1
|
Ashina M, Kido T, Shirai K, Masuda Y, Imagawa Y, Nakata Y, Nozu K, Fujioka K. A neonatal mouse model of meconium peritonitis generated using human meconium slurry. Pediatr Res 2024:10.1038/s41390-024-03470-3. [PMID: 39169223 DOI: 10.1038/s41390-024-03470-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 05/16/2024] [Accepted: 07/15/2024] [Indexed: 08/23/2024]
Abstract
BACKGROUND Meconium peritonitis is a noninfectious chemical peritonitis that occurs following fetal intestinal perforation and leakage of meconium into the abdominal cavity. Because of the lack of appropriate animal models, its pathophysiology has not yet been elucidated. We aimed to create a neonatal mouse model of meconium peritonitis using human meconium slurry (MS). METHODS A stock MS solution prepared from fresh meconium obtained from healthy term infants was administered intraperitoneally to 4-d-old newborn mice. An MS LD40 was then administered, and changes in body weight, hematology, serum biochemistry, and immunomodulatory gene expression were determined. The MS was subjected to antibiotic treatment and heat inactivation to validate the content. Finally, comparisons with nonsurgical neonatal sepsis mouse models were performed. RESULTS Dose-dependent mortality rates were observed, with an LD40 of 200 µL/body weight established. Substantial hematological and hepatorenal abnormalities and increased inflammatory gene expression were observed. Although antibiotic treatment was ineffective, the survival rate was improved by enzymatic inactivation of MS. Importantly, the systemic responses to MS were distinct from those observed in neonatal sepsis model mice. CONCLUSION The MS model closely reflects the pathology of human neonatal meconium peritonitis and maybe useful in research elucidating the pathophysiology of this condition. IMPACT In this study, we generated a neonatal mouse model of meconium peritonitis through intraperitoneal administration of human meconium slurry. We clarified that the pathogenic agent in meconium slurry is mainly a digestive enzyme, and that the systemic responses elicited by meconium slurry were distinct from those in a neonatal sepsis mouse model. As our mouse model is simple and highly reproducible, it is useful for elucidating the pathophysiology of meconium peritonitis.
Collapse
Affiliation(s)
- Mariko Ashina
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Takumi Kido
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Keisuke Shirai
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yu Masuda
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yukihito Imagawa
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yuki Nakata
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Kandai Nozu
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Kazumichi Fujioka
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan.
| |
Collapse
|
2
|
Nofi CP, Prince JM, Aziz M, Wang P. The Novel MFG-E8-derived Oligopeptide, MOP3, Improves Outcomes in a Preclinical Murine Model of Neonatal Sepsis. J Pediatr Surg 2024; 59:1282-1290. [PMID: 38582704 DOI: 10.1016/j.jpedsurg.2024.03.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 03/04/2024] [Indexed: 04/08/2024]
Abstract
INTRODUCTION Neonatal sepsis is a devastating inflammatory condition that remains a leading cause of morbidity and mortality. Milk fat globule-EGF-factor VIII (MFG-E8) is a glycoprotein that reduces inflammation, whereas extracellular cold-inducible RNA binding protein (eCIRP) worsens inflammation. This study aimed to determine the therapeutic potential of a novel MFG-E8-derived oligopeptide 3 (MOP3) designed to clear eCIRP and protect against inflammation, organ injury, and mortality in neonatal sepsis. METHODS C57BL6 mouse pups were injected intraperitoneally with cecal slurry (CS) and treated with MOP3 (20 μg/g) or vehicle. 10 h after injection, blood, lungs, and intestines were collected for analyses, and in a 7-day experiment, pups were monitored for differences in mortality. RESULTS MOP3 treatment protected septic pups from inflammation by reducing eCIRP, IL-6, TNFα, and LDH. MOP3 reduced lung and intestinal inflammation and injury as assessed by reductions in tissue mRNA levels of inflammatory markers, histopathologic injury, and apoptosis in lung and intestines. MOP3 also significantly improved 7-day overall survival for CS-septic mouse pups compared to vehicle (75% vs. 46%, respectively). CONCLUSION Deriving from MFG-E8 and designed to clear eCIRP, MOP3 protects against sepsis-induced inflammation, organ injury, and mortality in a preclinical model of neonatal sepsis, implicating it as an exciting potential new therapeutic. LEVEL OF EVIDENCE Level 1.
Collapse
Affiliation(s)
- Colleen P Nofi
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, USA; Elmezzi Graduate School of Molecular Medicine, Manhasset, NY, USA; Department of Surgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, USA.
| | - Jose M Prince
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, USA; Department of Surgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, USA
| | - Monowar Aziz
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, USA; Elmezzi Graduate School of Molecular Medicine, Manhasset, NY, USA; Department of Surgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, USA; Department of Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, USA
| | - Ping Wang
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, USA; Elmezzi Graduate School of Molecular Medicine, Manhasset, NY, USA; Department of Surgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, USA; Department of Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, USA
| |
Collapse
|
3
|
Odum JD, Akhter J, Verma V, Vollmer G, Davidson A, Hyndman KA, Bolisetty S. Myeloid-specific ferritin light chain deletion does not exacerbate sepsis-associated AKI. Am J Physiol Renal Physiol 2024; 327:F171-F183. [PMID: 38779751 DOI: 10.1152/ajprenal.00043.2024] [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: 02/08/2024] [Revised: 04/29/2024] [Accepted: 05/10/2024] [Indexed: 05/25/2024] Open
Abstract
Sepsis-associated acute kidney injury (SA-AKI) is a key contributor to the life-threatening sequelae attributed to sepsis. Mechanistically, SA-AKI is a consequence of unabated myeloid cell activation and oxidative stress that induces tubular injury. Iron mediates inflammatory pathways directly and through regulating the expression of myeloid-derived ferritin, an iron storage protein comprising ferritin light (FtL) and ferritin heavy chain (FtH) subunits. Previous work revealed that myeloid FtH deletion leads to a compensatory increase in intracellular and circulating FtL and is associated with amelioration of SA-AKI. We designed this study to test the hypothesis that loss of myeloid FtL and subsequently, circulating FtL will exacerbate the sepsis-induced inflammatory response and worsen SA-AKI. We generated a novel myeloid-specific FtL knockout mouse (FtLLysM-/-) and induced sepsis via cecal ligation and puncture or lipopolysaccharide endotoxemia. As expected, serum ferritin levels were significantly lower in the knockout mice, suggesting that myeloid cells dominantly contribute to circulating ferritin. Interestingly, although sepsis induction led to a marked production of pro- and anti-inflammatory cytokines, there was no statistical difference between the genotypes. There was a similar loss of kidney function, as evidenced by a rise in serum creatinine and cystatin C and renal injury identified by expression of kidney injury molecule-1 and neutrophil gelatinase-associated lipocalin. Finally, RNA sequencing revealed upregulation of pathways for cell cycle arrest and autophagy postsepsis, but no significant differences were observed between genotypes, including in key genes associated with ferroptosis, an iron-mediated form of cell death. The loss of FtL did not impact sepsis-mediated activation of NF-κB or HIF-1a signaling, key inflammatory pathways associated with dysregulated host response. Taken together, while FtL overexpression was shown to be protective against sepsis, the loss of FtL did not influence sepsis pathogenesis.NEW & NOTEWORTHY Hyperferritinemia in sepsis is often associated with a proinflammatory phenotype and poor prognosis. We previously showed the myeloid deletion of FtH results in a compensatory increase in FtL and is associated with reduced circulating cytokines and decreased rates of SA-AKI in animal sepsis models. Here, we show that myeloid deletion of FtL does not impact the severity of SA-AKI following CLP or LPS, suggesting that FtH plays the predominant role in propagating myeloid-induced proinflammatory pathways.
Collapse
Affiliation(s)
- James D Odum
- Division of Pediatric Critical Care, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Juheb Akhter
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Vivek Verma
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Giacynta Vollmer
- Division of Pediatric Critical Care, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Ahmad Davidson
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Kelly A Hyndman
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Subhashini Bolisetty
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| |
Collapse
|
4
|
Atre R, Sharma R, Obukhov AG, Saqib U, Umar S, Darwhekar GN, Baig MS. An improved mouse model of sepsis based on intraperitoneal injections of the enriched culture of cecum slurry. Life Sci 2024; 345:122584. [PMID: 38527668 DOI: 10.1016/j.lfs.2024.122584] [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: 12/08/2023] [Revised: 02/07/2024] [Accepted: 03/18/2024] [Indexed: 03/27/2024]
Abstract
AIM Sepsis is a life-threatening clinical syndrome comprising multiorgan dysfunctions caused by a disproportionate body immune response. There are several animal sepsis models which are based on cecum ligation, cecal puncture, and cecum slurry injection. The major limitation of all current sepsis models is the high variability owing to the variable degree of ligation, puncture and inconsistent microbial composition used for sepsis initiation. The primary objective of this work is to demonstrate the feasibility of a standardized method for sepsis development. MATERIALS AND METHODS The cecal slurry bacterial culture was developed and preserved in glycerol stocks. Antibiotics aztreonam and vancomycin were used for generating several defined, enriched cecal slurry bacterial cultures. Mice survival was assessed until 48 hrs post injection, and the tissue samples were collected after 10 hrs from sepsis initiation. KEY FINDINGS The results indicate that increasing polymicrobial load resulted in lower survival rates and was associated with the higher number of infiltrating immune cells and necrosis. H&E (haematoxylin & eosin) staining & serum markers revealed that septic mice exhibited increased inflammation and significant damage to the liver and kidneys. The defined Gram-negative and Gram-positive specific cecal slurry bacterial cultures were developed and their efficiency in inducing sepsis was characterized. SIGNIFICANCE Enriched cecal slurry bacterial cultures can be stored in glycerol stocks at -80 °C. This has an ethical advantage of avoiding unnecessary animal euthanasia for each experiment and provides a standardization capability of sepsis development.
Collapse
Affiliation(s)
- Rajat Atre
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Indore, MP 453552, India
| | - Rahul Sharma
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Indore, MP 453552, India
| | - Alexander G Obukhov
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Uzma Saqib
- School of Life Sciences, Devi Ahilya Vishwavidyalaya, Vigyan Bhawan, Indore, MP 452 001, India
| | - Sadiq Umar
- Department of Oral Biology, College of Dentistry, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Gajanan N Darwhekar
- Acropolis Institute of Pharmaceutical Education and Research (AIPER), Indore, MP 453771, India
| | - Mirza S Baig
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Indore, MP 453552, India.
| |
Collapse
|
5
|
Wakeley ME, Denning NL, Jiang J, De Paepe ME, Chung CS, Wang P, Ayala A. Herpes virus entry mediator signaling blockade produces mortality in neonatal sepsis through induced cardiac dysfunction. Front Immunol 2024; 15:1365174. [PMID: 38774873 PMCID: PMC11106455 DOI: 10.3389/fimmu.2024.1365174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 04/15/2024] [Indexed: 05/24/2024] Open
Abstract
Introduction Sepsis remains a major source of morbidity and mortality in neonates, and characterization of immune regulation in the neonatal septic response remains limited. HVEM is a checkpoint regulator which can both stimulate or inhibit immune responses and demonstrates altered expression after sepsis. We hypothesized that signaling via HVEM would be essential for the neonatal response to sepsis, and that therefore blockade of this pathway would improve survival to septic challenge. Methods To explore this, neonatal mice were treated with cecal slurry (CS), CS with Anti-HVEM antibody (CS-Ab) or CS with isotype (CS-IT) and followed for 7-day survival. Mice from all treatment groups had thymus, lung, kidney and peritoneal fluid harvested, weighed, and stained for histologic evaluation, and changes in cardiac function were assessed with echocardiography. Results Mortality was significantly higher for CS-Ab mice (72.2%) than for CS-IT mice (22.2%). CS resulted in dysregulated alveolar remodeling, but CS-Ab lungs demonstrated significantly less dysfunctional alveolar remodeling than CS alone (MCL 121.0 CS vs. 87.6 CS-Ab), as well as increased renal tubular vacuolization. No morphologic differences in alveolar septation or thymic karyorrhexis were found between CS-Ab and CS-IT. CS-Ab pups exhibited a marked decrease in heart rate (390.3 Sh vs. 342.1 CS-Ab), stroke volume (13.08 CS-IT vs. 8.83 CS-Ab) and ultimately cardiac output (4.90 Sh vs. 3.02 CS-Ab) as well as a significant increase in ejection fraction (73.74 Sh vs. 83.75 CS-Ab) and cardiac strain (40.74 Sh vs. 51.16 CS-Ab) as compared to CS-IT or Sham animals. Discussion While receptor ligation of aspects of HVEM signaling, via antibody blockade, appears to mitigate aspects of lung injury and thymic involution, stimulatory signaling via HVEM still seems to be necessary for vascular and hemodynamic resilience and overall neonatal mouse survival in response to this experimental polymicrobial septic insult. This dissonance in the activity of anti-HVEM neutralizing antibody in neonatal animals speaks to the differences in how septic cardiac dysfunction should be considered and approached in the neonatal population.
Collapse
Affiliation(s)
- Michelle E. Wakeley
- Division of Surgical Research, Department of Surgery, Brown University, Rhode Island Hospital, Providence, RI, United States
| | - Naomi-Liza Denning
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
- Department of Surgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, United States
| | - Jihong Jiang
- Division of Surgical Research, Department of Surgery, Brown University, Rhode Island Hospital, Providence, RI, United States
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Monique E. De Paepe
- Department of Pathology, Women and Infants Hospital, Providence, RI, United States
| | - Chun-Shiang Chung
- Division of Surgical Research, Department of Surgery, Brown University, Rhode Island Hospital, Providence, RI, United States
| | - Ping Wang
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
- Department of Surgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, United States
| | - Alfred Ayala
- Division of Surgical Research, Department of Surgery, Brown University, Rhode Island Hospital, Providence, RI, United States
| |
Collapse
|
6
|
Floyd D, Colbert JF, Feng F, Furgeson SB, Montford JR. Acute and Chronic Kidney Disease Worsen Outcomes in Experimental Sepsis. KIDNEY360 2024; 5:654-670. [PMID: 38353663 PMCID: PMC11146656 DOI: 10.34067/kid.0000000000000391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 02/01/2024] [Indexed: 02/22/2024]
Abstract
Key Points Acute kidney disease (AKD) and CKD are common conditions associated with high rates of incident infection, and poor outcomes once infection have been established. We successfully modeled AKD and CKD in rodents and then administered a cecal slurry solution to create peritonitis and tracked sepsis severity, end organ injury, and inflammatory changes. Our results indicate that AKD mice are more susceptible to infection than CKD mice, developing an aggravated inflammatory response and suggests that this condition predisposes to disparate infection risk. Background Infection is a leading cause of morbidity in individuals with acute kidney disease (AKD) and CKD. However, there is significant difficulty in modeling infection into an animal host with preexisting kidney disease. We report a novel method of peritoneal infection induced via cecal slurry (CS) inoculation deployed into mice with experimental aristolochic acid–induced AKD and CKD. Methods AKD, CKD, and paired control mice were injected with sham, low, or higher doses of donor–recipient matched CS solution. Animal survival, sepsis severity, and change in GFR were tracked longitudinally throughout the study. Histology for kidney injury, flow cytometry, plasma cytokines, and evidence of indirect organ injury from sepsis were also assessed. Results Infected AKD mice experienced significantly heightened sepsis severity, with 100% mortality by 24 hours after high CS doses versus no mortality in control mice. In addition, AKD mice receiving lower CS doses developed dramatically increased proinflammatory cytokines and persistent cytopenias. Infected CKD mice also had worse outcomes than paired CKD controls, although less severe than in AKD mice. Interestingly, animals with AKD had worse outcomes than mice with CKD after any CS dose or time point after inoculation, despite higher baseline kidney function and less uremic sequela. Conclusions These data confirm that acute bacterial infection can be modeled in animals with established kidney disease and suggest that the clinical state of kidney disease (AKD versus CKD) may influence host susceptibility to infection more than the degree of kidney failure alone.
