro. Furthermore, TXNIP was significantly upregulated in the intestinal mucosa of NASH mice and its knockout repaired the intestinal barrier disrupt, inhibited expression of inflammatory factors, and reduced LPS concentration as well as hepatic injury in vivo. Taken together, our findings demonstrated that inhibited the activation of the TXNIP-NLRP3 axis reduced MPO activity and oxidative stress and thus restoring the intestinal barrier function in NASH. TXNIP-NLRP3 axis may be a promising therapeutic strategy for the NASH treatment.
Collapse
Affiliation(s)
- Ru-Xue Bai
- Department of Gastroenterology, China-Japan Friendship Hospital, Beijing, China
| | - Ying-Ying Xu
- Department of Gastroenterology, China-Japan Friendship Hospital, Beijing, China
| | - Geng Qin
- Department of Gastroenterology, China-Japan Friendship Hospital, Beijing, China
| | - Yan-Ming Chen
- Department of Gastroenterology, China-Japan Friendship Hospital, Beijing, China
| | - Hui-Fen Wang
- Department of Gastroenterology, China-Japan Friendship Hospital, Beijing, China
| | - Miao Wang
- Department of Gastroenterology, China-Japan Friendship Hospital, Beijing, China
| | - Shi-Yu Du
- Department of Gastroenterology, China-Japan Friendship Hospital, Beijing, China
| |
Collapse
|
10
|
Jacob S, Herndon DN, Hawkins HK, Enkhbaatar P, Cox RA. Xanthine oxidase contributes to sustained airway epithelial oxidative stress after scald burn. INTERNATIONAL JOURNAL OF BURNS AND TRAUMA 2017; 7:98-106. [PMID: 29119062 PMCID: PMC5665841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 09/28/2017] [Indexed: 06/07/2023]
Abstract
Respiratory tract infections and pneumonia are major causes of morbidity and mortality in burn victims, however, limited studies have examined the effects of burn injury on airway epithelium. The current study examines the effect of scald burn injury on rat tracheal epithelium at 5 days after injury and tests the hypothesis that treatment with febuxostat (FBX), an inhibitor of xanthine oxidase (XO), can be protective of cell homeostasis. Sprague Dawley rats were randomly divided into uninjured (sham), injured (control) and injured and FBX treated groups, n = 8. Control and FBX treated groups received 60% total body surface area scald burn injury. The FBX group received an i. p. dose (1 mg/kg) at 1 hour after injury and every 24 hours. At 5 days after injury, the animals were sacrificed and tracheal epithelial cell lysates were collected. Malondialdehyde (MDA), ATP, and XO activity were measured. Formation of 8-OHdG in tracheal epithelium was determined using immunohistochemistry (IHC) and immunoreactivity was quantitated. MDA levels were significantly increased in injured control animals (24.8 ± 2.3) compared to sham (7.93 ± 1.2, p = 0.002). FBX treatment attenuated this response (12.6 ± 2.7, p = 0.02). ATP levels were significantly decreased in control (0.7 ± 0.16) compared to sham, (2 ± 0.14, p = 0.01). ATP levels were increased with FBX treatment (1.8 ± 0.1, p = 0.03) compared to controls. There was a significant increase in XO activity in control animals, 1.04 ± 0.06 compared to sham (0.34 ± 0.05, p = 0.03), and this response decreased with FBX treatment 0.46 ± 0.07 (p = 0.04). Immunolabeling of 8-OHdG in control animals was significantly increased (25.1 ± 0.7 compared to the sham group 5.5 ± 1.9 (p = 0.01)), and was decreased with FBX treatment (7.0 ± 2.3 compared to control (p = 0.03)). The current study indicates that lipid peroxidation and ATP depletion persist in tracheal epithelium for 5 days after injury along with increased XO activity and 8-OHdG. These effects were significantly attenuated by FBX treatment, suggesting that reactive oxygen species generated by XO contribute to airway epithelial injury following scald burn.
