1
|
Hunter CE, Mesfin FM, Manohar K, Liu J, Shelley WC, Brokaw JP, Pecoraro AR, Hosfield BD, Markel TA. Hydrogen Sulfide Improves Outcomes in a Murine Model of Necrotizing Enterocolitis via the Cys440 Residue on Endothelial Nitric Oxide Synthase. J Pediatr Surg 2023; 58:2391-2398. [PMID: 37684170 PMCID: PMC10841167 DOI: 10.1016/j.jpedsurg.2023.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 07/31/2023] [Accepted: 08/13/2023] [Indexed: 09/10/2023]
Abstract
BACKGROUND Hydrogen sulfide (H2S) has been shown to improve outcomes in a murine model of necrotizing enterocolitis (NEC). There is evidence in humans that H2S relies on endothelial nitric oxide synthase (eNOS) to exert its protective effects, potentially through the persulfidation of eNOS at the Cysteine 443 residue. We obtained a novel mouse strain with a mutation at this residue (eNOSC440G) and hypothesized that this locus would be critical for GYY4137 (an H2S donor) to exert its protective effects. METHODS Necrotizing enterocolitis was induced in 5-day old wild type (WT) and eNOSC440G mice using intermittent exposure to hypoxia and hypothermia in addition to gavage formula feeds. On postnatal day 9, mice were humanely euthanized. Data collected included daily weights, clinical sickness scores, histologic lung injury, intestinal injury (macroscopically and histologically), and intestinal perfusion. During the NEC model, pups received daily intraperitoneal injections of either GYY4137 (50 mg/kg) or PBS (vehicle). Data were tested for normality and compared using t-test or Mann-Whitney, and a p-value <0.05 was considered significant. RESULTS In WT mice, the administration of GYY4137 significantly improved clinical sickness scores, attenuated intestinal and lung injury, and improved mesenteric perfusion compared to vehicle (p < 0.05). In eNOSC440G mice, the treatment and vehicle groups had similar clinical sickness scores, intestinal and lung injury scores, and intestinal perfusion. CONCLUSIONS GYY4137 administration improves clinical outcomes, attenuates intestinal and lung injury, and improves perfusion in a murine model of necrotizing enterocolitis. The beneficial effects of GYY4137 are dependent on the Cys440 residue of eNOS.
Collapse
Affiliation(s)
- Chelsea E Hunter
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Fikir M Mesfin
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Krishna Manohar
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jianyun Liu
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | - John P Brokaw
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Anthony R Pecoraro
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Brian D Hosfield
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Troy A Markel
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, USA; Riley Hospital for Children at Indiana University Health, Indianapolis, IN, USA.
| |
Collapse
|
2
|
Ganji N, Alganabi M, Yamoto M, Chusilp S, Pierro A, Li B. Family care reduces the incidence of neonatal sepsis: A systematic review and meta-analysis. Front Pediatr 2023; 11:1089229. [PMID: 37124182 PMCID: PMC10130514 DOI: 10.3389/fped.2023.1089229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 03/29/2023] [Indexed: 05/02/2023] Open
Abstract
Purpose Family-involved care in the neonatal intensive care unit (NICU) helps to alleviate neonatal anxiety and promotes breastmilk intake, body growth and neurological development, but its effect on reducing the incidence of neonatal sepsis is not known. We conducted a systematic review and meta-analysis of randomized controlled trials (RCT) to evaluate whether neonates receiving family care have a lower incidence of neonatal sepsis compared to neonates receiving standard NICU care. Methods MEDLINE, Embase, Web of Science, and CENTRAL were searched for RCTs that compared preterm neonates receiving family care vs. standard NICU care. From 126 articles that were identified and screened, 34 full-text articles were assessed for eligibility, and 5 RCTs were included. The primary outcome was the development of sepsis. The RevMan 5.4 software was used to conduct the Meta-analysis. Results The metanalysis, based on 5 RCTs demonstrated that neonates receiving family-involved care had significantly lower incidence of sepsis (12.0% vs. 16.3%), increased body weight, and reduced length of hospital stay compared to those receiving standard NICU care. Conclusion This study suggests that family-involved care in NICU can (i) reduce the incidence of neonatal sepsis, (ii) improve growth, and (iii) reduce the length of hospital stay. This study highlights the need for evaluating whether family-involved care improves other neonatal outcomes.
Collapse
|
3
|
Hosfield BD, Hunter CE, Li H, Drucker NA, Pecoraro AR, Manohar K, Shelley WC, Markel TA. A hydrogen-sulfide derivative of mesalamine reduces the severity of intestinal and lung injury in necrotizing enterocolitis through endothelial nitric oxide synthase. Am J Physiol Regul Integr Comp Physiol 2022; 323:R422-R431. [PMID: 35912999 PMCID: PMC9512109 DOI: 10.1152/ajpregu.00229.2021] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 06/18/2022] [Accepted: 07/19/2022] [Indexed: 11/22/2022]
Abstract
Necrotizing enterocolitis (NEC) remains a devastating disease that affects preterm infants. Hydrogen sulfide (H2S) donors have been shown to reduce the severity of NEC, but the optimal compound has yet to be identified. We hypothesized that oral H2S-Mesalamine (ATB-429) would improve outcomes in experimental NEC, and its benefits would be dependent on endothelial nitric oxide synthase (eNOS) pathways. NEC was induced in 5-day-old wild-type (WT) and eNOS knockout (eNOSKO) pups by formula feeding and stress. Four groups were studied in both WT and eNOSKO mice: 1) breastfed controls, 2) NEC, 3) NEC + 50 mg/kg mesalamine, and 4) NEC + 130 mg/kg ATB-429. Mesalamine and ATB-429 doses were equimolar. Pups were monitored for sickness scores and perfusion to the gut was measured by Laser Doppler Imaging (LDI). After euthanasia of the pups, intestine and lung were hematoxylin and eosin-stained and scored for injury in a blind fashion. TLR4 expression was quantified by Western blot and IL-6 expression by ELISA. P < 0.05 was significant. Both WT and eNOSKO breastfed controls underwent normal development and demonstrated milder intestinal and pulmonary injury compared with NEC groups. For the WT groups, ATB-429 significantly improved weight gain, reduced clinical sickness score, and improved perfusion compared with the NEC group. In addition, WT ATB-429 pups had a significantly milder intestinal and pulmonary histologic injury when compared with NEC. ATB-429 attenuated the increase in TLR4 and IL-6 expression in the intestine. When the experiment was repeated in eNOSKO pups, ATB-429 offered no benefit in weight gain, sickness scores, perfusion, intestinal injury, pulmonary injury, or decreasing intestinal inflammatory markers. An H2S derivative of mesalamine improves outcomes in experimental NEC. Protective effects appear to be mediated through eNOS. Further research is warranted to explore whether ATB-429 may be an effective oral therapy to combat NEC.