Collapse
Affiliation(s)
- Deana Floyd
- Renal Section, Rocky Mountain Regional VA Medical Center, Aurora, Colorado
| | - James F. Colbert
- Infectious Disease Section, Rocky Mountain Regional VA Medical Center, Aurora, Colorado
- Division of Infectious Disease, University of Colorado School of Medicine, Aurora, Colorado
| | - Frances Feng
- Department of Pathology, University of Colorado School of Medicine, Aurora, Colorado
| | - Seth B. Furgeson
- Division of Nephrology and Hypertension, University of Colorado School of Medicine, Aurora, Colorado
| | - John R. Montford
- Renal Section, Rocky Mountain Regional VA Medical Center, Aurora, Colorado
- Division of Nephrology and Hypertension, University of Colorado School of Medicine, Aurora, Colorado
| |
Collapse
|
7
|
Epstein AA, Janos SN, Menozzi L, Pegram K, Jain V, Bisset LC, Davis JT, Morrison S, Shailaja A, Guo Y, Chao AS, Abdi K, Rikard B, Yao J, Gregory SG, Fisher K, Pittman R, Erkanli A, Gustafson KE, Carrico CWT, Malcolm WF, Inder TE, Cotten CM, Burt TD, Shinohara ML, Maxfield CM, Benner EJ. Subventricular zone stem cell niche injury is associated with intestinal perforation in preterm infants and predicts future motor impairment. Cell Stem Cell 2024; 31:467-483.e6. [PMID: 38537631 PMCID: PMC11129818 DOI: 10.1016/j.stem.2024.03.001] [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: 09/24/2023] [Revised: 02/11/2024] [Accepted: 03/01/2024] [Indexed: 04/07/2024]
Abstract
Brain injury is highly associated with preterm birth. Complications of prematurity, including spontaneous or necrotizing enterocolitis (NEC)-associated intestinal perforations, are linked to lifelong neurologic impairment, yet the mechanisms are poorly understood. Early diagnosis of preterm brain injuries remains a significant challenge. Here, we identified subventricular zone echogenicity (SVE) on cranial ultrasound in preterm infants following intestinal perforations. The development of SVE was significantly associated with motor impairment at 2 years. SVE was replicated in a neonatal mouse model of intestinal perforation. Examination of the murine echogenic subventricular zone (SVZ) revealed NLRP3-inflammasome assembly in multiciliated FoxJ1+ ependymal cells and a loss of the ependymal border in this postnatal stem cell niche. These data suggest a mechanism of preterm brain injury localized to the SVZ that has not been adequately considered. Ultrasound detection of SVE may serve as an early biomarker for neurodevelopmental impairment after inflammatory disease in preterm infants.
Collapse
Affiliation(s)
- Adrian A Epstein
- Department of Pediatrics, Division of Neonatology, Duke University School of Medicine, Durham, NC, USA
| | - Sara N Janos
- Department of Radiology, Duke University School of Medicine, Durham, NC, USA
| | - Luca Menozzi
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Kelly Pegram
- Department of Pediatrics, Division of Neonatology, Duke University School of Medicine, Durham, NC, USA
| | - Vaibhav Jain
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, USA
| | - Logan C Bisset
- Department of Radiology, Duke University School of Medicine, Durham, NC, USA
| | - Joseph T Davis
- Department of Radiology, Duke University School of Medicine, Durham, NC, USA
| | - Samantha Morrison
- Department of Biostatistics & Bioinformatics, Duke University School of Medicine, Durham, NC, USA
| | - Aswathy Shailaja
- Department of Pediatrics, Division of Neonatology, Duke University School of Medicine, Durham, NC, USA
| | - Yingqiu Guo
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Agnes S Chao
- Department of Pediatrics, Division of Neonatology, Duke University School of Medicine, Durham, NC, USA
| | - Khadar Abdi
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, USA
| | - Blaire Rikard
- Department of Pediatrics, Division of Neonatology, Duke University School of Medicine, Durham, NC, USA
| | - Junjie Yao
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Simon G Gregory
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, USA; Department of Neurosurgery, Duke University School of Medicine, Durham, NC, USA
| | - Kimberley Fisher
- Department of Pediatrics, Division of Neonatology, Duke University School of Medicine, Durham, NC, USA
| | - Rick Pittman
- Department of Pediatrics, Division of Neonatology, Duke University School of Medicine, Durham, NC, USA
| | - Al Erkanli
- Department of Biostatistics & Bioinformatics, Duke University School of Medicine, Durham, NC, USA
| | - Kathryn E Gustafson
- Department of Pediatrics, Division of Neonatology, Duke University School of Medicine, Durham, NC, USA
| | | | - William F Malcolm
- Department of Pediatrics, Division of Neonatology, Duke University School of Medicine, Durham, NC, USA
| | - Terrie E Inder
- Department of Pediatric Newborn Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - C Michael Cotten
- Department of Pediatrics, Division of Neonatology, Duke University School of Medicine, Durham, NC, USA
| | - Trevor D Burt
- Department of Pediatrics, Division of Neonatology, Duke University School of Medicine, Durham, NC, USA; Children's Health and Discovery Initiative, Duke University School of Medicine, Durham, NC, USA
| | - Mari L Shinohara
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC 27710, USA; Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Charles M Maxfield
- Department of Radiology, Duke University School of Medicine, Durham, NC, USA.
| | - Eric J Benner
- Department of Pediatrics, Division of Neonatology, Duke University School of Medicine, Durham, NC, USA; Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
8
|
Colón DF, Wanderley CW, Turato WM, Borges VF, Franchin M, Castanheira FVS, Nascimento D, Prado D, Haruo Fernandes de Lima M, Volpon LC, Kavaguti SK, Carlotti AP, Carmona F, Franklin BS, Cunha TM, Alves-Filho JC, Cunha FQ. Paediatric sepsis survivors are resistant to sepsis-induced long-term immune dysfunction. Br J Pharmacol 2024; 181:1308-1323. [PMID: 37990806 DOI: 10.1111/bph.16286] [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: 02/26/2023] [Revised: 07/19/2023] [Accepted: 08/17/2023] [Indexed: 11/23/2023] Open
Abstract
BACKGROUND AND PURPOSE Sepsis-surviving adult individuals commonly develop immunosuppression and increased susceptibility to secondary infections, an outcome mediated by the axis IL-33/ILC2s/M2 macrophages/Tregs. Nonetheless, the long-term immune consequences of paediatric sepsis are indeterminate. We sought to investigate the role of age in the genesis of immunosuppression following sepsis. EXPERIMENTAL APPROACH Here, we compared the frequency of Tregs, the activation of the IL-33/ILC2s axis in M2 macrophages and the DNA methylation of epithelial lung cells from post-septic infant and adult mice. Likewise, sepsis-surviving mice were inoculated intranasally with Pseudomonas aeruginosa or by subcutaneous inoculation of the B16 melanoma cell line. Finally, blood samples from sepsis-surviving patients were collected and the concentration of IL-33 and Tregs frequency were assessed. KEY RESULTS In contrast to 6-week-old mice, 2-week-old mice were resistant to secondary infection and did not show impairment in tumour controls upon melanoma challenge. Mechanistically, increased IL-33 levels, Tregs expansion, and activation of ILC2s and M2-macrophages were observed in 6-week-old but not 2-week-old post-septic mice. Moreover, impaired IL-33 production in 2-week-old post-septic mice was associated with increased DNA methylation in lung epithelial cells. Notably, IL-33 treatment boosted the expansion of Tregs and induced immunosuppression in 2-week-old mice. Clinically, adults but not paediatric post-septic patients exhibited higher counts of Tregs and seral IL-33 levels. CONCLUSION AND IMPLICATIONS These findings demonstrate a crucial and age-dependent role for IL-33 in post-sepsis immunosuppression. Thus, a better understanding of this process may lead to differential treatments for adult and paediatric sepsis.
Collapse
Affiliation(s)
- David F Colón
- Center of Research in Inflammatory Diseases (CRID), University of São Paulo, Ribeirão Preto, Brazil
- Departments of Biochemistry and Immunology, University of São Paulo, Ribeirão Preto, Brazil
| | - Carlos W Wanderley
- Center of Research in Inflammatory Diseases (CRID), University of São Paulo, Ribeirão Preto, Brazil
- Department of Pharmacology, University of São Paulo, Ribeirão Preto, Brazil
| | - Walter M Turato
- Center of Research in Inflammatory Diseases (CRID), University of São Paulo, Ribeirão Preto, Brazil
| | - Vanessa F Borges
- Center of Research in Inflammatory Diseases (CRID), University of São Paulo, Ribeirão Preto, Brazil
- Department of Pharmacology, University of São Paulo, Ribeirão Preto, Brazil
| | - Marcelo Franchin
- School of Dentistry, Alfenas Federal University, Alfenas, Brazil
| | | | - Daniele Nascimento
- Center of Research in Inflammatory Diseases (CRID), University of São Paulo, Ribeirão Preto, Brazil
- Departments of Biochemistry and Immunology, University of São Paulo, Ribeirão Preto, Brazil
| | - Douglas Prado
- Center of Research in Inflammatory Diseases (CRID), University of São Paulo, Ribeirão Preto, Brazil
- Department of Pharmacology, University of São Paulo, Ribeirão Preto, Brazil
| | - Mikhael Haruo Fernandes de Lima
- Center of Research in Inflammatory Diseases (CRID), University of São Paulo, Ribeirão Preto, Brazil
- Departments of Biochemistry and Immunology, University of São Paulo, Ribeirão Preto, Brazil
| | - Leila C Volpon
- Department of Pediatrics, University of São Paulo, Ribeirão Preto, Brazil
| | - Silvia K Kavaguti
- Department of Pediatrics, University of São Paulo, Ribeirão Preto, Brazil
| | - Ana P Carlotti
- Physiology & Pharmacology Calgary, University of Calgary, Calgary, Canada
| | - Fabio Carmona
- Department of Pediatrics, University of São Paulo, Ribeirão Preto, Brazil
| | - Bernardo S Franklin
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Thiago M Cunha
- Center of Research in Inflammatory Diseases (CRID), University of São Paulo, Ribeirão Preto, Brazil
- Department of Pharmacology, University of São Paulo, Ribeirão Preto, Brazil
| | - Jose Carlos Alves-Filho
- Center of Research in Inflammatory Diseases (CRID), University of São Paulo, Ribeirão Preto, Brazil
- Departments of Biochemistry and Immunology, University of São Paulo, Ribeirão Preto, Brazil
| | - Fernando Q Cunha
- Center of Research in Inflammatory Diseases (CRID), University of São Paulo, Ribeirão Preto, Brazil
- Department of Pharmacology, University of São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|
9
|
Kannan SK, Kim CY, Heidarian M, Berton RR, Jensen IJ, Griffith TS, Badovinac VP. Mouse Models of Sepsis. Curr Protoc 2024; 4:e997. [PMID: 38439603 PMCID: PMC10917121 DOI: 10.1002/cpz1.997] [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] [Indexed: 03/06/2024]
Abstract
Human sepsis is a complex disease that manifests with a diverse range of phenotypes and inherent variability among individuals, making it hard to develop a comprehensive animal model. Despite this difficulty, numerous models have been developed that capture many key aspects of human sepsis. The robustness of these models is vital for conducting pre-clinical studies to test and develop potential therapeutics. In this article, we describe four different models of murine sepsis that can be used to address different scientific questions relevant to the pathology and immune response during and after a septic event. Basic Protocol 1 details a non-synchronous cecal ligation and puncture (CLP) model of sepsis, where mice are subjected to polymicrobial exposure through surgery at different time points within 2 weeks. This variation in sepsis onset establishes each mouse at a different state of inflammation and cytokine levels that mimics the variability observed in humans when they present in the clinic. This model is ideal for studying the long-term impact of sepsis on the host. Basic Protocol 2 is also a type of polymicrobial sepsis, where injection of a specific amount of cecal slurry from a donor mouse into the peritoneum of recipient mice establishes immediate inflammation and sepsis without any need for surgery. Basic Protocol 3 describes infecting mice with a defined gram-positive or -negative bacterial strain to model a subset of sepsis observed in humans infected with a single pathogen. Basic Protocol 4 describes administering LPS to induce sterile endotoxemia. This form of sepsis is observed in humans exposed to bacterial toxins from the environment. © 2024 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: Non-synchronous cecal ligation and puncture Basic Protocol 2: Cecal slurry model of murine sepsis Basic Protocol 3: Monomicrobial model of murine sepsis Basic Protocol 4: LPS model of murine sepsis.
Collapse
Affiliation(s)
- Shravan-Kumar Kannan
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, USA
| | - Caleb Y. Kim
- Microbiology, Immunology, and Cancer Biology Program, University of Minnesota, Minneapolis, Minnesota, USA
| | | | - Roger R. Berton
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, USA
| | - Isaac J. Jensen
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, USA
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York City, New York, USA
| | - Thomas S. Griffith
- Microbiology, Immunology, and Cancer Biology Program, University of Minnesota, Minneapolis, Minnesota, USA
- Department of Urology, University of Minnesota, Minneapolis, Minnesota, USA
- Minneapolis Veterans Affairs Health Care System, Minneapolis, Minnesota, USA
| | - Vladimir P. Badovinac
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, USA
- Microbiology, Immunology, and Cancer Biology Program, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
10
|
Vergadi E, Kolliniati O, Lapi I, Ieronymaki E, Lyroni K, Alexaki VI, Diamantaki E, Vaporidi K, Hatzidaki E, Papadaki HA, Galanakis E, Hajishengallis G, Chavakis T, Tsatsanis C. An IL-10/DEL-1 axis supports granulopoiesis and survival from sepsis in early life. Nat Commun 2024; 15:680. [PMID: 38263289 PMCID: PMC10805706 DOI: 10.1038/s41467-023-44178-y] [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: 10/24/2021] [Accepted: 12/03/2023] [Indexed: 01/25/2024] Open
Abstract
The limited reserves of neutrophils are implicated in the susceptibility to infection in neonates, however the regulation of neutrophil kinetics in infections in early life remains poorly understood. Here we show that the developmental endothelial locus (DEL-1) is elevated in neonates and is critical for survival from neonatal polymicrobial sepsis, by supporting emergency granulopoiesis. Septic DEL-1 deficient neonate mice display low numbers of myeloid-biased multipotent and granulocyte-macrophage progenitors in the bone marrow, resulting in neutropenia, exaggerated bacteremia, and increased mortality; defects that are rescued by DEL-1 administration. A high IL-10/IL-17A ratio, observed in newborn sepsis, sustains tissue DEL-1 expression, as IL-10 upregulates while IL-17 downregulates DEL-1. Consistently, serum DEL-1 and blood neutrophils are elevated in septic adult and neonate patients with high serum IL-10/IL-17A ratio, and mortality is lower in septic patients with high serum DEL-1. Therefore, IL-10/DEL-1 axis supports emergency granulopoiesis, prevents neutropenia and promotes sepsis survival in early life.
Collapse
Affiliation(s)
- Eleni Vergadi
- Department of Paediatrics, School of Medicine, University of Crete, Heraklion, Greece.
- Institute of Molecular Biology and Biotechnology, IMMB, FORTH, Heraklion, Greece.
| | - Ourania Kolliniati
- Institute of Molecular Biology and Biotechnology, IMMB, FORTH, Heraklion, Greece
- Department of Clinical Chemistry, School of Medicine, University of Crete, Heraklion, Greece
| | - Ioanna Lapi
- Institute of Molecular Biology and Biotechnology, IMMB, FORTH, Heraklion, Greece
- Department of Clinical Chemistry, School of Medicine, University of Crete, Heraklion, Greece
| | - Eleftheria Ieronymaki
- Institute of Molecular Biology and Biotechnology, IMMB, FORTH, Heraklion, Greece
- Department of Clinical Chemistry, School of Medicine, University of Crete, Heraklion, Greece
| | - Konstantina Lyroni
- Institute of Molecular Biology and Biotechnology, IMMB, FORTH, Heraklion, Greece
- Department of Clinical Chemistry, School of Medicine, University of Crete, Heraklion, Greece
| | - Vasileia Ismini Alexaki
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Eleni Diamantaki
- Department of Intensive Care Medicine, School of Medicine, University of Crete, Heraklion, Greece
| | - Katerina Vaporidi
- Department of Intensive Care Medicine, School of Medicine, University of Crete, Heraklion, Greece
| | - Eleftheria Hatzidaki
- Department of Neonatology/Neonatal Intensive Care Unit, School of Medicine, University of Crete, Heraklion, Greece
| | - Helen A Papadaki
- Department of Hematology, School of Medicine, University of Crete, Heraklion, Greece
| | - Emmanouil Galanakis
- Department of Paediatrics, School of Medicine, University of Crete, Heraklion, Greece
| | - George Hajishengallis
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Triantafyllos Chavakis
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Christos Tsatsanis
- Institute of Molecular Biology and Biotechnology, IMMB, FORTH, Heraklion, Greece
- Department of Clinical Chemistry, School of Medicine, University of Crete, Heraklion, Greece
| |
Collapse
|
11
|
Qiao H, Zienkiewicz J, Liu Y, Hawiger J. Activation of thousands of genes in the lungs and kidneys by sepsis is countered by the selective nuclear blockade. Front Immunol 2023; 14:1221102. [PMID: 37638006 PMCID: PMC10450963 DOI: 10.3389/fimmu.2023.1221102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 07/24/2023] [Indexed: 08/29/2023] Open
Abstract
The steady rise of sepsis globally has reached almost 49 million cases in 2017, and 11 million sepsis-related deaths. The genomic response to sepsis comprising multi-system stage of raging microbial inflammation has been reported in the whole blood, while effective treatment is lacking besides anti-microbial therapy and supportive measures. Here we show that, astoundingly, 6,237 significantly expressed genes in sepsis are increased or decreased in the lungs, the site of acute respiratory distress syndrome (ARDS). Moreover, 5,483 significantly expressed genes in sepsis are increased or decreased in the kidneys, the site of acute injury (AKI). This massive genomic response to polymicrobial sepsis is countered by the selective nuclear blockade with the cell-penetrating Nuclear Transport Checkpoint Inhibitor (NTCI). It controlled 3,735 sepsis-induced genes in the lungs and 1,951 sepsis-induced genes in the kidneys. The NTCI also reduced without antimicrobial therapy the bacterial dissemination: 18-fold in the blood, 11-fold in the lungs, and 9-fold in the spleen. This enhancement of bacterial clearance was not significant in the kidneys. Cumulatively, identification of the sepsis-responsive host's genes and their control by the selective nuclear blockade advances a better understanding of the multi-system mechanism of sepsis. Moreover, it spurs much-needed new diagnostic, therapeutic, and preventive approaches.