Collapse
Affiliation(s)
- Sam Jacob
- Department of Pathology, Shriners Hospitals for Children and The University of Texas Medical BranchGalveston, Texas, USA
| | - David N Herndon
- Department of Surgery, Shriners Hospitals for Children and The University of Texas Medical BranchGalveston, Texas, USA
| | - Hal K Hawkins
- Department of Pathology, Shriners Hospitals for Children and The University of Texas Medical BranchGalveston, Texas, USA
| | - Perenlei Enkhbaatar
- Department of Anesthesiology, Shriners Hospitals for Children and The University of Texas Medical BranchGalveston, Texas, USA
| | - Robert A Cox
- Department of Pathology, Shriners Hospitals for Children and The University of Texas Medical BranchGalveston, Texas, USA
| |
Collapse
|
11
|
Wang T, Gross C, Desai AA, Zemskov E, Wu X, Garcia AN, Jacobson JR, Yuan JXJ, Garcia JGN, Black SM. Endothelial cell signaling and ventilator-induced lung injury: molecular mechanisms, genomic analyses, and therapeutic targets. Am J Physiol Lung Cell Mol Physiol 2016; 312:L452-L476. [PMID: 27979857 DOI: 10.1152/ajplung.00231.2016] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 12/08/2016] [Accepted: 12/11/2016] [Indexed: 12/13/2022] Open
Abstract
Mechanical ventilation is a life-saving intervention in critically ill patients with respiratory failure due to acute respiratory distress syndrome (ARDS). Paradoxically, mechanical ventilation also creates excessive mechanical stress that directly augments lung injury, a syndrome known as ventilator-induced lung injury (VILI). The pathobiology of VILI and ARDS shares many inflammatory features including increases in lung vascular permeability due to loss of endothelial cell barrier integrity resulting in alveolar flooding. While there have been advances in the understanding of certain elements of VILI and ARDS pathobiology, such as defining the importance of lung inflammatory leukocyte infiltration and highly induced cytokine expression, a deep understanding of the initiating and regulatory pathways involved in these inflammatory responses remains poorly understood. Prevailing evidence indicates that loss of endothelial barrier function plays a primary role in the development of VILI and ARDS. Thus this review will focus on the latest knowledge related to 1) the key role of the endothelium in the pathogenesis of VILI; 2) the transcription factors that relay the effects of excessive mechanical stress in the endothelium; 3) the mechanical stress-induced posttranslational modifications that influence key signaling pathways involved in VILI responses in the endothelium; 4) the genetic and epigenetic regulation of key target genes in the endothelium that are involved in VILI responses; and 5) the need for novel therapeutic strategies for VILI that can preserve endothelial barrier function.
Collapse
Affiliation(s)
- Ting Wang
- Department of Medicine, The University of Arizona Health Sciences, Tucson, Arizona
| | - Christine Gross
- Vascular Biology Center, Augusta University, Augusta, Georgia
| | - Ankit A Desai
- Department of Medicine, The University of Arizona Health Sciences, Tucson, Arizona
| | - Evgeny Zemskov
- Department of Medicine, The University of Arizona Health Sciences, Tucson, Arizona
| | - Xiaomin Wu
- Department of Medicine, The University of Arizona Health Sciences, Tucson, Arizona
| | - Alexander N Garcia
- Department of Pharmacology University of Illinois at Chicago, Chicago, Illinois; and
| | - Jeffrey R Jacobson
- Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Jason X-J Yuan
- Department of Medicine, The University of Arizona Health Sciences, Tucson, Arizona
| | - Joe G N Garcia
- Department of Medicine, The University of Arizona Health Sciences, Tucson, Arizona
| | - Stephen M Black
- Department of Medicine, The University of Arizona Health Sciences, Tucson, Arizona;
| |
Collapse
|
12
|
Shults JA, Curtis BJ, Boe DM, Ramirez L, Kovacs EJ. Ethanol intoxication prolongs post-burn pulmonary inflammation: role of alveolar macrophages. J Leukoc Biol 2016; 100:1037-1045. [PMID: 27531926 DOI: 10.1189/jlb.3ma0316-111r] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 07/26/2016] [Indexed: 02/06/2023] Open
Abstract
In this study, the role and fate of AMs were examined in pulmonary inflammation after intoxication and injury. Clinical evidence has revealed that half of all burn patients brought to the emergency department are intoxicated at the time of injury. This combined insult results in amplified neutrophil accumulation and pulmonary edema, with an increased risk of lung failure and mortality, relative to either insult alone. We believe that this excessive pulmonary inflammation, which also parallels decreased lung function, is mediated in part by AMs. Restoration of lung tissue homeostasis is dependent on the eradication of neutrophils and removal of apoptotic cells, both major functions of AMs. Thirty minutes after binge ethanol intoxication, mice were anesthetized and given a 15% total body surface area dorsal scald injury. At 24 h, we found a 50% decrease in the total number of AMs (P < 0.05) and observed a proinflammatory phenotype on the remaining lung AMs. Loss of AMs paralleled a 6-fold increase in the number of TUNEL+ lung apoptotic cells (P < 0.05) and a 3.5-fold increase in the percentage of annexin V+ apoptotic cells in BAL (P < 0.05), after intoxication and injury, relative to controls. In contrast to the reduction in the number of cells, AMs from intoxicated and injured mice had a 4-fold increase in efferocytosis (P < 0.05). In summary, these data suggest that loss of AMs may delay resolution of inflammation, resulting in the pulmonary complications and elevated mortality rates observed in intoxicated and burn-injured patients.