Collapse
Affiliation(s)
- Brian D Hosfield
- Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| | - Chelsea E Hunter
- Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| | - Hongge Li
- Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| | - Natalie A Drucker
- Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| | - Anthony R Pecoraro
- Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| | - Krishna Manohar
- Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| | | | - Troy A Markel
- Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana
- Riley Hospital for Children, Indiana University Health, Indianapolis, Indiana
| |
Collapse
|
4
|
Yuksel S, Erginel B, Bingul I, Ozluk Y, Karatay H, Aydın F, Keskin E. The effect of hydrogen sulfide on ischemi̇a /reperfusion injury in an experimental testicular torsion model. J Pediatr Urol 2022; 18:16.e1-16.e7. [PMID: 34937685 DOI: 10.1016/j.jpurol.2021.11.019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 09/30/2021] [Accepted: 11/29/2021] [Indexed: 02/05/2023]
Abstract
INTRODUCTION Testicular torsion is an acute pediatric surgical emergency requiring rapid diagnosis to prevent the permanent ischaemic damage of the testicles. Hydrogen Sulfide (H2S) have shown to cure tissue damage and has a role in the prevention of I/R damage. We aimed to evaluate the effect of H2S in testicular torsion. MATERIALS AND METHODS Eighteen male, Wistar albino rats were divided into 3 groups. The sham group which is applied surgical stress. The ischemia/reperfusion group (I/R) which detorsion performed 1 h later than testicular torsion application. I/R + NaHS treatment group, NaHS solution was injected intraperitoneally for 1 week. On the 7th day of the detorsion all left testes were fixed in Bouin solution and sent to Pathology Department for histopathological examination. All right testes were washed with normal saline, dried in a sterile way and stored in - 80 °C deepfreeze up to the date of biochemical processes. Testicular tissues were obtained for the detection of myeloperoxidase (MPO), malondialdehyde (MDA), AOPP (advanced oxidation protein product) for oxidant markers and ferric reducing antioxidant power (FRAP) levels, superoxide dismutase (SOD),glutathione peroxidase (GSH-Px) activities for antioxidant markers and histopathological exploration. RESULTS The effects of NaHs administration on oxidation were evaluated by determination of testicular MPO, MDA and AOPP levels. Increased testicular MPO (58.6%) activity was observed in the I/R group compared to the sham group. Following NaHS treatment, MPO (26.7%) activity was significantly decreased in rats exposed to I/R injury (Figure 1). MDA levels did not alter. Increases in AOPP (20.9%) levels were observed in the I/R group. NaHS treatment resulted in significant decreases in AOPP (25.1%) levels in testes tissues of rats exposed to I/R injury. The effects of NaHS treatment on antioxidant system FRAP, SOD, GSH and GSH-Px activities were evaluated. GSH levels were significantly increased in the IR + NaHS group compared to the I/R group. In histopathological examination degeneration of seminiferous tubules and spermatogenic cells were observed in the I/R group. After NaHS treatment, normal spermatogenic activity with many spermatozoa in the lumen of most seminiferous tubules were observed in the I/R injured rats. According to Johnsen's scoring (JS), the I/R group was significantly decreased compared to the sham group. JS values for the I/R + NaHS group were significantly increased compared to the I/R group. CONCLUSION Our study supports that ischemia/reperfusion injury plays an important role in the testicular torsion injury, and it is a pioneer study showing that H₂S may have a potential for therapeutic effect. The limitation of this work is this is an experimental study with limited number of animals. According to the results of our study, hydrogen sulfide treatment has beneficial effects on biochemical and histopathological results of testicular injury in testic torsion.
Collapse
Affiliation(s)
- Secil Yuksel
- Istanbul Faculty of Medicine, Department of Pediatric Surgery, Istanbul, Turkey
| | - Basak Erginel
- Istanbul Faculty of Medicine, Department of Pediatric Surgery, Istanbul, Turkey.
| | - Ilknur Bingul
- Istanbul Faculty of Medicine, Department of Biochemistry, Istanbul, Turkey
| | - Yasemin Ozluk
- Istanbul Faculty of Medicine, Department of Pathology, Istanbul, Turkey
| | - Huseyin Karatay
- Istanbul Faculty of Medicine, Department of Pathology, Istanbul, Turkey
| | - Fatih Aydın
- Istanbul Faculty of Medicine, Department of Biochemistry, Istanbul, Turkey
| | - Erbug Keskin
- Istanbul Faculty of Medicine, Department of Pediatric Surgery, Istanbul, Turkey
| |
Collapse
|
5
|
Merz T, McCook O, Denoix N, Radermacher P, Waller C, Kapapa T. Biological Connection of Psychological Stress and Polytrauma under Intensive Care: The Role of Oxytocin and Hydrogen Sulfide. Int J Mol Sci 2021; 22:9192. [PMID: 34502097 PMCID: PMC8430789 DOI: 10.3390/ijms22179192] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 08/06/2021] [Accepted: 08/20/2021] [Indexed: 12/12/2022] Open
Abstract
This paper explored the potential mediating role of hydrogen sulfide (H2S) and the oxytocin (OT) systems in hemorrhagic shock (HS) and/or traumatic brain injury (TBI). Morbidity and mortality after trauma mainly depend on the presence of HS and/or TBI. Rapid "repayment of the O2 debt" and prevention of brain tissue hypoxia are cornerstones of the management of both HS and TBI. Restoring tissue perfusion, however, generates an ischemia/reperfusion (I/R) injury due to the formation of reactive oxygen (ROS) and nitrogen (RNS) species. Moreover, pre-existing-medical-conditions (PEMC's) can aggravate the occurrence and severity of complications after trauma. In addition to the "classic" chronic diseases (of cardiovascular or metabolic origin), there is growing awareness of psychological PEMC's, e.g., early life stress (ELS) increases the predisposition to develop post-traumatic-stress-disorder (PTSD) and trauma patients with TBI show a significantly higher incidence of PTSD than patients without TBI. In fact, ELS is known to contribute to the developmental origins of cardiovascular disease. The neurotransmitter H2S is not only essential for the neuroendocrine stress response, but is also a promising therapeutic target in the prevention of chronic diseases induced by ELS. The neuroendocrine hormone OT has fundamental importance for brain development and social behavior, and, thus, is implicated in resilience or vulnerability to traumatic events. OT and H2S have been shown to interact in physical and psychological trauma and could, thus, be therapeutic targets to mitigate the acute post-traumatic effects of chronic PEMC's. OT and H2S both share anti-inflammatory, anti-oxidant, and vasoactive properties; through the reperfusion injury salvage kinase (RISK) pathway, where their signaling mechanisms converge, they act via the regulation of nitric oxide (NO).