Collapse
Affiliation(s)
- Huan Qiao
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, TN, United States
| | - Jozef Zienkiewicz
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, TN, United States
- Department of Veterans Affairs, Tennessee Valley Health Care System, Nashville, Tennessee, TN, United States
| | - Yan Liu
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, TN, United States
- Department of Veterans Affairs, Tennessee Valley Health Care System, Nashville, Tennessee, TN, United States
| | - Jacek Hawiger
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, TN, United States
- Department of Veterans Affairs, Tennessee Valley Health Care System, Nashville, Tennessee, TN, United States
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, TN, United States
| |
Collapse
|
12
|
Chao AS, Matak P, Pegram K, Powers J, Hutson C, Jo R, Dubois L, Thompson JW, Smith PB, Jain V, Liu C, Younge NE, Rikard B, Reyes EY, Shinohara ML, Gregory SG, Goldberg RN, Benner EJ. 20-αHydroxycholesterol, an oxysterol in human breast milk, reverses mouse neonatal white matter injury through Gli-dependent oligodendrogenesis. Cell Stem Cell 2023; 30:1054-1071.e8. [PMID: 37541211 PMCID: PMC10625465 DOI: 10.1016/j.stem.2023.07.010] [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: 10/27/2020] [Revised: 05/21/2023] [Accepted: 07/12/2023] [Indexed: 08/06/2023]
Abstract
White matter injuries (WMIs) are the leading cause of neurologic impairment in infants born premature. There are no treatment options available. The most common forms of WMIs in infants occur prior to the onset of normal myelination, making its pathophysiology distinctive, thus requiring a tailored approach to treatment. Neonates present a unique opportunity to repair WMIs due to a transient abundance of neural stem/progenitor cells (NSPCs) present in the germinal matrix with oligodendrogenic potential. We identified an endogenous oxysterol, 20-αHydroxycholesterol (20HC), in human maternal breast milk that induces oligodendrogenesis through a sonic hedgehog (shh), Gli-dependent mechanism. Following WMI in neonatal mice, injection of 20HC induced subventricular zone-derived oligodendrogenesis and improved myelination in the periventricular white matter, resulting in improved motor outcomes. Targeting the oligodendrogenic potential of postnatal NSPCs in neonates with WMIs may be further developed into a novel approach to mitigate this devastating complication of preterm birth.
Collapse
Affiliation(s)
- Agnes S Chao
- Division of Neonatology, Department of Pediatrics, Duke University Medical Center, The Jean and George Brumley, Jr. Neonatal-Perinatal Institute, Durham, NC 27710, USA
| | - Pavle Matak
- Division of Neonatology, Department of Pediatrics, Duke University Medical Center, The Jean and George Brumley, Jr. Neonatal-Perinatal Institute, Durham, NC 27710, USA
| | - Kelly Pegram
- Division of Neonatology, Department of Pediatrics, Duke University Medical Center, The Jean and George Brumley, Jr. Neonatal-Perinatal Institute, Durham, NC 27710, USA
| | - James Powers
- Division of Neonatology, Department of Pediatrics, Duke University Medical Center, The Jean and George Brumley, Jr. Neonatal-Perinatal Institute, Durham, NC 27710, USA
| | - Collin Hutson
- Division of Neonatology, Department of Pediatrics, Duke University Medical Center, The Jean and George Brumley, Jr. Neonatal-Perinatal Institute, Durham, NC 27710, USA
| | - Rebecca Jo
- Division of Neonatology, Department of Pediatrics, Duke University Medical Center, The Jean and George Brumley, Jr. Neonatal-Perinatal Institute, Durham, NC 27710, USA
| | - Laura Dubois
- Duke Proteomics and Metabolomics Shared Resource, Center for Genomics and Computational Biology, School of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - J Will Thompson
- Duke Proteomics and Metabolomics Shared Resource, Center for Genomics and Computational Biology, School of Medicine, Duke University Medical Center, Durham, NC 27710, USA; Department of Pharmacology and Cancer Biology, School of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - P Brian Smith
- Division of Neonatology, Department of Pediatrics, Duke University Medical Center, The Jean and George Brumley, Jr. Neonatal-Perinatal Institute, Durham, NC 27710, USA
| | - Vaibhav Jain
- Department of Neurology, Duke University Medical Center, Durham, NC 27710, USA; Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC 27710, USA
| | - Chunlei Liu
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, Berkeley, CA 94720, USA; Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Noelle E Younge
- Division of Neonatology, Department of Pediatrics, Duke University Medical Center, The Jean and George Brumley, Jr. Neonatal-Perinatal Institute, Durham, NC 27710, USA
| | - Blaire Rikard
- Division of Neonatology, Department of Pediatrics, Duke University Medical Center, The Jean and George Brumley, Jr. Neonatal-Perinatal Institute, Durham, NC 27710, USA
| | - Estefany Y Reyes
- Department of Immunology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Mari L Shinohara
- Department of Immunology, Duke University School of Medicine, Durham, NC 27710, USA; Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Simon G Gregory
- Department of Neurology, Duke University Medical Center, Durham, NC 27710, USA; Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC 27710, USA
| | - Ronald N Goldberg
- Division of Neonatology, Department of Pediatrics, Duke University Medical Center, The Jean and George Brumley, Jr. Neonatal-Perinatal Institute, Durham, NC 27710, USA
| | - Eric J Benner
- Division of Neonatology, Department of Pediatrics, Duke University Medical Center, The Jean and George Brumley, Jr. Neonatal-Perinatal Institute, Durham, NC 27710, USA; Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
13
|
Cai L, Rodgers E, Schoenmann N, Raju RP. Advances in Rodent Experimental Models of Sepsis. Int J Mol Sci 2023; 24:9578. [PMID: 37298529 PMCID: PMC10253762 DOI: 10.3390/ijms24119578] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 05/09/2023] [Accepted: 05/23/2023] [Indexed: 06/12/2023] Open
Abstract
In the development of therapeutic strategies for human diseases, preclinical experimental models have a key role. However, the preclinical immunomodulatory therapies developed using rodent sepsis were not successful in human clinical trials. Sepsis is characterized by a dysregulated inflammation and redox imbalance triggered by infection. Human sepsis is simulated in experimental models using methods that trigger inflammation or infection in the host animals, most often mice or rats. It remains unknown whether the characteristics of the host species, the methods used to induce sepsis, or the molecular processes focused upon need to be revisited in the development of treatment methods that will succeed in human clinical trials. Our goal in this review is to provide a survey of existing experimental models of sepsis, including the use of humanized mice and dirty mice, and to show how these models reflect the clinical course of sepsis. We will discuss the strengths and limitations of these models and present recent advances in this subject area. We maintain that rodent models continue to have an irreplaceable role in studies toward discovering treatment methods for human sepsis.
Collapse
Affiliation(s)
- Lun Cai
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Elizabeth Rodgers
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Nick Schoenmann
- Department of Emergency Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Raghavan Pillai Raju
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| |
Collapse
|
14
|
Denorme F, Rustad JL, Portier I, Crandell JL, de Araujo CV, Cody MJ, Campbell RA, Yost CC. Neutrophil extracellular trap inhibition improves survival in neonatal mouse infectious peritonitis. Pediatr Res 2023; 93:862-869. [PMID: 35902703 PMCID: PMC9331023 DOI: 10.1038/s41390-022-02219-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 06/25/2022] [Accepted: 07/09/2022] [Indexed: 12/01/2022]
Abstract
BACKGROUND Treatment of neonatal peritonitis and sepsis is challenging. Following infection, neutrophils elaborate neutrophil extracellular traps (NETs)-extracellular lattices of decondensed chromatin decorated with antimicrobial proteins. NETs, however, can augment pathogenic inflammation causing collateral damage. We hypothesized that NET inhibition would improve survival in experimental neonatal infectious peritonitis. METHODS We induced peritonitis in 7 to 10-day-old mice by intraperitoneal injection with cecal slurry. We targeted NETs by treating mice with neonatal NET-Inhibitory Factor (nNIF), an endogenous NET-inhibitor; Cl-amidine, a PAD4 inhibitor; DNase I, a NET degrading enzyme, or meropenem (an antibiotic). We determined peritoneal NET and cytokine levels and circulating platelet-neutrophil aggregates. Survival from peritonitis was followed for 6 days. RESULTS nNIF, Cl-amidine, and DNase I decreased peritoneal NET formation and inflammatory cytokine levels at 24 h compared to controls. nNIF, Cl-amidine, and DNase I decreased circulating platelet-neutrophil aggregates, and NET-targeting treatments significantly increased survival from infectious peritonitis compared to controls. Finally, nNIF administration significantly improved survival in mice treated with sub-optimal doses of meropenem even when treatment was delayed until 2 h after peritonitis induction. CONCLUSIONS NET inhibition improves survival in experimental neonatal infectious peritonitis, suggesting that NETs participate pathogenically in neonatal peritonitis and sepsis. IMPACT 1. Neutrophil extracellular trap formation participates pathogenically in experimental neonatal infectious peritonitis. 2. NET-targeting strategies improve outcomes in a translational model of neonatal infectious peritonitis. 3. NET inhibition represents a potential target for drug development in neonatal sepsis and infectious peritonitis.
Collapse
Affiliation(s)
| | - John L Rustad
- Molecular Medicine Program, Salt Lake City, UT, 84112, USA
| | - Irina Portier
- Molecular Medicine Program, Salt Lake City, UT, 84112, USA
| | | | - Claudia V de Araujo
- Molecular Medicine Program, Salt Lake City, UT, 84112, USA
- Department of Pediatrics/Neonatology, Salt Lake City, UT, 84112, USA
| | - Mark J Cody
- Molecular Medicine Program, Salt Lake City, UT, 84112, USA
- Department of Pediatrics/Neonatology, Salt Lake City, UT, 84112, USA
| | - Robert A Campbell
- Molecular Medicine Program, Salt Lake City, UT, 84112, USA
- Department of Internal Medicine, University of Utah, Salt Lake City, UT, 84112, USA
| | - Christian C Yost
- Molecular Medicine Program, Salt Lake City, UT, 84112, USA.
- Department of Pediatrics/Neonatology, Salt Lake City, UT, 84112, USA.
| |
Collapse
|
15
|
MicroRNA Profiles in Intestinal Epithelial Cells in a Mouse Model of Sepsis. Cells 2023; 12:cells12050726. [PMID: 36899862 PMCID: PMC10001189 DOI: 10.3390/cells12050726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 02/22/2023] [Accepted: 02/22/2023] [Indexed: 03/03/2023] Open
Abstract
Sepsis is a systemic inflammatory disorder that leads to the dysfunction of multiple organs. In the intestine, the deregulation of the epithelial barrier contributes to the development of sepsis by triggering continuous exposure to harmful factors. However, sepsis-induced epigenetic changes in gene-regulation networks within intestinal epithelial cells (IECs) remain unexplored. In this study, we analyzed the expression profile of microRNAs (miRNAs) in IECs isolated from a mouse model of sepsis generated via cecal slurry injection. Among 239 miRNAs, 14 miRNAs were upregulated, and 9 miRNAs were downregulated in the IECs by sepsis. Upregulated miRNAs in IECs from septic mice, particularly miR-149-5p, miR-466q, miR-495, and miR-511-3p, were seen to exhibit complex and global effects on gene regulation networks. Interestingly, miR-511-3p has emerged as a diagnostic marker in this sepsis model due to its increase in blood in addition to IECs. As expected, mRNAs in the IECs were remarkably altered by sepsis; specifically, 2248 mRNAs were decreased, while 612 mRNAs were increased. This quantitative bias may be possibly derived, at least partly, from the direct effects of the sepsis-increased miRNAs on the comprehensive expression of mRNAs. Thus, current in silico data indicate that there are dynamic regulatory responses of miRNAs to sepsis in IECs. In addition, the miRNAs that were increased with sepsis had enriched downstream pathways including Wnt signaling, which is associated with wound healing, and FGF/FGFR signaling, which has been linked to chronic inflammation and fibrosis. These modifications in miRNA networks in IECs may lead to both pro- and anti-inflammatory effects in sepsis. The four miRNAs discovered above were shown to putatively target LOX, PTCH1, COL22A1, FOXO1, or HMGA2, via in silico analysis, which were associated with Wnt or inflammatory pathways and selected for further study. The expressions of these target genes were downregulated in sepsis IECs, possibly through posttranscriptional modifications of these miRNAs. Taken together, our study suggests that IECs display a distinctive miRNA profile which is capable of comprehensively and functionally reshaping the IEC-specific mRNA landscape in a sepsis model.
Collapse
|
16
|
Zhai H, Zhang J, Shang D, Zhu C, Xiang X. The progress to establish optimal animal models for the study of acute-on-chronic liver failure. Front Med (Lausanne) 2023; 10:1087274. [PMID: 36844207 PMCID: PMC9947362 DOI: 10.3389/fmed.2023.1087274] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 01/23/2023] [Indexed: 02/11/2023] Open
Abstract
Acute-on-chronic liver failure (ACLF) defines a complicated and multifaceted syndrome characterized by acute liver dysfunction following an acute insult on the basis of chronic liver diseases. It is usually concurrent with bacterial infection and multi-organ failure resulting in high short-term mortality. Based on the cohort studies in ACLF worldwide, the clinical course of ACLF was demonstrated to comprise three major stages including chronic liver injury, acute hepatic/extrahepatic insult, and systemic inflammatory response caused by over-reactive immune system especially bacterial infection. However, due to the lack of optimal experimental animal models for ACLF, the progress of basic study on ACLF is limping. Though several experimental ACLF models were established, none of them can recapitulate and simulate the whole pathological process of ACLF patients. Recently, we have developed a novel mouse model for ACLF combining chronic liver injury [injection of carbon tetrachloride (CCl4) for 8 weeks], acute hepatic insult (injection of a double dose CCl4), and bacterial infection (intraperitoneal injection of Klebsiella pneumoniae), which could recapitulate the major clinical features of patients with ACLF worsened by bacterial infection.
Collapse
Affiliation(s)
- Hengben Zhai
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China,Translational Lab of Liver Diseases, Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jinming Zhang
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China,Translational Lab of Liver Diseases, Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dabao Shang
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China,Translational Lab of Liver Diseases, Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chuanwu Zhu
- Department of Infectious Diseases, The Fifth People’s Hospital of Suzhou, Suzhou, China,Chuanwu Zhu,
| | - Xiaogang Xiang
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China,Translational Lab of Liver Diseases, Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China,*Correspondence: Xiaogang Xiang,
| |
Collapse
|
17
|
Oncostatin M Receptor Type II Knockout Mitigates Inflammation and Improves Survival from Sepsis in Mice. Biomedicines 2023; 11:biomedicines11020483. [PMID: 36831019 PMCID: PMC9953488 DOI: 10.3390/biomedicines11020483] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 01/27/2023] [Accepted: 01/31/2023] [Indexed: 02/11/2023] Open
Abstract
Sepsis remains one of the leading causes of death worldwide. Oncostatin M (OSM), an interleukin (IL)-6 family cytokine, can be found at high levels in septic patients. However, little is known about its role in sepsis. This study aimed to determine if the genetic knockout of OSM receptor (OSMR) type II signaling would improve survival in a murine model of sepsis. Aged (>50 weeks) OSMR type II knockout (KO) mice and wild-type (WT) littermates received an intraperitoneal injection of fecal slurry (FS) or vehicle. The KO mice had better survival 48 h after the injection of FS than the WT mice (p = 0.005). Eighteen hours post-FS injection, the KO mice had reduced peritoneal, serum, and tissue cytokine levels (including IL-1β, IL-6, TNFα, KG/GRO, and IL-10) compared to the WT mice (p < 0.001 for all). Flow cytometry revealed decreased recruitment of CD11b+ F4/80+ Ly6chigh+ macrophages in the peritoneum of KO mice compared to WT mice (34 ± 6 vs. 4 ± 3%, PInt = 0.005). Isolated peritoneal macrophages from aged KO mice had better live E. coli killing capacity than those from WT mice (p < 0.001). Peritoneal lavage revealed greater bacterial counts in KO mice than in WT mice (KO: 305 ± 22 vs. 116 ± 6 CFU (×109)/mL; p < 0.001). In summary, deficiency in OSMR type II receptor signaling provided a survival benefit in the progression of sepsis. This coincided with reduced serum levels of pro-inflammatory (IL-1β, TNFα, and KC/GRO) and anti-inflammatory markers (IL-10), increased bacterial killing ability of macrophages, and reduced macrophage infiltration into to site of infection.