Collapse
Affiliation(s)
- Jill A Shults
- Alcohol Research Program, Loyola University Chicago, Health Sciences Campus, Stritch School of Medicine, Maywood, Illinois, USA.,Burn and Shock Trauma Research Institute, Loyola University Chicago, Health Sciences Campus, Stritch School of Medicine, Maywood, Illinois, USA.,Department of Surgery, Loyola University Chicago, Health Sciences Campus, Stritch School of Medicine, Maywood, Illinois, USA.,Integrative Cell Biology Program, Loyola University Chicago, Health Sciences Campus, Stritch School of Medicine, Maywood, Illinois, USA
| | - Brenda J Curtis
- Alcohol Research Program, Loyola University Chicago, Health Sciences Campus, Stritch School of Medicine, Maywood, Illinois, USA.,Burn and Shock Trauma Research Institute, Loyola University Chicago, Health Sciences Campus, Stritch School of Medicine, Maywood, Illinois, USA.,Department of Surgery, Loyola University Chicago, Health Sciences Campus, Stritch School of Medicine, Maywood, Illinois, USA
| | - Devin M Boe
- Alcohol Research Program, Loyola University Chicago, Health Sciences Campus, Stritch School of Medicine, Maywood, Illinois, USA.,Burn and Shock Trauma Research Institute, Loyola University Chicago, Health Sciences Campus, Stritch School of Medicine, Maywood, Illinois, USA.,Department of Surgery, Loyola University Chicago, Health Sciences Campus, Stritch School of Medicine, Maywood, Illinois, USA.,Integrative Cell Biology Program, Loyola University Chicago, Health Sciences Campus, Stritch School of Medicine, Maywood, Illinois, USA
| | - Luis Ramirez
- Alcohol Research Program, Loyola University Chicago, Health Sciences Campus, Stritch School of Medicine, Maywood, Illinois, USA.,Burn and Shock Trauma Research Institute, Loyola University Chicago, Health Sciences Campus, Stritch School of Medicine, Maywood, Illinois, USA.,Department of Surgery, Loyola University Chicago, Health Sciences Campus, Stritch School of Medicine, Maywood, Illinois, USA
| | - Elizabeth J Kovacs
- Alcohol Research Program, Loyola University Chicago, Health Sciences Campus, Stritch School of Medicine, Maywood, Illinois, USA; .,Burn and Shock Trauma Research Institute, Loyola University Chicago, Health Sciences Campus, Stritch School of Medicine, Maywood, Illinois, USA.,Department of Surgery, Loyola University Chicago, Health Sciences Campus, Stritch School of Medicine, Maywood, Illinois, USA.,Integrative Cell Biology Program, Loyola University Chicago, Health Sciences Campus, Stritch School of Medicine, Maywood, Illinois, USA
| |
Collapse
|
13
|
Zhu J, Yang F, He F, Tian X, Tang S, Chen X. A tubular gelatin scaffold capable of the time-dependent controlled release of epidermal growth factor and mitomycin C. Colloids Surf B Biointerfaces 2015; 135:416-424. [DOI: 10.1016/j.colsurfb.2015.06.049] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2015] [Revised: 05/24/2015] [Accepted: 06/24/2015] [Indexed: 10/23/2022]
|
14
|
Ménoret A, Crocker SJ, Rodriguez A, Rathinam VA, Clark RB, Vella AT. Transition from identity to bioactivity-guided proteomics for biomarker discovery with focus on the PF2D platform. Proteomics Clin Appl 2015. [PMID: 26201056 DOI: 10.1002/prca.201500029] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Proteomic strategies provide a valuable tool kit to identify proteins involved in diseases. With recent progress in MS technology, high throughput proteomics has accelerated protein identification for potential biomarkers. Numerous biomarker candidates have been identified in several diseases, and many are common among pathologies. An overall strategy that could complement and strengthen the search for biomarkers is combining protein identity with biological outcomes. This review describes an emerging framework of bridging bioactivity to protein identity, exploring the possibility that some biomarkers will have a mechanistic role in the disease process. A review of pulmonary, cardiovascular, and CNS biomarkers will be discussed to demonstrate the utility of combining bioactivity with identification as a means to not only find meaningful biomarkers, but also to uncover functional mediators of disease.
Collapse
Affiliation(s)
- Antoine Ménoret
- Department of Immunology, University of Connecticut Health Center, Farmington, CT, USA
| | - Stephen J Crocker
- Department of Neuroscience, University of Connecticut Health Center, Farmington, CT, USA
| | - Annabelle Rodriguez
- Department of Cell Biology, University of Connecticut Health Center, Farmington, CT, USA
| | - Vijay A Rathinam
- Department of Immunology, University of Connecticut Health Center, Farmington, CT, USA
| | - Robert B Clark
- Department of Immunology, University of Connecticut Health Center, Farmington, CT, USA
| | - Anthony T Vella
- Department of Immunology, University of Connecticut Health Center, Farmington, CT, USA
| |
Collapse
|
15
|
Ribeiro A, Almeida VI, Costola-de-Souza C, Ferraz-de-Paula V, Pinheiro ML, Vitoretti LB, Gimenes-Junior JA, Akamine AT, Crippa JA, Tavares-de-Lima W, Palermo-Neto J. Cannabidiol improves lung function and inflammation in mice submitted to LPS-induced acute lung injury. Immunopharmacol Immunotoxicol 2014; 37:35-41. [DOI: 10.3109/08923973.2014.976794] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|