Collapse
Affiliation(s)
- Tamara Merz
- Institute for Anesthesiological Pathophysiology and Process Engineering, Medical Center, Ulm University, Helmholtzstraße 8/1, 89081 Ulm, Germany; (T.M.); (N.D.); (P.R.)
| | - Oscar McCook
- Institute for Anesthesiological Pathophysiology and Process Engineering, Medical Center, Ulm University, Helmholtzstraße 8/1, 89081 Ulm, Germany; (T.M.); (N.D.); (P.R.)
| | - Nicole Denoix
- Institute for Anesthesiological Pathophysiology and Process Engineering, Medical Center, Ulm University, Helmholtzstraße 8/1, 89081 Ulm, Germany; (T.M.); (N.D.); (P.R.)
- Clinic for Psychosomatic Medicine and Psychotherapy, Medical Center, Ulm University, 89081 Ulm, Germany
| | - Peter Radermacher
- Institute for Anesthesiological Pathophysiology and Process Engineering, Medical Center, Ulm University, Helmholtzstraße 8/1, 89081 Ulm, Germany; (T.M.); (N.D.); (P.R.)
| | - Christiane Waller
- Department of Psychosomatic Medicine and Psychotherapy, Nuremberg General Hospital, Paracelsus Medical University, 90471 Nuremberg, Germany;
| | - Thomas Kapapa
- Clinic for Neurosurgery, Medical Center, Ulm University, 89081 Ulm, Germany;
| |
Collapse
|
6
|
McCook O, Denoix N, Radermacher P, Waller C, Merz T. H 2S and Oxytocin Systems in Early Life Stress and Cardiovascular Disease. J Clin Med 2021; 10:jcm10163484. [PMID: 34441780 PMCID: PMC8397059 DOI: 10.3390/jcm10163484] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 08/02/2021] [Accepted: 08/03/2021] [Indexed: 02/07/2023] Open
Abstract
Today it is well established that early life stress leads to cardiovascular programming that manifests in cardiovascular disease, but the mechanisms by which this occurs, are not fully understood. This perspective review examines the relevant literature that implicates the dysregulation of the gasomediator hydrogen sulfide and the neuroendocrine oxytocin systems in heart disease and their putative mechanistic role in the early life stress developmental origins of cardiovascular disease. Furthermore, interesting hints towards the mutual interaction of the hydrogen sulfide and OT systems are identified, especially with regards to the connection between the central nervous and the cardiovascular system, which support the role of the vagus nerve as a communication link between the brain and the heart in stress-mediated cardiovascular disease.
Collapse
Affiliation(s)
- Oscar McCook
- Institute for Anesthesiological Pathophysiology and Process Engineering, Ulm University Medical Center, 89081 Ulm, Germany; (N.D.); (P.R.); (T.M.)
- Correspondence: ; Tel.: +49-731-500-60185; Fax: +49-731-500-60162
| | - Nicole Denoix
- Institute for Anesthesiological Pathophysiology and Process Engineering, Ulm University Medical Center, 89081 Ulm, Germany; (N.D.); (P.R.); (T.M.)
- Clinic for Psychosomatic Medicine and Psychotherapy, Ulm University Medical Center, 89081 Ulm, Germany
| | - Peter Radermacher
- Institute for Anesthesiological Pathophysiology and Process Engineering, Ulm University Medical Center, 89081 Ulm, Germany; (N.D.); (P.R.); (T.M.)
| | - Christiane Waller
- Department of Psychosomatic Medicine and Psychotherapy, Nuremberg General Hospital, Paracelsus Medical University, 90471 Nuremberg, Germany;
| | - Tamara Merz
- Institute for Anesthesiological Pathophysiology and Process Engineering, Ulm University Medical Center, 89081 Ulm, Germany; (N.D.); (P.R.); (T.M.)
| |
Collapse
|
7
|
Koike Y, Li B, Ganji N, Zhu H, Miyake H, Chen Y, Lee C, Janssen Lok M, Zozaya C, Lau E, Lee D, Chusilp S, Zhang Z, Yamoto M, Wu RY, Inoue M, Uchida K, Kusunoki M, Delgado-Olguin P, Mertens L, Daneman A, Eaton S, Sherman PM, Pierro A. Remote ischemic conditioning counteracts the intestinal damage of necrotizing enterocolitis by improving intestinal microcirculation. Nat Commun 2020; 11:4950. [PMID: 33009377 PMCID: PMC7532542 DOI: 10.1038/s41467-020-18750-9] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 09/09/2020] [Indexed: 02/07/2023] Open
Abstract
Necrotizing enterocolitis (NEC) is a devastating disease of premature infants with high mortality rate, indicating the need for precision treatment. NEC is characterized by intestinal inflammation and ischemia, as well derangements in intestinal microcirculation. Remote ischemic conditioning (RIC) has emerged as a promising tool in protecting distant organs against ischemia-induced damage. However, the effectiveness of RIC against NEC is unknown. To address this gap, we aimed to determine the efficacy and mechanism of action of RIC in experimental NEC. NEC was induced in mouse pups between postnatal day (P) 5 and 9. RIC was applied through intermittent occlusion of hind limb blood flow. RIC, when administered in the early stages of disease progression, decreases intestinal injury and prolongs survival. The mechanism of action of RIC involves increasing intestinal perfusion through vasodilation mediated by nitric oxide and hydrogen sulfide. RIC is a viable and non-invasive treatment strategy for NEC. Necrotizing enterocolitis (NEC) is one of the most lethal gastrointestinal emergencies in neonates needing precision treatment. Here the authors show that remote ischemic conditioning is a non-invasive therapeutic method that enhances blood flow in the intestine, reduces damage, and improves NEC outcome.