Collapse
|
18
|
Green EA, Garrick SP, Peterson B, Berger PJ, Galinsky R, Hunt RW, Cho SX, Bourke JE, Nold MF, Nold-Petry CA. The Role of the Interleukin-1 Family in Complications of Prematurity. Int J Mol Sci 2023; 24:2795. [PMID: 36769133 PMCID: PMC9918069 DOI: 10.3390/ijms24032795] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/20/2023] [Accepted: 01/22/2023] [Indexed: 02/05/2023] Open
Abstract
Preterm birth is a major contributor to neonatal morbidity and mortality. Complications of prematurity such as bronchopulmonary dysplasia (BPD, affecting the lung), pulmonary hypertension associated with BPD (BPD-PH, heart), white matter injury (WMI, brain), retinopathy of prematurity (ROP, eyes), necrotizing enterocolitis (NEC, gut) and sepsis are among the major causes of long-term morbidity in infants born prematurely. Though the origins are multifactorial, inflammation and in particular the imbalance of pro- and anti-inflammatory mediators is now recognized as a key driver of the pathophysiology underlying these illnesses. Here, we review the involvement of the interleukin (IL)-1 family in perinatal inflammation and its clinical implications, with a focus on the potential of these cytokines as therapeutic targets for the development of safe and effective treatments for early life inflammatory diseases.
Collapse
Affiliation(s)
- Elys A. Green
- Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC 3168, Australia
- Department of Paediatrics, Monash University, Melbourne, VIC 3168, Australia
- Monash Newborn, Monash Children’s Hospital, Melbourne, VIC 3168, Australia
| | - Steven P. Garrick
- Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC 3168, Australia
- Department of Paediatrics, Monash University, Melbourne, VIC 3168, Australia
| | - Briana Peterson
- Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC 3168, Australia
- Department of Paediatrics, Monash University, Melbourne, VIC 3168, Australia
| | - Philip J. Berger
- Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC 3168, Australia
- Department of Paediatrics, Monash University, Melbourne, VIC 3168, Australia
| | - Robert Galinsky
- Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC 3168, Australia
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC 3168, Australia
| | - Rod W. Hunt
- Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC 3168, Australia
- Department of Paediatrics, Monash University, Melbourne, VIC 3168, Australia
- Monash Newborn, Monash Children’s Hospital, Melbourne, VIC 3168, Australia
| | - Steven X. Cho
- Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC 3168, Australia
- Department of Paediatrics, Monash University, Melbourne, VIC 3168, Australia
| | - Jane E. Bourke
- Department of Pharmacology, Biomedicine Discovery Institute, Monash University, Melbourne, VIC 3168, Australia
| | - Marcel F. Nold
- Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC 3168, Australia
- Department of Paediatrics, Monash University, Melbourne, VIC 3168, Australia
- Monash Newborn, Monash Children’s Hospital, Melbourne, VIC 3168, Australia
| | - Claudia A. Nold-Petry
- Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC 3168, Australia
- Department of Paediatrics, Monash University, Melbourne, VIC 3168, Australia
| |
Collapse
|
19
|
Probiotic Potential of the Marine Isolate Enterococcus faecium EA9 and In Vivo Evaluation of Its Antisepsis Action in Rats. Mar Drugs 2023; 21:md21010045. [PMID: 36662218 PMCID: PMC9860781 DOI: 10.3390/md21010045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 01/03/2023] [Accepted: 01/04/2023] [Indexed: 01/11/2023] Open
Abstract
This study aims to obtain a novel probiotic strain adapted to marine habitats and to assess its antisepsis properties using a cecal ligation and puncture (CLP) model in rodents. The marine Enterococcus faecium EA9 was isolated from marine shrimp samples and evaluated for probiotic potential after phenotypical and molecular identification. In septic animals, hepatic and renal tissues were histologically and biochemically evaluated for inflammation and oxidative stress following the probiotic treatment. Moreover, gene expressions of multiple signaling cascades were determined using RT-PCR. EA9 was identified and genotyped as Enterococcus faecium with a 99.88% identity. EA9 did not exhibit any signs of hemolysis and survived at low pH and elevated concentrations of bile salts. Moreover, EA9 isolate had antibacterial activity against different pathogenic bacteria and could thrive in 6.5% NaCl. Septic animals treated with EA9 had improved liver and kidney functions, lower inflammatory and lipid peroxidation biomarkers, and enhanced antioxidant enzymes. The CLP-induced necrotic histological changes and altered gene expressions of IL-10, IL-1β, INF-γ, COX-2, SOD-1, SOD-2, HO-1, AKT, mTOR, iNOS, and STAT-3 were abolished by the EA9 probiotic in septic animals. The isolate Enterococcus faecium EA9 represents a promising marine probiotic. The in vivo antisepsis testing of EA9 highlighted its potential and effective therapeutic approach.
Collapse
|
20
|
Zhou H, Lu X, Huang J, Jordan P, Ma S, Xu L, Hu F, Gui H, Zhao H, Bai Z, Redmond HP, Wang JH, Wang J. Induction of Trained Immunity Protects Neonatal Mice Against Microbial Sepsis by Boosting Both the Inflammatory Response and Antimicrobial Activity. J Inflamm Res 2022; 15:3829-3845. [PMID: 35836719 PMCID: PMC9273902 DOI: 10.2147/jir.s363995] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 06/21/2022] [Indexed: 11/28/2022] Open
Abstract
Background Neonates are susceptible to a wide range of microbial infection and at a high risk to develop severe sepsis and septic shock. Emerged evidence has shown that induction of trained immunity triggers a much stronger inflammatory response in adult monocytes/macrophages, thereby conferring protection against microbial infection. Methods This study was carried out to examine whether trained immunity is inducible and exerts its protection against microbial sepsis in neonates. Results Induction of trained immunity by Bacillus Calmette-Guerin (BCG) plus bacterial lipoprotein (BLP) protected neonatal mice against cecal slurry peritonitis-induced polymicrobial sepsis, and this protection is associated with elevated circulating inflammatory cytokines, increased neutrophil recruitment, and accelerated bacterial clearance. In vitro stimulation of neonatal murine macrophages with BCG+BLP augmented both inflammatory response and antimicrobial activity. Notably, BCG+BLP stimulation resulted in epigenetic remodeling characterized by histone modifications with enhanced H3K4me3, H3K27Ac, and suppressed H3K9me3 at the promoters of the targeted inflammatory and antimicrobial genes. Critically, BCG+BLP stimulation led to a shift in cellular metabolism with increased glycolysis, which is the prerequisite for subsequent BCG+BLP-triggered epigenetic reprogramming and augmented inflammatory response and antimicrobial capacity. Conclusion These results illustrate that BCG+BLP induces trained immunity in neonates, thereby protecting against microbial infection by boosting both inflammatory and antimicrobial responses.
Collapse
Affiliation(s)
- Huiting Zhou
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, People's Republic of China
| | - Xiaying Lu
- Department of Academic Surgery, University College Cork, Cork University Hospital, Cork, Ireland.,Department of Physiology, Gannan Medical University, Ganzhou, People's Republic of China
| | - Jie Huang
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, People's Republic of China
| | - Patrick Jordan
- Department of Academic Surgery, University College Cork, Cork University Hospital, Cork, Ireland
| | - Shurong Ma
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, People's Republic of China
| | - Lingqi Xu
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, People's Republic of China
| | - Fangjie Hu
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, People's Republic of China
| | - Huan Gui
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, People's Republic of China
| | - He Zhao
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, People's Republic of China
| | - Zhenjiang Bai
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, People's Republic of China
| | - H Paul Redmond
- Department of Academic Surgery, University College Cork, Cork University Hospital, Cork, Ireland
| | - Jiang Huai Wang
- Department of Academic Surgery, University College Cork, Cork University Hospital, Cork, Ireland
| | - Jian Wang
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, People's Republic of China
| |
Collapse
|
21
|
Hensler E, Petros H, Gray CC, Chung CS, Ayala A, Fallon EA. The Neonatal Innate Immune Response to Sepsis: Checkpoint Proteins as Novel Mediators of This Response and as Possible Therapeutic/Diagnostic Levers. Front Immunol 2022; 13:940930. [PMID: 35860251 PMCID: PMC9289477 DOI: 10.3389/fimmu.2022.940930] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 06/07/2022] [Indexed: 11/23/2022] Open
Abstract
Sepsis, a dysfunctional immune response to infection leading to life-threatening organ injury, represents a significant global health issue. Neonatal sepsis is disproportionately prevalent and has a cost burden of 2-3 times that of adult patients. Despite this, no widely accepted definition for neonatal sepsis or recommendations for management exist and those created for pediatric patients are significantly limited in their applicability to this unique population. This is in part due to neonates' reliance on an innate immune response (which is developmentally more prominent in the neonate than the immature adaptive immune response) carried out by dysfunctional immune cells, including neutrophils, antigen-presenting cells such as macrophages/monocytes, dendritic cells, etc., natural killer cells, and innate lymphoid regulatory cell sub-sets like iNKT cells, γδ T-cells, etc. Immune checkpoint inhibitors are a family of proteins with primarily suppressive/inhibitory effects on immune and tumor cells and allow for the maintenance of self-tolerance. During sepsis, these proteins are often upregulated and are thought to contribute to the long-term immunosuppression seen in adult patients. Several drugs targeting checkpoint inhibitors, including PD-1 and PD-L1, have been developed and approved for the treatment of various cancers, but no such therapeutics have been approved for the management of sepsis. In this review, we will comparatively discuss the role of several checkpoint inhibitor proteins, including PD-1, PD-L1, VISTA, and HVEM, in the immune response to sepsis in both adults and neonates, as well as posit how they may uniquely propagate their actions through the neonatal innate immune response. We will also consider the possibility of leveraging these proteins in the clinical setting as potential therapeutics/diagnostics that might aid in mitigating neonatal septic morbidity/mortality.
Collapse
Affiliation(s)
- Emily Hensler
- Division of Surgical Research, Department of Surgery, Rhode Island Hospital, Providence, RI, United States,Graduate Program in Biotechnology, Brown University, Providence, RI, United States
| | - Habesha Petros
- Graduate Program in Biotechnology, Brown University, Providence, RI, United States
| | - Chyna C. Gray
- Division of Surgical Research, Department of Surgery, Rhode Island Hospital, Providence, RI, United States,Graduate Program in Biotechnology, Brown University, Providence, RI, United States
| | - Chun-Shiang Chung
- Division of Surgical Research, Department of Surgery, Rhode Island Hospital, Providence, RI, United States,Graduate Program in Biotechnology, Brown University, Providence, RI, United States
| | - Alfred Ayala
- Division of Surgical Research, Department of Surgery, Rhode Island Hospital, Providence, RI, United States,Graduate Program in Biotechnology, Brown University, Providence, RI, United States,*Correspondence: Alfred Ayala,
| | - Eleanor A. Fallon
- Division of Surgical Research, Department of Surgery, Rhode Island Hospital, Providence, RI, United States,Graduate Program in Biotechnology, Brown University, Providence, RI, United States
| |
Collapse
|
22
|
Meegan JE, Komalavilas P, Cheung‐Flynn J, Yim TW, Putz ND, Jesse JJ, Smith KD, Sidorova TN, Lee HNR, Tomasek T, Shaver CM, Ware LB, Brophy CM, Bastarache JA. Blocking P2X7 receptor with AZ 10606120 exacerbates vascular hyperpermeability and inflammation in murine polymicrobial sepsis. Physiol Rep 2022; 10:e15290. [PMID: 35668576 PMCID: PMC9170948 DOI: 10.14814/phy2.15290] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 04/03/2022] [Accepted: 04/13/2022] [Indexed: 11/25/2022] Open
Abstract
Sepsis is a devastating disease with high morbidity and mortality and no specific treatments. The pathophysiology of sepsis involves a hyperinflammatory response and release of damage-associated molecular patterns (DAMPs), including adenosine triphosphate (ATP), from activated and dying cells. Purinergic receptors activated by ATP have gained attention for their roles in sepsis, which can be pro- or anti-inflammatory depending on the context. Current data regarding the role of ATP-specific purinergic receptor P2X7 (P2X7R) in vascular function and inflammation during sepsis are conflicting, and its role on the endothelium has not been well characterized. In this study, we hypothesized that the P2X7R antagonist AZ 10606120 (AZ106) would prevent endothelial dysfunction during sepsis. As proof of concept, we first demonstrated the ability of AZ106 (10 µM) to prevent endothelial dysfunction in intact rat aorta in response to IL-1β, an inflammatory mediator upregulated during sepsis. Likewise, blocking P2X7R with AZ106 (10 µg/g) reduced the impairment of endothelial-dependent relaxation in mice subjected to intraperitoneal injection of cecal slurry (CS), a model of polymicrobial sepsis. However, contrary to our hypothesis, AZ106 did not improve microvascular permeability or injury, lung apoptosis, or illness severity in mice subjected to CS. Instead, AZ106 elevated spleen bacterial burden and circulating inflammatory markers. In conclusion, antagonism of P2X7R signaling during sepsis appears to disrupt the balance between its roles in inflammatory, antimicrobial, and vascular function.
Collapse
Affiliation(s)
- Jamie E. Meegan
- Division of Allergy, Pulmonary and Critical Care MedicineDepartment of MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Padmini Komalavilas
- Division of Vascular SurgeryVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Joyce Cheung‐Flynn
- Division of Vascular SurgeryVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Tsz Wing Yim
- Division of Vascular SurgeryVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Nathan D. Putz
- Division of Allergy, Pulmonary and Critical Care MedicineDepartment of MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Jordan J. Jesse
- Division of Allergy, Pulmonary and Critical Care MedicineDepartment of MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Kyle D. Smith
- Division of Allergy, Pulmonary and Critical Care MedicineDepartment of MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Tatiana N. Sidorova
- Division of Allergy, Pulmonary and Critical Care MedicineDepartment of MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Han Noo Ri Lee
- Division of Allergy, Pulmonary and Critical Care MedicineDepartment of MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Toria Tomasek
- Division of Allergy, Pulmonary and Critical Care MedicineDepartment of MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Ciara M. Shaver
- Division of Allergy, Pulmonary and Critical Care MedicineDepartment of MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Lorraine B. Ware
- Division of Allergy, Pulmonary and Critical Care MedicineDepartment of MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
- Department of Pathology, Microbiology and ImmunologyVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Colleen M. Brophy
- Division of Vascular SurgeryVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Julie A. Bastarache
- Division of Allergy, Pulmonary and Critical Care MedicineDepartment of MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
- Department of Pathology, Microbiology and ImmunologyVanderbilt University Medical CenterNashvilleTennesseeUSA
- Department of Cell and Developmental BiologyVanderbilt University Medical CenterNashvilleTennesseeUSA
| |
Collapse
|
23
|
Davenport P, Fan HH, Nolton E, Feldman HA, Lorenz V, Canas J, Acosta-Zaldívar M, Yakah W, Arthur C, Martin C, Stowell S, Koehler J, Mager D, Sola-Visner M. Platelet transfusions in a murine model of neonatal polymicrobial sepsis: Divergent effects on inflammation and mortality. Transfusion 2022; 62:1177-1187. [PMID: 35522536 PMCID: PMC11465244 DOI: 10.1111/trf.16895] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 03/11/2022] [Accepted: 03/18/2022] [Indexed: 12/19/2022]
Abstract
BACKGROUND Platelet transfusions (PTxs) are often given to septic preterm neonates at high platelet count thresholds in an attempt to reduce bleeding risk. However, the largest randomized controlled trial (RCT) of neonatal transfusion thresholds found higher mortality and/or major bleeding in infants transfused at higher thresholds. Using a murine model, we investigated the effects of adult PTx on neonatal sepsis-induced mortality, systemic inflammation, and platelet consumption. STUDY DESIGN AND METHODS Polymicrobial sepsis was induced via intraperitoneal injection of cecal slurry preparations (CS1, 2, 3) into P10 pups. Two hours after infection, pups were transfused with washed adult Green Flourescent Protein (GFP+) platelets or control. Weights, platelet counts, and GFP% were measured before 4 and 24 h post-infection. At 24 h, blood was collected for quantification of plasma cytokines. RESULTS The CS batches varied in 24 h mortality (11%, 73%, and 30% in CS1, 2, and 3, respectively), due to differences in bacterial composition. PTx had differential effects on sepsis-induced mortality and systemic inflammatory cytokines, increasing both in mice infected with CS1 (low mortality) and decreasing both in mice infected with CS2 and 3. In a mathematical model of platelet kinetics, the consumption of transfused adult platelets was higher than that of endogenous neonatal platelets, regardless of CS batch. DISCUSSION Our findings support the hypothesis that transfused adult platelets are consumed faster than endogenous neonatal platelets in sepsis and demonstrate that PTx can enhance or attenuate neonatal inflammation and mortality in a model of murine polymicrobial sepsis, depending on the composition of the inoculum and/or the severity of sepsis.