Collapse
Affiliation(s)
- Yuhki Koike
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada.,Division of General and Thoracic Surgery, Translational Medicine, The Hospital for Sick Children, Toronto, ON, Canada.,Departments of Gastrointestinal and Pediatric Surgery, Mie University Graduate School of Medicine, Tsu, Japan
| | - Bo Li
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada.,Division of General and Thoracic Surgery, Translational Medicine, The Hospital for Sick Children, Toronto, ON, Canada
| | - Niloofar Ganji
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada.,Division of General and Thoracic Surgery, Translational Medicine, The Hospital for Sick Children, Toronto, ON, Canada
| | - Haitao Zhu
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada.,Division of General and Thoracic Surgery, Translational Medicine, The Hospital for Sick Children, Toronto, ON, Canada
| | - Hiromu Miyake
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada.,Division of General and Thoracic Surgery, Translational Medicine, The Hospital for Sick Children, Toronto, ON, Canada
| | - Yong Chen
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada.,Division of General and Thoracic Surgery, Translational Medicine, The Hospital for Sick Children, Toronto, ON, Canada
| | - Carol Lee
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada.,Division of General and Thoracic Surgery, Translational Medicine, The Hospital for Sick Children, Toronto, ON, Canada
| | - Maarten Janssen Lok
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada.,Division of General and Thoracic Surgery, Translational Medicine, The Hospital for Sick Children, Toronto, ON, Canada
| | - Carlos Zozaya
- Division of Neonatology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Ethan Lau
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada.,Division of General and Thoracic Surgery, Translational Medicine, The Hospital for Sick Children, Toronto, ON, Canada
| | - Dorothy Lee
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada.,Division of General and Thoracic Surgery, Translational Medicine, The Hospital for Sick Children, Toronto, ON, Canada
| | - Sinobol Chusilp
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada.,Division of General and Thoracic Surgery, Translational Medicine, The Hospital for Sick Children, Toronto, ON, Canada
| | - Zhen Zhang
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada.,Division of General and Thoracic Surgery, Translational Medicine, The Hospital for Sick Children, Toronto, ON, Canada
| | - Masaya Yamoto
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada.,Division of General and Thoracic Surgery, Translational Medicine, The Hospital for Sick Children, Toronto, ON, Canada
| | - Richard Y Wu
- Cell Biology Program, Research Institute, Division of Gastroenterology, Hepatology and Nutrition, Hospital for Sick Children, Toronto, ON, Canada.,Department of Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Mikihiro Inoue
- Departments of Gastrointestinal and Pediatric Surgery, Mie University Graduate School of Medicine, Tsu, Japan
| | - Keiichi Uchida
- Departments of Gastrointestinal and Pediatric Surgery, Mie University Graduate School of Medicine, Tsu, Japan
| | - Masato Kusunoki
- Departments of Gastrointestinal and Pediatric Surgery, Mie University Graduate School of Medicine, Tsu, Japan
| | - Paul Delgado-Olguin
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada.,Heart & Stroke Richard Lewar Centre of Excellence, Toronto, ON, Canada
| | - Luc Mertens
- The Labatt Family Heart Center, Cardiology, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Alan Daneman
- Department of Diagnostic Imaging, Division of Nuclear Medicine, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Simon Eaton
- UCL Great Ormond Street Institute of Child Health, London, UK
| | - Philip M Sherman
- Cell Biology Program, Research Institute, Division of Gastroenterology, Hepatology and Nutrition, Hospital for Sick Children, Toronto, ON, Canada.,Faculty of Dentistry, University of Toronto, Toronto, ON, Canada
| | - Agostino Pierro
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada. .,Division of General and Thoracic Surgery, Translational Medicine, The Hospital for Sick Children, Toronto, ON, Canada. .,Department of Surgery, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
8
|
Denoix N, McCook O, Ecker S, Wang R, Waller C, Radermacher P, Merz T. The Interaction of the Endogenous Hydrogen Sulfide and Oxytocin Systems in Fluid Regulation and the Cardiovascular System. Antioxidants (Basel) 2020; 9:E748. [PMID: 32823845 PMCID: PMC7465147 DOI: 10.3390/antiox9080748] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 08/10/2020] [Accepted: 08/11/2020] [Indexed: 12/11/2022] Open
Abstract
The purpose of this review is to explore the parallel roles and interaction of hydrogen sulfide (H2S) and oxytocin (OT) in cardiovascular regulation and fluid homeostasis. Their interaction has been recently reported to be relevant during physical and psychological trauma. However, literature reports on H2S in physical trauma and OT in psychological trauma are abundant, whereas available information regarding H2S in psychological trauma and OT in physical trauma is much more limited. This review summarizes recent direct and indirect evidence of the interaction of the two systems and their convergence in downstream nitric oxide-dependent signaling pathways during various types of trauma, in an effort to better understand biological correlates of psychosomatic interdependencies.
Collapse
Affiliation(s)
- Nicole Denoix
- Clinic for Psychosomatic Medicine and Psychotherapy, Ulm University Medical Center, 89081 Ulm, Germany;
- Institute for Anesthesiological Pathophysiology and Process Engineering, Ulm University Medical Center, 89081 Ulm, Germany; (S.E.); (P.R.); (T.M.)
| | - Oscar McCook
- Institute for Anesthesiological Pathophysiology and Process Engineering, Ulm University Medical Center, 89081 Ulm, Germany; (S.E.); (P.R.); (T.M.)
| | - Sarah Ecker
- Institute for Anesthesiological Pathophysiology and Process Engineering, Ulm University Medical Center, 89081 Ulm, Germany; (S.E.); (P.R.); (T.M.)
| | - Rui Wang
- Faculty of Science, York University, Toronto, ON M3J 1P3, Canada;
| | - Christiane Waller
- Department of Psychosomatic Medicine and Psychotherapy, Nuremberg General Hospital, Paracelsus Medical University, 90419 Nuremberg, Germany;
| | - Peter Radermacher
- Institute for Anesthesiological Pathophysiology and Process Engineering, Ulm University Medical Center, 89081 Ulm, Germany; (S.E.); (P.R.); (T.M.)
| | - Tamara Merz
- Institute for Anesthesiological Pathophysiology and Process Engineering, Ulm University Medical Center, 89081 Ulm, Germany; (S.E.); (P.R.); (T.M.)
| |
Collapse
|
9
|
Liu J, Miyake H, Zhu H, Li B, Alganabi M, Lee C, Pierro A. Fecal microbiota transplantation by enema reduces intestinal injury in experimental necrotizing enterocolitis. J Pediatr Surg 2020; 55:1094-1098. [PMID: 32234317 DOI: 10.1016/j.jpedsurg.2020.02.035] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 02/20/2020] [Indexed: 01/05/2023]
Abstract
PURPOSE Necrotizing Enterocolitis (NEC) is a devastating neonatal disease with a high mortality rate. Fecal Microbiota Transplantation (FMT) has been used to treat a variety of gastrointestinal diseases. We aimed to investigate the role of FMT in NEC. METHODS NEC was induced by hypoxia, LPS, and hyperosmolar gavage feeding between postnatal days P5 and P9 (n = 8). Breastfed mice were used as control (n = 7). FMT (30 μl/g) was administered by gavage or enema at P6 during NEC induction. Distal ileum was harvested on P9. Disease severity was evaluated by H&E staining. Gene expression of inflammatory markers IL6 and TNFa was measured. Expression of intestinal barrier function was investigated by measuring Claudin-7. Microbiota composition in ileum and colon was analyzed by quantitative PCR. RESULTS FMT by gavage further increased terminal ileum inflammation and did not improve the histological damage owing to experimental NEC. Conversely, FMT by enema decreased intestinal inflammation and improved histology of the NEC-like injury in the ileum. In addition, compared with NEC alone, FMT by enema increased Claudin-7 expression indicating an improvement in barrier function. These beneficial effects occurred despite no change in microbiota. CONCLUSION Our results show that FMT by enema may be an effective strategy to reduce NEC progression as it attenuates intestinal inflammation and enhances intestinal barrier function. FMT by enema is a potential novel treatment for NEC. LEVEL OF EVIDENCE Level IV, Evidence from well-designed case-control or cohort studies.
Collapse
Affiliation(s)
- Jia Liu
- The Hospital for Sick Children, University of Toronto, Toronto, Canada; Department of Pediatric Surgery, Children's Hospital of Fudan University, Shanghai, China
| | - Hiromu Miyake
- The Hospital for Sick Children, University of Toronto, Toronto, Canada
| | - Haitao Zhu
- The Hospital for Sick Children, University of Toronto, Toronto, Canada; Department of Pediatric Surgery, Children's Hospital of Fudan University, Shanghai, China
| | - Bo Li
- The Hospital for Sick Children, University of Toronto, Toronto, Canada
| | - Mashriq Alganabi
- The Hospital for Sick Children, University of Toronto, Toronto, Canada
| | - Carol Lee
- The Hospital for Sick Children, University of Toronto, Toronto, Canada
| | - Agostino Pierro
- The Hospital for Sick Children, University of Toronto, Toronto, Canada.
| |
Collapse
|
10
|
Guan R, Wang J, Li D, Li Z, Liu H, Ding M, Cai Z, Liang X, Yang Q, Long Z, Chen L, Liu W, Sun D, Yao H, Lu W. Hydrogen sulfide inhibits cigarette smoke-induced inflammation and injury in alveolar epithelial cells by suppressing PHD2/HIF-1α/MAPK signaling pathway. Int Immunopharmacol 2020; 81:105979. [PMID: 31771816 DOI: 10.1016/j.intimp.2019.105979] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 09/28/2019] [Accepted: 10/13/2019] [Indexed: 02/07/2023]
Abstract
Chronic obstructive pulmonary fibrosis (COPD) is a chronic and fatal lung disease with few treatment options. Sodium hydrosulfide (NaHS), a donor of hydrogen sulfide (H2S), was found to alleviate cigarette smoke (CS)-induced emphysema in mice, however, the underlying mechanisms have not yet been clarified. In this study, we investigated its effects on COPD in a CS-induced mouse model in vivo and in cigarette smoke extract (CSE)-stimulated alveolar epithelial A549 cells in vitro. The results showed that NaHS not only relieved emphysema, but also improved pulmonary function in CS-exposed mice. NaHS significantly increased the expressions of tight junction proteins (i.e., ZO-1, Occludin and claudin-1), and reduced apoptosis and secretion of pro-inflammatory cytokines (i.e., TNF-α, IL-6 and IL-1β) in CS-exposed mouse lungs and CSE-incubated A549 cells, indicating H2S inhibits CS-induced inflammation, injury and apoptosis in alveolar epithelial cells. NaHS also upregulated prolyl hydroxylase (PHD)2, and suppressed hypoxia-inducible factor (HIF)-1α expression in vivo and in vitro, suggesting H2S inhibits CS-induced activation of PHD2/HIF-1α axis. Moreover, NaHS inhibited CS-induced phosphorylation of ERK, JNK and p38 MAPK in vivo and in vitro, and treatment with their inhibitors reversed CSE-induced ZO-1 expression and inflammation in A549 cells. These results suggest that NaHS may prevent emphysema via the suppression of PHD2/HIF-1α/MAPK signaling pathway, and subsequently inhibition of inflammation, epithelial cell injury and apoptosis, and may be a novel strategy for the treatment of COPD.