Collapse
Affiliation(s)
- Patricia Davenport
- Division of Newborn Medicine, Boston Children’s Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | - Hsuan-Hao Fan
- Department of Pharmaceutical Sciences, University of Buffalo, State University of New York, Buffalo, NY
| | - Emily Nolton
- Division of Newborn Medicine, Boston Children’s Hospital, Boston, MA
| | - Henry A. Feldman
- Division of Newborn Medicine, Boston Children’s Hospital, Boston, MA
- Harvard Medical School, Boston, MA
- Institutional Centers for Clinical and Translational Research, Boston Children’s Hospital, Boston, MA
| | - Viola Lorenz
- Division of Newborn Medicine, Boston Children’s Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | - Jorge Canas
- Division of Newborn Medicine, Boston Children’s Hospital, Boston, MA
| | | | - William Yakah
- Harvard Medical School, Boston, MA
- Division of Neonatology, Beth Israel Medical Center, Boston, MA
| | - Connie Arthur
- Harvard Medical School, Boston, MA
- Transfusion Medicine, Brigham and Women Hospital, Boston, MA
| | - Camilia Martin
- Harvard Medical School, Boston, MA
- Division of Neonatology, Beth Israel Medical Center, Boston, MA
| | - Sean Stowell
- Harvard Medical School, Boston, MA
- Transfusion Medicine, Brigham and Women Hospital, Boston, MA
| | - Julia Koehler
- Harvard Medical School, Boston, MA
- Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA
| | - Donald Mager
- Department of Pharmaceutical Sciences, University of Buffalo, State University of New York, Buffalo, NY
| | - Martha Sola-Visner
- Division of Newborn Medicine, Boston Children’s Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| |
Collapse
|
24
|
Tsuchida T, Wada T, Mizugaki A, Oda Y, Kayano K, Yamakawa K, Tanaka S. Protocol for a Sepsis Model Utilizing Fecal Suspension in Mice: Fecal Suspension Intraperitoneal Injection Model. Front Med (Lausanne) 2022; 9:765805. [PMID: 35646946 PMCID: PMC9134078 DOI: 10.3389/fmed.2022.765805] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 04/08/2022] [Indexed: 11/17/2022] Open
Abstract
Background Various animal models of sepsis have been developed to optimize sepsis treatment. However, therapeutic agents that were successful in animal models were rarely effective in human clinical trials. The cecal ligation and puncture (CLP) model is currently the gold standard for sepsis studies. However, its limitations include the high variability among researchers and the difficulty in comparing animals with different cecum shapes and sizes. In this study, we established a protocol for the creation of a simple and accessible sepsis rodent model using fecal suspensions that minimized differences in technical effects among researchers and individual differences in animals. Methods A mouse model of sepsis using fecal suspension intraperitoneal injection (FSI) was created using fresh stool excreted within 24 h. The collected fresh stool was dissolved in saline solution and filtered. The obtained fecal suspension was injected intraperitoneally into the mice. Moreover, fecal suspensions with different concentrations were prepared, and the survival rates were compared among the fecal suspensions for each concentration. To assess the validity of the FSI as a sepsis model, CLP and FSI with similar mortality rates were compared pathologically, physiologically, immunologically, and bacteriologically. Histopathological comparison was evaluated by hematoxylin-eosin and Gram staining of the parenchymal organs. Physiological evaluation was performed by comparing the respiratory rate, body temperature, and blood gas analysis results. Immunological assessment was performed using multiplex analysis. Bacteriological comparisons were performed by culturing ascites fluid. Results The FSI model increased mortality in proportion to the fecal suspension concentration. The mortality rate was reduced with antibiotic administration. In various comparative experiments conducted using the FSI and CLP models, both models showed findings consistent with sepsis. Furthermore, the FSI model showed less variability among the individuals in each test. Conclusion This is the first detailed and accurate report of a protocol for creating a sepsis model using fecal suspension. The FSI model is a minimally invasive and accessible sepsis rodent model. Its clinical validity as a sepsis model was proven via histological, physiological, microbiological, and immunological evaluation methods. The FSI model minimizes individual differences between mice and helps to conduct accurate studies after the onset of sepsis.
Collapse
Affiliation(s)
- Takumi Tsuchida
- Division of Acute and Critical Care Medicine, Department of Anesthesiology and Critical Care Medicine, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Takeshi Wada
- Division of Acute and Critical Care Medicine, Department of Anesthesiology and Critical Care Medicine, Faculty of Medicine, Hokkaido University, Sapporo, Japan
- *Correspondence: Takeshi Wada,
| | - Asumi Mizugaki
- Division of Acute and Critical Care Medicine, Department of Anesthesiology and Critical Care Medicine, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Yoshitaka Oda
- Department of Cancer Pathology, Faculty of Medicine, WPI-ICReDD, Hokkaido University, Sapporo, Japan
| | - Katsuhide Kayano
- Department of Emergency Medicine, Osaka Medical and Pharmaceutical University, Takatsuki, Japan
| | - Kazuma Yamakawa
- Department of Emergency Medicine, Osaka Medical and Pharmaceutical University, Takatsuki, Japan
| | - Shinya Tanaka
- Department of Cancer Pathology, Faculty of Medicine, WPI-ICReDD, Hokkaido University, Sapporo, Japan
| |
Collapse
|
25
|
Campion S, Inselman A, Hayes B, Casiraghi C, Joseph D, Facchinetti F, Salomone F, Schmitt G, Hui J, Davis-Bruno K, Van Malderen K, Morford L, De Schaepdrijver L, Wiesner L, Kourula S, Seo S, Laffan S, Urmaliya V, Chen C. The benefits, limitations and opportunities of preclinical models for neonatal drug development. Dis Model Mech 2022; 15:dmm049065. [PMID: 35466995 PMCID: PMC9066504 DOI: 10.1242/dmm.049065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Increased research to improve preclinical models to inform the development of therapeutics for neonatal diseases is an area of great need. This article reviews five common neonatal diseases - bronchopulmonary dysplasia, retinopathy of prematurity, necrotizing enterocolitis, perinatal hypoxic-ischemic encephalopathy and neonatal sepsis - and the available in vivo, in vitro and in silico preclinical models for studying these diseases. Better understanding of the strengths and weaknesses of specialized neonatal disease models will help to improve their utility, may add to the understanding of the mode of action and efficacy of a therapeutic, and/or may improve the understanding of the disease pathology to aid in identification of new therapeutic targets. Although the diseases covered in this article are diverse and require specific approaches, several high-level, overarching key lessons can be learned by evaluating the strengths, weaknesses and gaps in the available models. This Review is intended to help guide current and future researchers toward successful development of therapeutics in these areas of high unmet medical need.
Collapse
Affiliation(s)
- Sarah Campion
- Pfizer Worldwide Research, Development, and Medical, Groton, CT 06340, USA
| | - Amy Inselman
- U.S. Food and Drug Administration, National Center for Toxicological Research, Division of Systems Biology, Jefferson, AR 72079, USA
| | - Belinda Hayes
- U.S. Food and Drug Administration, Center for Drug Evaluation and Research, Office of New Drugs, Silver Spring, MD 20993, USA
| | - Costanza Casiraghi
- Department of Experimental Pharmacology and Translational Science, Chiesi Farmaceutici S.p.A., 43122 Parma, Italy
| | - David Joseph
- U.S. Food and Drug Administration, Center for Drug Evaluation and Research, Office of New Drugs, Silver Spring, MD 20993, USA
| | - Fabrizio Facchinetti
- Department of Experimental Pharmacology and Translational Science, Chiesi Farmaceutici S.p.A., 43122 Parma, Italy
| | - Fabrizio Salomone
- Department of Experimental Pharmacology and Translational Science, Chiesi Farmaceutici S.p.A., 43122 Parma, Italy
| | - Georg Schmitt
- Pharma Research and Early Development, Roche Innovation Center Basel, Pharmaceutical Sciences, F. Hoffmann-La Roche, 4070 Basel, Switzerland
| | - Julia Hui
- Bristol Myers Squibb, Nonclinical Research and Development, Summit, NJ 07901, USA
| | - Karen Davis-Bruno
- U.S. Food and Drug Administration, Center for Drug Evaluation and Research, Office of New Drugs, Silver Spring, MD 20993, USA
| | - Karen Van Malderen
- Federal Agency for Medicines and Health Products (FAMHP), Department DG PRE authorization, 1210 Brussels, Belgium
| | - LaRonda Morford
- Eli Lilly, Global Regulatory Affairs, Indianapolis, IN 46285, USA
| | | | - Lutz Wiesner
- Federal Institute for Drugs and Medical Devices, Clinical Trials, 53175 Bonn, Germany
| | - Stephanie Kourula
- Janssen R&D, Drug Metabolism & Pharmacokinetics, 2340 Beerse, Belgium
| | - Suna Seo
- U.S. Food and Drug Administration, Center for Drug Evaluation and Research, Office of New Drugs, Silver Spring, MD 20993, USA
| | - Susan Laffan
- GlaxoSmithKline, Non-Clinical Safety, Collegeville, PA 19406, USA
| | | | - Connie Chen
- Health and Environmental Sciences Institute, Washington, DC 20005, USA
| |
Collapse
|
26
|
Mechanisms and modulation of sepsis-induced immune dysfunction in children. Pediatr Res 2022; 91:447-453. [PMID: 34952937 PMCID: PMC9752201 DOI: 10.1038/s41390-021-01879-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 10/20/2021] [Accepted: 11/19/2021] [Indexed: 02/06/2023]
Abstract
Immunologic responses during sepsis vary significantly among patients and evolve over the course of illness. Sepsis has a direct impact on the immune system due to adverse alteration of the production, maturation, function, and apoptosis of immune cells. Dysregulation in both the innate and adaptive immune responses during sepsis leads to a range of phenotypes consisting of both hyperinflammation and immunosuppression that can result in immunoparalysis. In this review, we discuss components of immune dysregulation in sepsis, biomarkers and functional immune assays to aid in immunophenotyping patients, and evolving immunomodulatory therapies. Important research gaps for the future include: (1) Defining how age, host factors including prior exposures, and genetics impact the trajectory of sepsis in children, (2) Developing tools for rapid assessment of immune function in sepsis, and (3) Assessing how evolving pediatric sepsis endotypes respond differently to immunomodulation. Although multiple promising immunomodulatory agents exist or are in development, access to rapid immunophenotyping will be needed to identify which children are most likely to benefit from which therapy. Advancements in the ability to perform multidimensional endotyping will be key to developing a personalized approach to children with sepsis. IMPACT: Immunologic responses during sepsis vary significantly among patients and evolve over the course of illness. The resulting spectrum of immunoparalysis that can occur due to sepsis can increase morbidity and mortality in children and adults. This narrative review summarizes the current literature surrounding biomarkers and functional immunologic assays for immune dysregulation in sepsis, with a focus on immunomodulatory therapies that have been evaluated in sepsis. A precision approach toward diagnostic endotyping and therapeutics, including gene expression, will allow for optimal clinical trials to evaluate the efficacy of individualized and targeted treatments for pediatric sepsis.
Collapse
|
27
|
Pharmacological Effects of Marine-Derived Enterococcus faecium EA9 against Acute Lung Injury and Inflammation in Cecal Ligated and Punctured Septic Rats. BIOMED RESEARCH INTERNATIONAL 2021; 2021:5801700. [PMID: 34912891 PMCID: PMC8668278 DOI: 10.1155/2021/5801700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 09/12/2021] [Accepted: 11/16/2021] [Indexed: 11/25/2022]
Abstract
Microorganisms obtained from the marine environment may represent a potential therapeutic value for multiple diseases. This study explored the possible protective role of marine-derived potential probiotic Enterococcus faecium EA9 (E. faecium) against pulmonary inflammation and oxidative stress using the cecal ligation and puncture (CLP) model of sepsis in Wistar rats. Animals were pretreated with E. faecium for 10 days before either sham or CLP surgeries. Animals were sacrificed 72 hours following the surgical intervention. The histological architecture of lung tissues was evaluated as indicated by the lung injury score. In addition, the extend of pulmonary edema was determined as wet/dry weight ratio. The inflammatory cytokines were estimated in lung tissues, including tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), and interleukin-1 beta (IL-1β) using the enzyme-linked-immunosorbent-assay (ELISA) technique. Moreover, markers for lipid peroxidation such as thiobarbituric acid reaction substances (TBARs), and endogenous antioxidants, including reduced glutathione (GSH) were determined in lung tissues. Finally, the enzymatic activities of antioxidant enzymes such as catalase (CAT), superoxide dismutase (SOD), glutathione peroxidase (GPx), and glutathione reductase (GR) were assayed in the lungs. Pretreatment with E. faecium markedly attenuated CLP-induced lung injury and pulmonary edema. Markers for inflammation, including TNF-α, IL-6, and IL-1β were augmented in the lung tissues of CLP animals, while E. faecium ameliorated their augmented levels. E. faecium pretreatment also restored the elevated TBARS levels and the prohibited CAT, SOD, and GPx enzymatic activities in CLP animals. GSH levels were corrected by E. faecium in CLP animals. The inflammatory and lipid peroxidation mediators were positively correlated, while antioxidant enzymatic activities were negatively correlated with CLP-induced lung injury and pulmonary edema. Collectively, marine-derived Enterococcus faecium EA9 might be considered as a prospective therapeutic tool for the management of pulmonary dysfunction associated with sepsis.
Collapse
|
28
|
Nakasone R, Ashina M, Kido T, Miyauchi H, Saito M, Inoue S, Shinohara M, Nozu K, Fujioka K. Protective Role of an Initial Low-Dose Septic Challenge against Lethal Sepsis in Neonatal Mice: A Pilot Study. J Clin Med 2021; 10:jcm10245823. [PMID: 34945120 PMCID: PMC8705039 DOI: 10.3390/jcm10245823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/10/2021] [Accepted: 12/10/2021] [Indexed: 11/16/2022] Open
Abstract
Neonatal sepsis is characterized by systemic bacterial invasion followed by a massive inflammatory response. At present, no therapeutic strategy has been found that significantly reduces the mortality of neonatal sepsis. We aimed to investigate the protective role of an initial low-dose septic challenge for the prevention of subsequent lethal sepsis in a mouse model. A stock cecal slurry (CS) solution was prepared from adult ceca. The LD83 (1.5 mg CS/g) was used for all animals. An initial challenge of normal saline (NS) or 0.5 mg CS/g (non-lethal dose) was administered at four days of age, then 1.5 mg CS/g was administered intraperitoneally at seven days of age (72 h post-initial challenge), and survival was monitored. Initial exposure to NS (n = 10) resulted in 90% mortality following exposure to the LD83 CS dose in contrast to an initial exposure to CS (n = 16), which significantly decreased mortality to 6% (p < 0.0001), reduced blood bacterial counts, attenuated inflammatory responses, and suppressed lipid mediators. Initial exposure to a non-lethal CS dose prior to exposure to a lethal CS dose significantly reduces sepsis mortality, a protective effect that might be mediated by modulating abnormal systemic inflammatory responses.
Collapse
Affiliation(s)
- Ruka Nakasone
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (R.N.); (M.A.); (T.K.); (K.N.)
| | - Mariko Ashina
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (R.N.); (M.A.); (T.K.); (K.N.)
| | - Takumi Kido
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (R.N.); (M.A.); (T.K.); (K.N.)
| | - Harunori Miyauchi
- Department of Pediatric Surgery, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan;
| | - Masafumi Saito
- Department of Disaster and Emergency and Critical Care Medicine, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (M.S.); (S.I.)
| | - Shigeaki Inoue
- Department of Disaster and Emergency and Critical Care Medicine, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (M.S.); (S.I.)
| | - Masakazu Shinohara
- Division of Epidemiology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan;
- The Integrated Center for Mass Spectrometry, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Kandai Nozu
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (R.N.); (M.A.); (T.K.); (K.N.)
| | - Kazumichi Fujioka
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (R.N.); (M.A.); (T.K.); (K.N.)
- Correspondence: ; Tel.: +81-78-382-6090
| |
Collapse
|
29
|
Li B, Niu S, Geng H, Yang C, Zhao C. Berberine Attenuates Neonatal Sepsis in Mice By Inhibiting FOXA1 and NF-κB Signal Transduction Via the Induction of MiR-132-3p. Inflammation 2021; 44:2395-2406. [PMID: 34523052 DOI: 10.1007/s10753-021-01510-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 06/25/2021] [Indexed: 01/23/2023]
Abstract
Neonatal sepsis (NS) is a severe syndrome in newborns that is induced by infections, and the initiation and development of NS are closely associated with the function of miRs. In the current study, the effects of berberine, which is a functional component in traditional Chinese medicine (TCM), against NS were assessed by focusing on the interaction of berberine with miR-132-3p-mediated signaling. An NS model was induced using cecal slurry (CS) in vivo and LPS in vitro, and berberine treatment was applies. The changes in survival rate, intestinal structure, and systemic inflammation in mice and the viability, apoptosis, and inflammatory response in intestinal cells were measured. At the molecular level, miR-132-3p levels and the activities of the FOXA1 and NF-κB pathways were analyzed. The data showed that berberine increased the survival rates of CS-induced mice. The intestinal injuries induced by CS were also attenuated by berberine, which was associated with inhibition of the production of systemic IL-6, IL-1β, and TNF-α. At the molecular level, the expression of miR-132-3p was upregulated, suppressing the expression of FOXA1, p-IκBα, and p65 while inducing the expression of IκBα. The effects of berberine on NS-induced impairments were blocked by the injection of the miR-132-3p antagomir, which exacerbated intestinal injuries, induced systemic inflammation, and reactivated the FOXA1 and NF-κB pathways. The findings in the in vivo model were validated with in vitro assays. Collectively, the findings outlined in the current study indicated that berberine had solid protective effects against NS-induced symptoms in newborn mice, and the effects depended on the upregulation of miR-132-3p.
Collapse
Affiliation(s)
- Binghui Li
- Maternal and Child Health Care Hospital of Zibo, Zhangdian District, No. 66 Tianjin South Road, 255000, Zibo, Shandong, China
| | - Shiping Niu
- Maternal and Child Health Care Hospital of Zibo, Zhangdian District, No. 66 Tianjin South Road, 255000, Zibo, Shandong, China
| | - Hongli Geng
- Maternal and Child Health Care Hospital of Zibo, Zhangdian District, No. 66 Tianjin South Road, 255000, Zibo, Shandong, China
| | - Changkun Yang
- Maternal and Child Health Care Hospital of Zibo, Zhangdian District, No. 66 Tianjin South Road, 255000, Zibo, Shandong, China
| | - Changliang Zhao
- Maternal and Child Health Care Hospital of Zibo, Zhangdian District, No. 66 Tianjin South Road, 255000, Zibo, Shandong, China.
| |
Collapse
|
30
|
Campbell RA, Campbell HD, Bircher JS, de Araujo CV, Denorme F, Crandell JL, Rustad JL, Monts J, Cody MJ, Kosaka Y, Yost CC. Placental HTRA1 cleaves α1-antitrypsin to generate a NET-inhibitory peptide. Blood 2021; 138:977-988. [PMID: 34192300 PMCID: PMC9069473 DOI: 10.1182/blood.2020009021] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 06/30/2021] [Accepted: 06/13/2021] [Indexed: 11/20/2022] Open
Abstract
Neutrophil extracellular traps (NETs) are important components of innate immunity. Neonatal neutrophils (polymorphonuclear leukocytes [PMNs]) fail to form NETs due to circulating NET-inhibitory peptides (NIPs), cleavage fragments of α1-antitrypsin (A1AT). How fetal and neonatal blood NIPs are generated remains unknown, however. The placenta expresses high-temperature requirement serine protease A1 (HTRA1) during fetal development, which can cleave A1AT. We hypothesized that placentally expressed HTRA1 regulates the formation of NIPs and that NET competency changed in PMNs isolated from neonatal HTRA1 knockout mice (HTRA1-/-). We found that umbilical cord blood plasma has elevated HTRA1 levels compared with adult plasma and that recombinant and placenta-eluted HTRA1 cleaves A1AT to generate an A1AT cleavage fragment (A1ATM383S-CF) of molecular weight similar to previously identified NIPs that block NET formation by adult neutrophils. We showed that neonatal mouse pup plasma contains A1AT fragments that inhibit NET formation by PMNs isolated from adult mice, indicating that NIP generation during gestation is conserved across species. Lipopolysaccharide-stimulated PMNs isolated from HTRA1+/+ littermate control pups exhibit delayed NET formation after birth. However, plasma from HTRA1-/- pups had no detectable NIPs, and PMNs from HTRA1-/- pups became NET competent earlier after birth compared with HTRA1+/+ littermate controls. Finally, in the cecal slurry model of neonatal sepsis, A1ATM383S-CF improved survival in C57BL/6 pups by preventing pathogenic NET formation. Our data indicate that placentally expressed HTRA1 is a serine protease that cleaves A1AT in utero to generate NIPs that regulate NET formation by human and mouse PMNs.
Collapse
Affiliation(s)
- Robert A Campbell
- University of Utah Molecular Medicine Program, Salt Lake City, UT; and
- Department of Internal Medicine
| | | | | | | | - Frederik Denorme
- University of Utah Molecular Medicine Program, Salt Lake City, UT; and
| | - Jacob L Crandell
- University of Utah Molecular Medicine Program, Salt Lake City, UT; and
| | - John L Rustad
- University of Utah Molecular Medicine Program, Salt Lake City, UT; and
| | - Josh Monts
- Flow Cytometry Core, University of Utah, Salt Lake City, UT
| | - Mark J Cody
- University of Utah Molecular Medicine Program, Salt Lake City, UT; and
- Department of Pediatrics, and
| | - Yasuhiro Kosaka
- University of Utah Molecular Medicine Program, Salt Lake City, UT; and
| | - Christian C Yost
- University of Utah Molecular Medicine Program, Salt Lake City, UT; and
- Department of Pediatrics, and
| |
Collapse
|
31
|
Brook B, Harbeson DJ, Shannon CP, Cai B, He D, Ben-Othman R, Francis F, Huang J, Varankovich N, Liu A, Bao W, Bjerregaard-Andersen M, Schaltz-Buchholzer F, Sanca L, Golding CN, Larsen KL, Levy O, Kampmann B, Tan R, Charles A, Wynn JL, Shann F, Aaby P, Benn CS, Tebbutt SJ, Kollmann TR, Amenyogbe N. BCG vaccination-induced emergency granulopoiesis provides rapid protection from neonatal sepsis. Sci Transl Med 2021; 12:12/542/eaax4517. [PMID: 32376769 DOI: 10.1126/scitranslmed.aax4517] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 03/13/2020] [Accepted: 04/16/2020] [Indexed: 12/11/2022]
Abstract
Death from sepsis in the neonatal period remains a serious threat for millions. Within 3 days of administration, bacille Calmette-Guérin (BCG) vaccination can reduce mortality from neonatal sepsis in human newborns, but the underlying mechanism for this rapid protection is unknown. We found that BCG was also protective in a mouse model of neonatal polymicrobial sepsis, where it induced granulocyte colony-stimulating factor (G-CSF) within hours of administration. This was necessary and sufficient to drive emergency granulopoiesis (EG), resulting in a marked increase in neutrophils. This increase in neutrophils was directly and quantitatively responsible for protection from sepsis. Rapid induction of EG after BCG administration also occurred in three independent cohorts of human neonates.
Collapse
Affiliation(s)
- Byron Brook
- Department of Experimental Medicine, University of British Columbia, 2775 Laurel Street, 10th Floor, Room 10117, Vancouver, BC V5Z 1M9, Canada
| | - Danny J Harbeson
- Department of Experimental Medicine, University of British Columbia, 2775 Laurel Street, 10th Floor, Room 10117, Vancouver, BC V5Z 1M9, Canada
| | - Casey P Shannon
- PROOF Centre of Excellence, British Columbia, 10th floor, 1190 Hornby Street, Vancouver, BC V6Z 2K5, Canada.,UBC Centre for Heart Lung Innovation, St. Paul's Hospital, 1081 Burrard Street, Vancouver, BC V6Z 1Y6, Canada
| | - Bing Cai
- Department of Pediatrics, University of British Columbia, and BC Children's Hospital, 4480 Oak Street, Vancouver, BC V6H 3V4, Canada
| | - Daniel He
- Department of Experimental Medicine, University of British Columbia, 2775 Laurel Street, 10th Floor, Room 10117, Vancouver, BC V5Z 1M9, Canada.,PROOF Centre of Excellence, British Columbia, 10th floor, 1190 Hornby Street, Vancouver, BC V6Z 2K5, Canada.,UBC Centre for Heart Lung Innovation, St. Paul's Hospital, 1081 Burrard Street, Vancouver, BC V6Z 1Y6, Canada
| | - Rym Ben-Othman
- Department of Pediatrics, University of British Columbia, and BC Children's Hospital, 4480 Oak Street, Vancouver, BC V6H 3V4, Canada
| | - Freddy Francis
- Department of Experimental Medicine, University of British Columbia, 2775 Laurel Street, 10th Floor, Room 10117, Vancouver, BC V5Z 1M9, Canada
| | - Joe Huang
- Department of Pediatrics, University of British Columbia, and BC Children's Hospital, 4480 Oak Street, Vancouver, BC V6H 3V4, Canada
| | - Natallia Varankovich
- Department of Pediatrics, University of British Columbia, and BC Children's Hospital, 4480 Oak Street, Vancouver, BC V6H 3V4, Canada
| | - Aaron Liu
- Department of Experimental Medicine, University of British Columbia, 2775 Laurel Street, 10th Floor, Room 10117, Vancouver, BC V5Z 1M9, Canada
| | - Winnie Bao
- Department of Pediatrics, University of British Columbia, and BC Children's Hospital, 4480 Oak Street, Vancouver, BC V6H 3V4, Canada
| | - Morten Bjerregaard-Andersen
- Bandim Health Project, Indepth Network, Apartado 861, 1004 Bissau, Guinea-Bissau.,Research Center for Vitamins and Vaccines (CVIVA), Statens Serum Institut (SSI), Artillerivej 5, 2300 Copenhagen S, Denmark.,Department of Endocrinology, Odense University Hospital, Kløvervænget 6, 5000 Odense C, Denmark
| | - Frederik Schaltz-Buchholzer
- Bandim Health Project, Indepth Network, Apartado 861, 1004 Bissau, Guinea-Bissau.,Research Center for Vitamins and Vaccines (CVIVA), Statens Serum Institut (SSI), Artillerivej 5, 2300 Copenhagen S, Denmark.,OPEN, Institute of Clinical Research and Danish Institute for Advanced Science, University of Southern Denmark, and Odense University Hospital, J.B. Winsløws Vej, 5000 Odense C, Denmark
| | - Lilica Sanca
- Bandim Health Project, Indepth Network, Apartado 861, 1004 Bissau, Guinea-Bissau
| | - Christian N Golding
- Bandim Health Project, Indepth Network, Apartado 861, 1004 Bissau, Guinea-Bissau.,Research Center for Vitamins and Vaccines (CVIVA), Statens Serum Institut (SSI), Artillerivej 5, 2300 Copenhagen S, Denmark
| | - Kristina Lindberg Larsen
- Bandim Health Project, Indepth Network, Apartado 861, 1004 Bissau, Guinea-Bissau.,Research Center for Vitamins and Vaccines (CVIVA), Statens Serum Institut (SSI), Artillerivej 5, 2300 Copenhagen S, Denmark
| | - Ofer Levy
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA 02115, USA.,Harvard Medical School, Boston, MA 02115, USA.,Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Beate Kampmann
- Vaccines and Immunity Theme, Medical Research Council Unit, The Gambia at the London School of Hygiene and Tropical Medicine, Atlantic Boulevard, P.O. Box 273, Banjul, The Gambia.,Vaccine Centre, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London WC1E 7HT, UK
| | | | - Rusung Tan
- Department of Pathology, Sidra Medicine and Weill Cornell Medicine, Doha, Qatar
| | - Adrian Charles
- Department of Pathology, Sidra Medicine and Weill Cornell Medicine, Doha, Qatar
| | - James L Wynn
- Department of Paediatrics and Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, P.O. Box 100296, Gainesville, FL 32610-0296, USA
| | - Frank Shann
- Department of Paediatrics, University of Melbourne, Melbourne, VIC 3052, Australia
| | - Peter Aaby
- Bandim Health Project, Indepth Network, Apartado 861, 1004 Bissau, Guinea-Bissau
| | - Christine S Benn
- Bandim Health Project, Indepth Network, Apartado 861, 1004 Bissau, Guinea-Bissau.,Research Center for Vitamins and Vaccines (CVIVA), Statens Serum Institut (SSI), Artillerivej 5, 2300 Copenhagen S, Denmark.,OPEN, Institute of Clinical Research and Danish Institute for Advanced Science, University of Southern Denmark, and Odense University Hospital, J.B. Winsløws Vej, 5000 Odense C, Denmark
| | - Scott J Tebbutt
- PROOF Centre of Excellence, British Columbia, 10th floor, 1190 Hornby Street, Vancouver, BC V6Z 2K5, Canada.,UBC Centre for Heart Lung Innovation, St. Paul's Hospital, 1081 Burrard Street, Vancouver, BC V6Z 1Y6, Canada.,Department of Medicine, Division of Respiratory Medicine, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| | - Tobias R Kollmann
- Department of Experimental Medicine, University of British Columbia, 2775 Laurel Street, 10th Floor, Room 10117, Vancouver, BC V5Z 1M9, Canada. .,Department of Pediatrics, University of British Columbia, and BC Children's Hospital, 4480 Oak Street, Vancouver, BC V6H 3V4, Canada.,Telethon Kids Institute, 100 Roberts Road, Subiaco, Western Australia 6008, Australia
| | - Nelly Amenyogbe
- Department of Experimental Medicine, University of British Columbia, 2775 Laurel Street, 10th Floor, Room 10117, Vancouver, BC V5Z 1M9, Canada. .,Telethon Kids Institute, 100 Roberts Road, Subiaco, Western Australia 6008, Australia
| |
Collapse
|
32
|
Exploring Clinically-Relevant Experimental Models of Neonatal Shock and Necrotizing Enterocolitis. Shock 2021; 53:596-604. [PMID: 31977960 DOI: 10.1097/shk.0000000000001507] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Neonatal shock and necrotizing enterocolitis (NEC) are leading causes of morbidity and mortality in premature infants. NEC is a life-threatening gastrointestinal illness, the precise etiology of which is not well understood, but is characterized by an immaturity of the intestinal barrier, altered function of the adaptive immune system, and intestinal dysbiosis. The complexities of NEC and shock in the neonatal population necessitate relevant clinical modeling using newborn animals that mimic the disease in human neonates to better elucidate the pathogenesis and provide an opportunity for the discovery of potential therapeutics. A wide variety of animal species-including rats, mice, piglets, and primates-have been used in developing experimental models of neonatal diseases such as NEC and shock. This review aims to highlight the immunologic differences in neonates compared with adults and provide an assessment of the advantages and drawbacks of established animal models of both NEC and shock using enteral or intraperitoneal induction of bacterial pathogens. The selection of a model has benefits unique to each type of animal species and provides individual opportunities for the development of targeted therapies. This review discusses the clinical and physiologic relevance of animal models and the insight they contribute to the complexities of the specific neonatal diseases: NEC and shock.
Collapse
|
33
|
Fallon EA, Chung CS, Heffernan DS, Chen Y, De Paepe ME, Ayala A. Survival and Pulmonary Injury After Neonatal Sepsis: PD1/PDL1's Contributions to Mouse and Human Immunopathology. Front Immunol 2021; 12:634529. [PMID: 33746973 PMCID: PMC7965961 DOI: 10.3389/fimmu.2021.634529] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Accepted: 02/08/2021] [Indexed: 12/14/2022] Open
Abstract
Morbidity and mortality associated with neonatal sepsis remains a healthcare crisis. PD1-/- neonatal mice endured experimental sepsis, in the form of cecal slurry (CS), and showed improved rates of survival compared to wildtype (WT) counterparts. End-organ injury, particularly of the lung, contributes to the devastation set forth by neonatal sepsis. PDL1-/- neonatal mice, in contrast to PD1-/- neonatal mice did not have a significant improvement in survival after CS. Because of this, we focused subsequent studies on the impact of PD1 gene deficiency on lung injury. Here, we observed that at 24 h post-CS (but not at 4 or 12 h) there was a marked increase in pulmonary edema (PE), neutrophil influx, myeloperoxidase (MPO) levels, and cytokine expression sham (Sh) WT mice. Regarding pulmonary endothelial cell (EC) adhesion molecule expression, we observed that Zona occludens-1 (ZO-1) within the cell shifted from a membranous location to a peri-nuclear location after CS in WT murine cultured ECs at 24hrs, but remained membranous among PD1-/- lungs. To expand the scope of this inquiry, we investigated human neonatal lung tissue. We observed that the lungs of human newborns exposed to intrauterine infection had significantly higher numbers of PD1+ cells compared to specimens who died from non-infectious causes. Together, these data suggest that PD1/PDL1, a pathway typically thought to govern adaptive immune processes in adult animals, can modulate the largely innate neonatal pulmonary immune response to experimental septic insult. The potential future significance of this area of study includes that PD1/PDL1 checkpoint proteins may be viable therapeutic targets in the septic neonate.