Collapse
Affiliation(s)
- Ruijuan Guan
- State Key Laboratory of Respiratory Diseases, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jian Wang
- State Key Laboratory of Respiratory Diseases, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Defu Li
- State Key Laboratory of Respiratory Diseases, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China; The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Ziying Li
- State Key Laboratory of Respiratory Diseases, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Hanwei Liu
- State Key Laboratory of Respiratory Diseases, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Mingjing Ding
- State Key Laboratory of Respiratory Diseases, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China; Departments of Respiratory and Critical Diseases, Inner Mongolia Autonomous Region People's Hospital, Hohhot, China
| | - Zhou Cai
- State Key Laboratory of Respiratory Diseases, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xue Liang
- State Key Laboratory of Respiratory Diseases, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China; The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Qian Yang
- State Key Laboratory of Respiratory Diseases, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Zhen Long
- State Key Laboratory of Respiratory Diseases, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Lingzhu Chen
- State Key Laboratory of Respiratory Diseases, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Wei Liu
- State Key Laboratory of Respiratory Diseases, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Dejun Sun
- Departments of Respiratory and Critical Diseases, Inner Mongolia Autonomous Region People's Hospital, Hohhot, China
| | - Hongwei Yao
- State Key Laboratory of Respiratory Diseases, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.
| | - Wenju Lu
- State Key Laboratory of Respiratory Diseases, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
11
|
Maternal Separation Induces Long-Term Alterations in the Cardiac Oxytocin Receptor and Cystathionine γ-Lyase Expression in Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:4309605. [PMID: 32082478 PMCID: PMC7007946 DOI: 10.1155/2020/4309605] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 12/13/2019] [Accepted: 12/28/2019] [Indexed: 12/21/2022]
Abstract
We recently showed that blunt chest trauma reduced the expression of the myocardial oxytocin receptor (Oxtr), which was further aggravated by genetic deletion of the H2S-producing enzyme cystathionine γ-lyase (CSE). Exogenous H2S supplementation restored myocardial Oxtr expression under these conditions. Early life stress (ELS) is a risk factor for cardiovascular disease by affecting vascular and heart structures. Therefore, we tested the hypotheses that (i) ELS affects cardiac Oxtr and CSE expressions and (ii) Oxtr and CSE expression patterns depend on the duration of stress exposure. Thus, two stress paradigms were compared: long- and short-term separation stress (LTSS and STSS, respectively). Cardiac Oxtr expression was differentially affected by the two stress paradigms with a significant reduction after LTSS and a significant increase after STSS. CSE expression, which was significantly reduced in Oxtr−/− knockout hearts, was downregulated and directly related to Oxtr expression in LTSS hearts (r = 0.657, p = 0.012). In contrast, CSE expression was not related to Oxtr upregulation in STSS. Plasma Oxt levels were not affected by either ELS paradigm. The coincidence of LTSS-induced reduction of cardiac Oxtr and reduced CSE expression may suggest a novel pathophysiological link between early life adversities and increased risk for the development of cardiovascular disorders in adulthood.
Collapse
|
12
|
Dutcher EG, Pama EC, Lynall ME, Khan S, Clatworthy MR, Robbins TW, Bullmore ET, Dalley JW. Early-life stress and inflammation: A systematic review of a key experimental approach in rodents. Brain Neurosci Adv 2020; 4:2398212820978049. [PMID: 33447663 PMCID: PMC7780197 DOI: 10.1177/2398212820978049] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 11/11/2020] [Indexed: 12/11/2022] Open
Abstract
Repeated maternal separation is the most widely used pre-clinical approach to investigate the relationship between early-life chronic stress and its neuropsychiatric and physical consequences. In this systematic review, we identified 46 studies that conducted repeated maternal separation or single-episode maternal separation and reported measurements of interleukin-1b, interleukin-6, interleukin-10, tumour necrosis factor-alpha, or microglia activation and density. We report that in the short-term and in the context of later-life stress, repeated maternal separation has pro-inflammatory immune consequences in diverse tissues. Repeated maternal separation animals exhibit greater microglial activation and elevated pro-inflammatory cytokine signalling in key brain regions implicated in human psychiatric disorders. Notably, repeated maternal separation generally has no long-term effect on cytokine expression in any tissue in the absence of later-life stress. These observations suggest that the elevated inflammatory signalling that has been reported in humans with a history of early-life stress may be the joint consequence of ongoing stressor exposure together with potentiated neural and/or immune responsiveness to stressors. Finally, our findings provide detailed guidance for future studies interrogating the causal roles of early-life stress and inflammation in disorders such as major depression.
Collapse
Affiliation(s)
- Ethan G. Dutcher
- Department of Psychology, University of Cambridge, Cambridge, UK
| | | | - Mary-Ellen Lynall
- Department of Psychiatry, University of Cambridge, Cambridge, UK
- Molecular Immunity Unit, MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Shahid Khan
- GlaxoSmithKline Research & Development, Stevenage, UK
| | | | | | | | - Jeffrey W. Dalley
- Department of Psychology, University of Cambridge, Cambridge, UK
- Department of Psychiatry, University of Cambridge, Cambridge, UK
| |
Collapse
|
13
|
Liu J, Li B, Lee C, Zhu H, Zheng S, Pierro A. Protective effects of lactoferrin on injured intestinal epithelial cells. J Pediatr Surg 2019; 54:2509-2513. [PMID: 31668400 DOI: 10.1016/j.jpedsurg.2019.08.046] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 08/24/2019] [Indexed: 12/25/2022]
Abstract
PURPOSE Lactoferrin has been used as a milk supplement to prevent disease progression in necrotizing enterocolitis. However, the underlying mechanism is still unclear. We hypothesize that lactoferrin administration can modulate intestinal epithelial cell injury. METHODS We established an in vitro model of epithelial cell injury by treating rat intestinal epithelial cells IEC-18 and human Caco-2 cells with hydrogen peroxide (H2O2), while lactoferrin was added as treatment at the same time. Live/dead cells were detected by immunofluorescence. Gene expression of inflammatory markers interleukin-6 (IL-6), intestinal stem cells (Lgr5), and proliferation marker (Wnt/β-catenin) were measured. Data was presented as mean ± SEM and compared using one-way ANOVA. A p-value <0.05 was considered significant. RESULTS Compared to control cells, H2O2 induced cell death in both IEC-18 and Caco-2 cells, whereas treatment with lactoferrin maintained cell viability. In addition, lactoferrin reduced gene expression of IL-6, while it increased gene expression of Lgr5 and Wnt/β-catenin. CONCLUSIONS Intestinal cell injury can be induced by exposure to H2O2, mimicking epithelial damage during intestinal injury. This damage can be reversed by lactoferrin administration by reducing inflammation and inducing cell proliferation. Lactoferrin can be a potential pharmacological intervention for the prevention and recovery of intestinal epithelial injury.