Collapse
Affiliation(s)
- Eleanor A. Fallon
- Division of Surgical Research, Department of Surgery, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI, United States
| | - Chun-Shiang Chung
- Division of Surgical Research, Department of Surgery, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI, United States
| | - Daithi S. Heffernan
- Division of Surgical Research, Department of Surgery, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI, United States
- Department of Surgery, Providence Veterans Affairs Medical Center, Providence, RI, United States
| | - Yaping Chen
- Division of Surgical Research, Department of Surgery, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI, United States
| | - Monique E. De Paepe
- Department of Pathology, Women & Infants Hospital and Alpert Medical School of Brown University, Providence, RI, United States
| | - Alfred Ayala
- Division of Surgical Research, Department of Surgery, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI, United States
| |
Collapse
|
34
|
He W, Xiao K, Xu J, Guan W, Xie S, Wang K, Yan P, Fang M, Xie L. Recurrent Sepsis Exacerbates CD4 + T Cell Exhaustion and Decreases Antiviral Immune Responses. Front Immunol 2021; 12:627435. [PMID: 33717146 PMCID: PMC7946831 DOI: 10.3389/fimmu.2021.627435] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 01/18/2021] [Indexed: 12/29/2022] Open
Abstract
Sepsis is a life-threatening organ dysfunction caused by a dysregulated host response to an infection. It is a disease with a high incidence, mortality, and recurrence rate and frequently results in its survivors requiring readmission into hospitals. The readmission is mainly due to recurrent sepsis. Patients with recurrent sepsis are more susceptible to secondary infections partly due to immune dysfunction, leading to a higher mortality in the long term. However, there remains a gap in the understanding of immunological characteristics and underlying mechanisms of recurrent sepsis. In this study, we used mouse models of acute and recurrent sepsis to investigate their different immunological characteristics. And then we subjected the two mouse models to a secondary influenza A virus (H1N1) infection and characterized the different immune responses. Here, we demonstrated that CD4+ T cells present an exacerbated exhaustion phenotype in response to recurrent sepsis as illustrated by the decreased frequency of CD4+ T cells, reduced co-stimulatory CD28 and increased inhibitory PD-1 and Tim-3 expression on CD4+ T cells, increased frequency of regulatory T cells, and reduced MHC-II expression on antigen-presenting cells. Moreover, we showed that antiviral immune responses decrease in the recurrent sepsis mouse model subjected to a secondary infection as illustrated by the reduced pathogen clearance and inflammatory response. This may be a consequence of the exacerbated CD4+ T cell exhaustion. In summary, recurrent sepsis exacerbates CD4+ T cell exhaustion and decreases antiviral immune responses, contributing to significant morbidity, increased late mortality, and increased health care burden in recurrent sepsis patients.
Collapse
Affiliation(s)
- Wanxue He
- College of Pulmonary and Critical Care Medicine, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Kun Xiao
- College of Pulmonary and Critical Care Medicine, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Jiaruo Xu
- College of Pulmonary and Critical Care Medicine, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Wei Guan
- College of Pulmonary and Critical Care Medicine, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Sheling Xie
- College of Pulmonary and Critical Care Medicine, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Kaifei Wang
- College of Pulmonary and Critical Care Medicine, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Peng Yan
- College of Pulmonary and Critical Care Medicine, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Min Fang
- Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Lixin Xie
- College of Pulmonary and Critical Care Medicine, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| |
Collapse
|
35
|
Chen Q, Zheng J, Wang D, Liu Q, Kang L, Gao X, Lin Z. Nitrosonisoldipine is a selective inhibitor of inflammatory caspases and protects against pyroptosis and related septic shock. Eur J Immunol 2021; 51:1234-1245. [PMID: 33454984 DOI: 10.1002/eji.202048937] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 12/02/2020] [Accepted: 01/15/2021] [Indexed: 12/14/2022]
Abstract
Pyroptosis is a type of acute cell death that mainly occurs in immune cells. It is characterized with robust release of inflammatory cytokines and has emerged to play a critical role in the pathogenesis of sepsis-associated immune disorders. In this study, we screened for pyroptotic inhibitors with the ultimate goal to benefit sepsis treatments. Accidentally, we identified that nitrosonisoldipine (NTS), a photodegradation product of calcium channel inhibitor nisoldipine, inhibits noncanonical pyroptosis. Using murine immortalized BM-derived macrophage and human THP-1 cell line, we further discovered that NTS not only inhibits noncanonical pyroptosis mediated by caspase-11 or caspase-4 but also canonical pyroptosis mediated by caspase-1. Mechanistically, NTS directly inhibits the enzyme activities of these inflammatory caspases, and these inhibitory effects persist despite extensive washout of the drug. By contrast, apoptosis mediated by caspase-3/-7 was not affected by NTS. Mice pretreated with NTS intraperitoneally displayed improved survival rate and extended survival time in LPS- and polymicrobe-induced septic models, respectively. In conclusion, NTS is a selective inhibitor of inflammatory caspases that blocks both the noncanonical and canonical pyroptotic pathways. It is safe for intraperitoneal administration and might be used as a prototype to develop drugs for sepsis treatments.
Collapse
Affiliation(s)
- Qianyue Chen
- MOE Key Laboratory of Model Animals for Disease Study, Model Animal Research Center, State Key Laboratory of Pharmaceutical Biotechnology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing University, Nanjing, China
| | - Jiashuo Zheng
- Helmholtz Zentrum München, Institute of Metabolism and Cell Death, Neuherberg, Germany
| | - Dingyu Wang
- Jiangsu GemPharmatech Co., Ltd., Nanjing, China
| | - Qiyao Liu
- MOE Key Laboratory of Model Animals for Disease Study, Model Animal Research Center, State Key Laboratory of Pharmaceutical Biotechnology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing University, Nanjing, China
| | - Lulu Kang
- MOE Key Laboratory of Model Animals for Disease Study, Model Animal Research Center, State Key Laboratory of Pharmaceutical Biotechnology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing University, Nanjing, China
| | - Xiang Gao
- MOE Key Laboratory of Model Animals for Disease Study, Model Animal Research Center, State Key Laboratory of Pharmaceutical Biotechnology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing University, Nanjing, China
| | - Zhaoyu Lin
- MOE Key Laboratory of Model Animals for Disease Study, Model Animal Research Center, State Key Laboratory of Pharmaceutical Biotechnology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing University, Nanjing, China
| |
Collapse
|
36
|
Rincon JC, Hawkins RB, Hollen M, Nacionales DC, Ungaro R, Efron PA, Moldawer LL, Larson SD. Aluminum Adjuvant Improves Survival Via NLRP3 Inflammasome and Myeloid Non-Granulocytic Cells in a Murine Model of Neonatal Sepsis. Shock 2021; 55:274-282. [PMID: 32769820 PMCID: PMC8025597 DOI: 10.1097/shk.0000000000001623] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
ABSTRACT Neonatal sepsis leads to significant morbidity and mortality with the highest risk of death occurring in preterm (<37 weeks) and low birth weight (<2,500 g) infants. The neonatal immune system is developmentally immature with well-described defects in innate and adaptive immune responses. Immune adjuvants used to enhance the vaccine response have emerged as potential therapeutic options, stimulating non-specific immunity and preventing sepsis mortality. Aluminum salts ("alum") have been used as immune adjuvants for over a century, but their mechanism of action remains poorly understood. This study aims to identify potential mechanisms by which pretreatment with alum induces host protective immunity to polymicrobial sepsis in neonatal mice. Utilizing genetic and cell-depletion studies, we demonstrate here that the prophylactic administration of aluminum adjuvants in neonatal mice improves sepsis survival via activation of the nucleotide oligomerization domain-like receptor family, pyrin-domain-containing 3 inflammasome and dendritic cell activation. Furthermore, this beneficial effect is dependent on myeloid, non-granulocytic Gr1-positive cells, and MyD88-signaling pathway activation. These findings suggest a promising therapeutic role for aluminum-based vaccine adjuvants to prevent development of neonatal sepsis and improve mortality in this highly vulnerable population.
Collapse
Affiliation(s)
- Jaimar C Rincon
- Department of Surgery, University of Florida College of Medicine, Gainesville, Florida
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Abstract
Aging is a grave problem in sepsis, and T cell exhaustion is the main cause of sepsis-induced immunosuppression. Sepsis- and aging-induced T cell exhaustion is related to secondary infection with a poor long-term outcome in the elderly. However, the trend, impact, and mechanism of T cell exhaustion are still unclear. Interleukin (IL)-15 improves survival rate of septic mice via its antiapoptotic effect on T cells; however, it is still unclear how IL-15 reverses prolonged T cell exhaustion in aged septic mice. The purpose of this study was to clarify the trend of sepsis-induced T cell exhaustion and whether IL-15 prevents aging-induced persistent T cell exhaustion in septic mice. Preserved cecal slurry was injected intraperitoneally into young (6-week-old) and aged mice (18-24-month-old) 4 times, to induce clinically relevant repeated sepsis. IL-15 (1.5 μg) or phosphate-buffered saline was injected subcutaneously 3 times, body weight was serially measured, and peripheral blood cells from their cheek were serially collected for 50 days. Sepsis-induced T cell exhaustion was significantly severe in aged mice than in young mice and was accompanied with decreased naive CD4 and CD8 T cells (P < 0.01) and increased expression of program death 1 on T cell (P < 0.01) and regulatory T cell population (P < 0.01). IL-15 significantly improved sepsis-induced T exhaustion, with significantly increased numbers of natural killer cells and macrophages, and significantly enhanced phagocytosis activity in aged septic mice (P < 0.05). It decreased the long-term mortality associated with sepsis survivors by improving T cell exhaustion over an extended duration and also ameliorated aging-induced persistent T cell exhaustion in septic mice.
Collapse
|
38
|
Steinhagen F, Hilbert T, Cramer N, Senzig S, Parcina M, Bode C, Boehm O, Frede S, Klaschik S. Development of a minimal invasive and controllable murine model to study polymicrobial abdominal sepsis. ALL LIFE 2021. [DOI: 10.1080/26895293.2021.1909663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Affiliation(s)
- Folkert Steinhagen
- Department of Anesthesiology and Critical Care Medicine, University Hospital Bonn, Bonn, Germany
| | - Tobias Hilbert
- Department of Anesthesiology and Critical Care Medicine, University Hospital Bonn, Bonn, Germany
| | - Nina Cramer
- Department of Anesthesiology and Critical Care Medicine, University Hospital Bonn, Bonn, Germany
| | - Sebastian Senzig
- Department of Anesthesiology and Critical Care Medicine, University Hospital Bonn, Bonn, Germany
| | - Marijo Parcina
- Department of Medical Microbiology, Immunology and Parasitology (IMMIP), University Hospital Bonn, Bonn, Germany
| | - Christian Bode
- Department of Anesthesiology and Critical Care Medicine, University Hospital Bonn, Bonn, Germany
| | - Olaf Boehm
- Department of Anesthesiology and Critical Care Medicine, University Hospital Bonn, Bonn, Germany
| | - Stilla Frede
- Department of Anesthesiology and Critical Care Medicine, University Hospital Bonn, Bonn, Germany
| | - Sven Klaschik
- Department of Anesthesiology and Critical Care Medicine, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
39
|
Abstract
Studying the pathophysiology of sepsis still requires animal models, and the mouse remains the most commonly used species. Here we discuss the "cecal slurry" (CS) model of polymicrobial, peritoneal sepsis and compare and contrast it to other commonly used methods. Among the different murine models of sepsis, cecal ligation and puncture (CLP), and not the CS, is often considered the "gold standard" to induce polymicrobial sepsis in laboratory animals. CLP is a well-described model involving a simple surgical procedure that closely mimics the clinical course of intra-abdominal sepsis. However, CLP may not be an option for experiments involving newborn pups, where the cecum is indistinguishable from small bowel, where differences in microbiome content may affect the experiment, or where surgical procedures/anesthesia exposure needs to be limited. An important alternative method is the CS model, involving the intraperitoneal injection of cecal contents from a donor animal into the peritoneal cavity of a recipient animal to induce polymicrobial sepsis. Furthermore, CS is an effective alternative model of intraperitoneal polymicrobial sepsis in adult mice and can now be considered the "gold standard" for experiments in neonatal mice.
Collapse
|
40
|
Denning NL, Aziz M, Diao L, Prince JM, Wang P. Targeting the eCIRP/TREM-1 interaction with a small molecule inhibitor improves cardiac dysfunction in neonatal sepsis. Mol Med 2020; 26:121. [PMID: 33276725 PMCID: PMC7716442 DOI: 10.1186/s10020-020-00243-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 11/12/2020] [Indexed: 02/06/2023] Open
Abstract
Background Neonatal sepsis and the associated myocardial dysfunction remain a leading cause of infant mortality. Extracellular cold-inducible RNA-binding protein (eCIRP) acts as a ligand of triggering receptor expressed on myeloid cells-1 (TREM-1). M3 is a small CIRP-derived peptide that inhibits the eCIRP/TREM-1 interaction. We hypothesize that the eCIRP/TREM-1 interaction in cardiomyocytes contributes to sepsis-induced cardiac dysfunction in neonatal sepsis, while M3 is cardioprotective. Methods Serum was collected from neonates in the Neonatal Intensive Care Unit (NICU). 5–7-day old C57BL/6 mouse pups were used in this study. Primary murine neonatal cardiomyocytes were stimulated with recombinant murine (rm) CIRP with M3. TREM-1 mRNA and supernatant cytokine levels were assayed. Mitochondrial oxidative stress, ROS, and membrane potential were assayed. Neonatal mice were injected with rmCIRP and speckle-tracking echocardiography was conducted to measure cardiac strain. Sepsis was induced by i.p. cecal slurry. Mouse pups were treated with M3 or vehicle. After 16 h, echocardiography was performed followed by euthanasia for tissue analysis. A 7-day survival study was conducted. Results Serum eCIRP levels were elevated in septic human neonates. rmCIRP stimulation of cardiomyocytes increased TREM-1 gene expression. Stimulation of cardiomyocytes with rmCIRP upregulated TNF-α and IL-6 in the supernatants, while this upregulation was inhibited by M3. Stimulation of cardiomyocytes with rmCIRP resulted in a reduction in mitochondrial membrane potential (MMP) while M3 treatment returned MMP to near baseline. rmCIRP caused mitochondrial calcium overload; this was inhibited by M3. rmCIRP injection impaired longitudinal and radial cardiac strain. Sepsis resulted in cardiac dysfunction with a reduction in cardiac output and left ventricular end diastolic diameter. Both were improved by M3 treatment. Treatment with M3 attenuated serum, cardiac, and pulmonary levels of pro-inflammatory cytokines compared to vehicle-treated septic neonates. M3 dramatically increased sepsis survival. Conclusions Inhibition of eCIRP/TREM-1 interaction with M3 is cardioprotective, decreases inflammation, and improves survival in neonatal sepsis. Trial registration Retrospectively registered.
Collapse
Affiliation(s)
- Naomi-Liza Denning
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, 350 Community Dr, Manhasset, NY, 11030, USA.,Elmezzi Graduate School of Molecular Medicine, Manhasset, NY, USA.,Department of Surgery, Donald and Barbara Zucker School of Medicine At Hofstra/Northwell, Hempstead, NY, USA
| | - Monowar Aziz
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, 350 Community Dr, Manhasset, NY, 11030, USA.,Elmezzi Graduate School of Molecular Medicine, Manhasset, NY, USA
| | - Li Diao
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, 350 Community Dr, Manhasset, NY, 11030, USA
| | - Jose M Prince
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, 350 Community Dr, Manhasset, NY, 11030, USA.,Elmezzi Graduate School of Molecular Medicine, Manhasset, NY, USA.,Department of Surgery, Donald and Barbara Zucker School of Medicine At Hofstra/Northwell, Hempstead, NY, USA.,Division of Pediatric Surgery, Cohen Children's Medical Center At Hofstra/Northwell, New Hyde Park, NY, USA
| | - Ping Wang
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, 350 Community Dr, Manhasset, NY, 11030, USA. .,Elmezzi Graduate School of Molecular Medicine, Manhasset, NY, USA. .,Department of Surgery, Donald and Barbara Zucker School of Medicine At Hofstra/Northwell, Hempstead, NY, USA.
| |
Collapse
|
41
|
Schwarzkopf KM, Eberle L, Uschner FE, Klein S, Schierwagen R, Mücke MM, Schaefer L, Clària J, Zeuzem S, Hintermann E, Christen U, Lange CM, Trebicka J, Welsch C. Interleukin-22 in acute-on-chronic liver failure: A matter of ineffective levels, receptor dysregulation or defective signalling? J Hepatol 2020; 73:980-982. [PMID: 32698966 DOI: 10.1016/j.jhep.2020.05.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 05/12/2020] [Accepted: 05/12/2020] [Indexed: 01/16/2023]
Affiliation(s)
- Katharina Maria Schwarzkopf
- Department of Internal Medicine I, University Hospital Frankfurt, Goethe University, Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Larissa Eberle
- Department of Internal Medicine I, University Hospital Frankfurt, Goethe University, Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Frank Erhard Uschner
- Department of Internal Medicine I, University Hospital Frankfurt, Goethe University, Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Sabine Klein
- Department of Internal Medicine I, University Hospital Frankfurt, Goethe University, Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Robert Schierwagen
- Department of Internal Medicine I, University Hospital Frankfurt, Goethe University, Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Marcus Maximilian Mücke
- Department of Internal Medicine I, University Hospital Frankfurt, Goethe University, Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Liliana Schaefer
- Pharmazentrum Frankfurt/ZAFES, Goethe University Hospital Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Joan Clària
- European Foundation for the Study of Chronic Liver Failure, Barcelona, Spain; Hospital Clínic-IDIBAPS, Barcelona, Spain
| | - Stefan Zeuzem
- Department of Internal Medicine I, University Hospital Frankfurt, Goethe University, Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Edith Hintermann
- Pharmazentrum Frankfurt/ZAFES, Goethe University Hospital Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Urs Christen
- Pharmazentrum Frankfurt/ZAFES, Goethe University Hospital Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Christian Markus Lange
- Department of Gastroenterology and Hepatology, University Hospital Essen, University of Duisburg-Essen, Hufelandstr. 55, 45147 Essen, Germany
| | - Jonel Trebicka
- Department of Internal Medicine I, University Hospital Frankfurt, Goethe University, Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany; European Foundation for the Study of Chronic Liver Failure, Barcelona, Spain.
| | - Christoph Welsch
- Department of Internal Medicine I, University Hospital Frankfurt, Goethe University, Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| |
Collapse
|
42
|
Evaluating the Timeliness and Specificity of CD69, CD64, and CD25 as Biomarkers of Sepsis in Mice. Shock 2020; 55:507-518. [PMID: 32890312 DOI: 10.1097/shk.0000000000001650] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
ABSTRACT Sepsis occurs when an infection induces a dysregulated immune response, and is most commonly bacterial in origin. This condition requires rapid treatment for successful patient outcomes. However, the current method to confirm infection (blood culture) requires up to 48 h for a positive result and many true cases remain culture-negative. Therefore, new diagnostic tests are urgently needed. Recent clinical studies suggest that CD69, CD64, and CD25 may serve as useful biomarkers of sepsis. In this study, we evaluated the cecal ligation and puncture and cecal slurry mouse models as tools to study these biomarkers in young and aged mice, and elucidate the timeliness and specificity of sepsis diagnosis. Fluorescence-activated cell sorting analysis revealed that all three biomarkers were elevated on blood leukocytes during sepsis. CD69 was specifically upregulated during sepsis, while CD64 and CD25 were also transiently upregulated in response to sham surgery. The optimal biomarker, or combination of biomarkers, depended on the timing of detection, mouse age, and presence of surgery. CD69 demonstrated an excellent capacity to distinguish sepsis, and in some scenarios the diagnostic performance was enhanced by combining CD69 with CD64. We also analyzed biomarker expression levels on specific cell populations (lymphocytes, monocytes, and neutrophils) and determined the cell types that upregulate each biomarker. Elevations in blood biomarkers were also detected via microfluidic analyses; in this case CD64 distinguished septic mice from naive controls. Our results suggest that CD69 and CD64 are valuable biomarkers to rapidly detect sepsis, and that mouse models are useful to study and validate sepsis biomarkers.
Collapse
|
43
|
Can the Cecal Ligation and Puncture Model Be Repurposed To Better Inform Therapy in Human Sepsis? Infect Immun 2020; 88:IAI.00942-19. [PMID: 32571986 DOI: 10.1128/iai.00942-19] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
A recent report by the National Institutes of Health on sepsis research has implied there is a trend to move away from mouse models of sepsis. The most commonly used animal model to study the pathogenesis of human sepsis is cecal ligation and puncture (CLP) in mice. The model has been the mainstay of sepsis research for decades and continues to be considered the gold standard to inform novel pathways of sepsis physiology and its therapeutic direction. As there have been many criticisms of the model, particularly regarding its relevance to human disease, how this model might be repurposed to be more reflective of the human condition begs discussion. In this piece, we compare and contrast the mouse microbiome of the CLP model to the emerging science of the microbiome of human sepsis and discuss the relevance for mice to harbor the specific pathogens present in the human microbiome during sepsis, as well as an underlying disease process to mimic the characteristics of those patients with undesirable outcomes. How to repurpose this model to incorporate these "human factors" is discussed in detail and suggestions offered.
Collapse
|
44
|
Garcia-Valtanen P, van Diermen BA, Lakhan N, Lousberg EL, Robertson SA, Hayball JD, Diener KR. Maternal host responses to poly(I:C) during pregnancy leads to both dysfunctional immune profiles and altered behaviour in the offspring. Am J Reprod Immunol 2020; 84:e13260. [PMID: 32365239 DOI: 10.1111/aji.13260] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 04/21/2020] [Accepted: 04/23/2020] [Indexed: 01/01/2023] Open
Abstract
PROBLEM Autism spectrum disorder (ASD)-like phenotypes in murine models are linked to elevated pro-inflammatory cytokine profiles caused by maternal immune activation (MIA), but whether MIA alters the immune response in the offspring remains unclear. METHOD OF STUDY Polyinosinic:polycytidylic acid (poly:[IC]) was used to induce MIA in immunocompetent and control TLR3-deficient pregnant mice, and cytokine levels were measured in maternal and foetal organs. Furthermore, cytokines and behaviour responses were tested after challenge with lipopolysaccharide in 7-day-old and adult mice. RESULTS MIA induced on E12 resulted in changes in the cytokine expression profile in maternal and foetal organs and correlated with TNFα and IL-18 dysregulation in immune organs and brains from neonatal mice born to MIA-induced dams. Such changes further correlated with altered behavioural responses in adulthood. CONCLUSION MIA induced by pathogens during pregnancy can interfere with the development of the foetal immune and nervous systems leading to dysfunctional immune responses and behaviour in offspring.
Collapse
Affiliation(s)
- Pablo Garcia-Valtanen
- Experimental Therapeutics Laboratory, School of Pharmacy and Medical Science, UniSA Cancer Research Institute, University of South Australia, Adelaide, SA, Australia
| | - Bianca A van Diermen
- Experimental Therapeutics Laboratory, School of Pharmacy and Medical Science, UniSA Cancer Research Institute, University of South Australia, Adelaide, SA, Australia.,Robinson Research Institute and Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
| | - Nerissa Lakhan
- Experimental Therapeutics Laboratory, School of Pharmacy and Medical Science, UniSA Cancer Research Institute, University of South Australia, Adelaide, SA, Australia.,Robinson Research Institute and Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
| | - Erin L Lousberg
- Experimental Therapeutics Laboratory, School of Pharmacy and Medical Science, UniSA Cancer Research Institute, University of South Australia, Adelaide, SA, Australia.,Robinson Research Institute and Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
| | - Sarah A Robertson
- Robinson Research Institute and Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
| | - John D Hayball
- Experimental Therapeutics Laboratory, School of Pharmacy and Medical Science, UniSA Cancer Research Institute, University of South Australia, Adelaide, SA, Australia.,Robinson Research Institute and Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
| | - Kerrilyn R Diener
- Experimental Therapeutics Laboratory, School of Pharmacy and Medical Science, UniSA Cancer Research Institute, University of South Australia, Adelaide, SA, Australia.,Robinson Research Institute and Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
45
|
Abstract
Sepsis is a heterogeneous clinical syndrome that is complicated commonly by acute kidney injury (sepsis-AKI). Currently, no approved pharmacologic therapies exist to either prevent sepsis-AKI or to treat sepsis-AKI once it occurs. A growing body of evidence supports a connection between red blood cell biology and sepsis-AKI. Increased levels of circulating cell-free hemoglobin (CFH) released from red blood cells during hemolysis are common during sepsis and can contribute to sepsis-AKI through several mechanisms including tubular obstruction, nitric oxide depletion, oxidative injury, and proinflammatory signaling. A number of potential pharmacologic therapies targeting CFH in sepsis have been identified including haptoglobin, hemopexin, and acetaminophen, and early phase clinical trials have suggested that acetaminophen may have beneficial effects on lipid peroxidation and kidney function in patients with sepsis. Bedside measurement of CFH levels may facilitate predictive enrichment for future clinical trials of CFH-targeted therapeutics. However, rapid and reliable bedside tests for plasma CFH will be required for such trials to move forward.
Collapse
Affiliation(s)
- V Eric Kerchberger
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN; Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN
| | - Lorraine B Ware
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN; Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville TN.
| |
Collapse
|
46
|
Meegan JE, Shaver CM, Putz ND, Jesse JJ, Landstreet SR, Lee HNR, Sidorova TN, McNeil JB, Wynn JL, Cheung-Flynn J, Komalavilas P, Brophy CM, Ware LB, Bastarache JA. Cell-free hemoglobin increases inflammation, lung apoptosis, and microvascular permeability in murine polymicrobial sepsis. PLoS One 2020; 15:e0228727. [PMID: 32012200 PMCID: PMC6996826 DOI: 10.1371/journal.pone.0228727] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 01/22/2020] [Indexed: 12/28/2022] Open
Abstract
Increased endothelial permeability is central to the pathogenesis of sepsis and leads to organ dysfunction and death but the endogenous mechanisms that drive increased endothelial permeability are not completely understood. We previously reported that cell-free hemoglobin (CFH), elevated in 80% of patients with sepsis, increases lung microvascular permeability in an ex vivo human lung model and cultured endothelial cells. In this study, we augmented a murine model of polymicrobial sepsis with elevated circulating CFH to test the hypothesis that CFH increases microvascular endothelial permeability by inducing endothelial apoptosis. Mice were treated with an intraperitoneal injection of cecal slurry with or without a single intravenous injection of CFH. Severity of illness, mortality, systemic and lung inflammation, endothelial injury and dysfunction and lung apoptosis were measured at selected time points. We found that CFH added to CS increased sepsis mortality, plasma inflammatory cytokines as well as lung apoptosis, edema and inflammation without affecting large vessel reactivity or vascular injury marker concentrations. These results suggest that CFH is an endogenous mediator of increased endothelial permeability and apoptosis in sepsis and may be a promising therapeutic target.
Collapse
Affiliation(s)
- Jamie E. Meegan
- Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - Ciara M. Shaver
- Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - Nathan D. Putz
- Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - Jordan J. Jesse
- Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - Stuart R. Landstreet
- Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - Han Noo Ri Lee
- Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - Tatiana N. Sidorova
- Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - J. Brennan McNeil
- Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - James L. Wynn
- Departments of Pediatrics, Pathology, Immunology, and Experimental Medicine, University of Florida Health, Gainesville, FL, United States of America
| | - Joyce Cheung-Flynn
- Division of Vascular Surgery, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - Padmini Komalavilas
- Division of Vascular Surgery, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - Colleen M. Brophy
- Division of Vascular Surgery, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - Lorraine B. Ware
- Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, United States of America
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - Julie A. Bastarache
- Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, United States of America
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States of America
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, United States of America
| |
Collapse
|
47
|
Recombinant human thrombomodulin attenuated sepsis severity in a non-surgical preterm mouse model. Sci Rep 2020; 10:333. [PMID: 31941991 PMCID: PMC6962223 DOI: 10.1038/s41598-019-57265-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Accepted: 12/26/2019] [Indexed: 11/08/2022] Open
Abstract
Neonatal sepsis is characterised by dysregulated immune responses. Lipid mediators (LMs) are involved in the regulation of inflammation. Human recombinant thrombomodulin (rhTM), an anticoagulant, has anti-inflammatory effects and might be useful for sepsis treatment. A stock caecal slurry (CS) solution was prepared from adult caeca. To induce sepsis, 1.5 mg/g of CS was administered intraperitoneally to 4 d-old wild-type FVB mouse pups. Saline (Veh-CS) or rhTM (3 or 10 mg/kg; rhTM3-CS or rhTM10-CS) was administered subcutaneously 6 h prior to sepsis induction, and liver LM profiles at 3 and 6 h post-sepsis induction and survival up to 7 days were examined. Mortality was significantly lower (47%) in the rhTM3-CS group and significantly higher (100%) in the rhTM10-CS group, compared with the Veh-CS group (79%, p < 0.05). Eleven LMs (12-HEPE, EPA, 14-HDHA, DHA, PD1, PGD2, 15d-PGJ2, 12S-HHT, lipoxin B4, 12-HETE, AA) were significantly increased at 3 h, and five LMs (5-HEPE, 15-HEPE, 18-HEPE, 17-HDHA, PD1) were significantly increased at 6 h post-sepsis induction. Increased EPA, DHA, 12S-HHT, lipoxin B4, and AA were significantly suppressed by rhTM pre-treatment. rhTM was protective against neonatal sepsis. This protective effect might be mediated via LM modulation. Further post-sepsis studies are needed to determine clinical plausibility.
Collapse
|
48
|
Carpenter KC, Hakenjos JM, Fry CD, Nemzek JA. The Influence of Pain and Analgesia in Rodent Models of Sepsis. Comp Med 2019; 69:546-554. [PMID: 31213216 PMCID: PMC6935706 DOI: 10.30802/aalas-cm-19-000004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 02/15/2019] [Accepted: 04/01/2019] [Indexed: 12/17/2022]
Abstract
Sepsis is a multifaceted host response to infection that dramatically affects patient outcomes and the cost of health care. Animal models are necessary to replicate the complexity and heterogeneity of clinical sepsis. However, these models entail a high risk of pain and distress due to tissue trauma, inflammation, endotoxin-mediated hyperalgesia, and other mechanisms. Several recent studies and initiatives address the need to improve the welfare of animals through analgesics and standardize the models used in preclinical sepsis research. Ultimately, the goal is to provide high-fidelity, humane animal models that better replicate the clinical course of sepsis, to provide more effective translation and advance therapeutic discovery. The purpose of this review is to discuss the current understanding of the roles of pain and analgesia in rodent models of sepsis. The current definitions of sepsis along with an overview of pain in human sepsis are described. Finally, welfare concerns associated with animal models of sepsis and the most recent considerations for relief of pain and distress are reviewed.
Collapse
Affiliation(s)
- Kelsey C Carpenter
- Unit for Laboratory Animal Medicine, University of Michigan, Ann Arbor, Michigan
| | - John M Hakenjos
- Unit for Laboratory Animal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Christopher D Fry
- Unit for Laboratory Animal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Jean A Nemzek
- Unit for Laboratory Animal Medicine, University of Michigan, Ann Arbor, Michigan;,
| |
Collapse
|
49
|
Kerchberger VE, Bastarache JA, Shaver CM, Nagata H, McNeil JB, Landstreet SR, Putz ND, Yu WK, Jesse J, Wickersham NE, Sidorova TN, Janz DR, Parikh CR, Siew ED, Ware LB. Haptoglobin-2 variant increases susceptibility to acute respiratory distress syndrome during sepsis. JCI Insight 2019; 4:131206. [PMID: 31573976 DOI: 10.1172/jci.insight.131206] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 09/20/2019] [Indexed: 01/15/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS) is an inflammatory lung disorder that frequently complicates critical illness and commonly occurs in sepsis. Although numerous clinical and environmental risk factors exist, not all patients with risk factors develop ARDS, raising the possibility of genetic underpinnings for ARDS susceptibility. We have previously reported that circulating cell-free hemoglobin (CFH) is elevated during sepsis, and higher levels predict worse outcomes. Excess CFH is rapidly scavenged by haptoglobin (Hp). A common HP genetic variant, HP2, is unique to humans and is common in many populations worldwide. HP2 haptoglobin has reduced ability to inhibit CFH-mediated inflammation and oxidative stress compared with the alternative HP1. We hypothesized that HP2 increases ARDS susceptibility during sepsis when plasma CFH levels are elevated. In a murine model of sepsis with elevated CFH, transgenic mice homozygous for Hp2 had increased lung inflammation, pulmonary vascular permeability, lung apoptosis, and mortality compared with wild-type mice. We then tested the clinical relevance of our findings in 496 septic critically ill adults, finding that HP2 increased ARDS susceptibility after controlling for clinical risk factors and plasma CFH. These observations identify HP2 as a potentially novel genetic ARDS risk factor during sepsis and may have important implications in the study and treatment of ARDS.
Collapse
Affiliation(s)
- V Eric Kerchberger
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine.,Department of Biomedical Informatics
| | - Julie A Bastarache
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine.,Department of Cell and Developmental Biology, and.,Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Ciara M Shaver
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine
| | - Hiromasa Nagata
- Department of Anesthesiology, Keio University School of Medicine, Tokyo, Japan
| | - J Brennan McNeil
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine
| | - Stuart R Landstreet
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine
| | - Nathan D Putz
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine
| | - Wen-Kuang Yu
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine.,Department of Chest Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.,School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Jordan Jesse
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine
| | - Nancy E Wickersham
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine
| | - Tatiana N Sidorova
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine
| | - David R Janz
- Section of Pulmonary and Critical Care Medicine, Louisiana State University School of Medicine, New Orleans, Louisiana, USA
| | - Chirag R Parikh
- Division of Nephrology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Edward D Siew
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Lorraine B Ware
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine.,Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
50
|
Neonatal Sepsis Alters the Excitability of Regular Spiking Cells in the Nucleus of the Solitary Tract in Rats. Shock 2019; 54:265-271. [DOI: 10.1097/shk.0000000000001453] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|