Collapse
Affiliation(s)
- Jia Liu
- The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada; Department of Pediatric Surgery, Children's Hospital of Fudan University, Shanghai Key Laboratory of Birth Defect, Shanghai, China
| | - Bo Li
- The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Carol Lee
- The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Haitao Zhu
- The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada; Department of Pediatric Surgery, Children's Hospital of Fudan University, Shanghai Key Laboratory of Birth Defect, Shanghai, China
| | - Shan Zheng
- Department of Pediatric Surgery, Children's Hospital of Fudan University, Shanghai Key Laboratory of Birth Defect, Shanghai, China
| | - Agostino Pierro
- The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
14
|
Abstract
The developmental period constitutes a critical window of sensitivity to stress. Indeed, early-life adversity increases the risk to develop psychiatric diseases, but also gastrointestinal disorders such as the irritable bowel syndrome at adulthood. In the past decade, there has been huge interest in the gut-brain axis, especially as regards stress-related emotional behaviours. Animal models of early-life adversity, in particular, maternal separation (MS) in rodents, demonstrate lasting deleterious effects on both the gut and the brain. Here, we review the effects of MS on both systems with a focus on stress-related behaviours. In addition, we discuss more recent findings showing the impact of gut-directed interventions, including nutrition with pre- and probiotics, illustrating the role played by gut microbiota in mediating the long-term effects of MS. Overall, preclinical studies suggest that nutritional approaches with pro- and prebiotics may constitute safe and efficient strategies to attenuate the effects of early-life stress on the gut-brain axis. Further research is required to understand the complex mechanisms underlying gut-brain interaction dysfunctions after early-life stress as well as to determine the beneficial impact of gut-directed strategies in a context of early-life adversity in human subjects.
Collapse
|
15
|
Li B, Yu FZ, Minich A, Hock A, Lee C, Pierro A. Neonatal intestinal injury induced by maternal separation: pathogenesis and pharmacological targets 1. Can J Physiol Pharmacol 2018; 97:193-196. [PMID: 30383976 DOI: 10.1139/cjpp-2018-0370] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Maternal separation (MS) is a well-studied phenomenon thought to play a role in the pathogenesis of many diseases ranging from neuropsychiatric to early intestinal disorders such as necrotizing enterocolitis. The existing evidence suggests that MS initiates a variety of processes that in turn lead to early intestinal injury. Although there are many theories as to how MS alters normal physiological processes, the exact mechanism of action remains to be elucidated. This review aims to describe some of the pathological processes affecting the intestine that are caused by MS, including (i) brain-gut axis, (ii) intestinal epithelial barrier function, (iii) microbiome, (iv) oxidative stress and endoplasmic reticulum stress, and (v) gut inflammation.
Collapse
Affiliation(s)
- Bo Li
- a Division of General and Thoracic Surgery, Translation Medicine Program, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Fang Zhou Yu
- a Division of General and Thoracic Surgery, Translation Medicine Program, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada.,b School of Medicine, University of St Andrews, St Andrews, Fife, United Kingdom
| | - Adam Minich
- a Division of General and Thoracic Surgery, Translation Medicine Program, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Alison Hock
- a Division of General and Thoracic Surgery, Translation Medicine Program, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Carol Lee
- a Division of General and Thoracic Surgery, Translation Medicine Program, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Agostino Pierro
- a Division of General and Thoracic Surgery, Translation Medicine Program, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| |
Collapse
|
16
|
Neonatal- maternal separation primes zymogenic cells in the rat gastric mucosa through glucocorticoid receptor activity. Sci Rep 2018; 8:9823. [PMID: 29959361 PMCID: PMC6026145 DOI: 10.1038/s41598-018-28223-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 06/19/2018] [Indexed: 12/21/2022] Open
Abstract
Neonatal- Maternal Separation (NMS) deprives mammals from breastfeeding and maternal care, influencing growth during suckling- weaning transition. In the gastric mucosa, Mist1 (encoded by Bhlha15 gene) and moesin organize the secretory apparatus for pepsinogen C in zymogenic cells. Our current hypothesis was that NMS would change corticosterone activity through receptors (GR), which would modify molecules involved in zymogenic cell differentiation in rats. We found that NMS increased corticosterone levels from 18 days onwards, as GR decreased in the gastric mucosa. However, as nuclear GR was detected, we investigated receptor binding to responsive elements (GRE) and observed an augment in NMS groups. Next, we demonstrated that NMS increased zymogenic population (18 and and 30 days), and targeted Mist1 and moesin. Finally, we searched for evolutionarily conserved sequences that contained GRE in genes involved in pepsinogen C secretion, and found that the genomic regions of Bhlha15 and PgC contained sites highly likely to be responsive to glucocorticoids. We suggest that NMS triggers GR- GRE to enhance the expression and to prime genes that organize cellular architecture in zymogenic population for PgC function. As pepsinogen C- pepsin is essential for digestion, disturbance of parenting through NMS might alter functions of gastric mucosa in a permanent manner.
Collapse
|
17
|
Abstract
Hydrogen sulfide (H2S) is a novel signaling molecule most recently found to be of fundamental importance in cellular function as a regulator of apoptosis, inflammation, and perfusion. Mechanisms of endogenous H2S signaling are poorly understood; however, signal transmission is thought to occur via persulfidation at reactive cysteine residues on proteins. Although much has been discovered about how H2S is synthesized in the body, less is known about how it is metabolized. Recent studies have discovered a multitude of different targets for H2S therapy, including those related to protein modification, intracellular signaling, and ion channel depolarization. The most difficult part of studying hydrogen sulfide has been finding a way to accurately and reproducibly measure it. The purpose of this review is to: elaborate on the biosynthesis and catabolism of H2S in the human body, review current knowledge of the mechanisms of action of this gas in relation to ischemic injury, define strategies for physiological measurement of H2S in biological systems, and review potential novel therapies that use H2S for treatment.
Collapse
|
18
|
Jensen AR, Drucker NA, te Winkel JP, Ferkowicz MJ, Markel TA. The route and timing of hydrogen sulfide therapy critically impacts intestinal recovery following ischemia and reperfusion injury. J Pediatr Surg 2018; 53:1111-1117. [PMID: 29622397 PMCID: PMC5994359 DOI: 10.1016/j.jpedsurg.2018.02.072] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 02/27/2018] [Indexed: 01/16/2023]
Abstract
PURPOSE Hydrogen sulfide (H2S) has many beneficial properties and may serve as a novel treatment in patients suffering from intestinal ischemia-reperfusion injury (I/R). The purpose of this study was to examine the method of delivery and timing of administration of H2S for intestinal therapy during ischemic injury. We hypothesized that 1) route of administration of hydrogen sulfide would impact intestinal recovery following acute mesenteric ischemia and 2) preischemic H2S conditioning using the optimal mode of administration as determined above would provide superior protection compared to postischemic application. METHODS Male C57BL/6J mice underwent intestinal ischemia by temporary occlusion of the superior mesenteric artery. Following ischemia, animals were treated according to one of the following (N=6 per group): intraperitoneal or intravenous injection of GYY4137 (H2S-releasing donor, 50mg/kg in PBS), vehicle, inhalation of oxygen only, inhalation of 80ppm hydrogen sulfide gas. Following 24-h recovery, perfusion was assessed via laser Doppler imaging, and animals were euthanized. Perfusion and histology data were assessed, and terminal ileum samples were analyzed for cytokine production following ischemia. Once the optimal route of administration was determined, preischemic conditioning with H2S was undertaken using that route of administration. All data were analyzed using Mann-Whitney. P-values <0.05 were significant. RESULTS Mesenteric perfusion following intestinal I/R was superior in mice treated with intraperitoneal (IP) GYY4137 (IP vehicle: 25.6±6.0 vs. IP GYY4137: 79.7±15.1; p=0.02) or intravenous (IV) GYY4137 (IV vehicle: 36.3±5.9 vs. IV GYY4137: 100.7±34.0; p=0.03). This benefit was not observed with inhaled H2S gas (O2 vehicle: 66.6±11.4 vs. H2S gas: 81.8±6.0; p=0.31). However, histological architecture was only preserved with intraperitoneal administration of GYY4127 (IP vehicle: 3.4±0.4 vs. IP GYY4137: 2±0.3; p=0.02). Additionally, IP GYY4137 allowed for significant attenuation of inflammatory chemokine production of IL-6, IP-10 and MIP-2. We then analyzed whether there was a difference between pre- and postischemic administration of IP GYY4137. We found that preconditioning of animals with intraperitoneal GYY4137 only added minor improvements in outcomes compared to postischemic application. CONCLUSION Therapeutic benefits of H2S are superior with intraperitoneal application of an H2S donor compared to other administration routes. Additionally, while intraperitoneal treatment in both the pre- and postischemic period is beneficial, preischemic application of an H2S donor was found to be slightly better. Further studies are needed to examine long term outcomes and further mechanisms of action prior to widespread clinical application. TYPE OF STUDY Basic science. LEVEL OF EVIDENCE N/A.
Collapse
Affiliation(s)
- Amanda R. Jensen
- Department of Surgery, Section of Pediatric Surgery,The Indiana University School of Medicine Indianapolis, IN
| | - Natalie A. Drucker
- Department of Surgery, Section of Pediatric Surgery,The Indiana University School of Medicine Indianapolis, IN
| | - Jan P. te Winkel
- Department of Surgery, Section of Pediatric Surgery,The Indiana University School of Medicine Indianapolis, IN
| | - Michael J. Ferkowicz
- Department of Surgery, Section of Pediatric Surgery,The Indiana University School of Medicine Indianapolis, IN
| | - Troy A. Markel
- Department of Surgery, Section of Pediatric Surgery,Riley Hospital for Children at Indiana University Health,The Indiana University School of Medicine Indianapolis, IN
| |
Collapse
|
19
|
Lee C, Minich A, Li B, Miyake H, Seo S, Pierro A. Influence of stress factors on intestinal epithelial injury and regeneration. Pediatr Surg Int 2018; 34:155-160. [PMID: 29018960 DOI: 10.1007/s00383-017-4183-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/21/2017] [Indexed: 12/19/2022]
Abstract
PURPOSE Lgr5+ intestinal epithelial stem cells (ISCs) crucial for intestinal epithelial regeneration are impaired during necrotizing enterocolitis. This study aims to investigate the influence of different stressors on intestinal epithelial injury and regeneration in vitro. METHODS Intestinal epithelial cells (IEC-18) were exposed to stressors such as lipopolysaccharide, hydrogen peroxide, and serum. Cell viability was assessed using MTT assay at 18 and 24 h. IL-6 and Lgr5 gene expressions were measured using qPCR. RESULTS IEC-18 cell viability decreased 18 h following administration of lipopolysaccharide, hydrogen peroxide, and low serum concentration. However, after 24 h, the decrease in cell viability was observed only in higher, but not in lower concentrations of lipopolysaccharide and hydrogen peroxide. IL-6 expression increased in all groups compared to control. Lgr5 expression was up-regulated in cells exposed to a single stressor, but down-regulated when multiple stressors were administered. CONCLUSION Lipopolysaccharide, hydrogen peroxide, or low serum induced IEC-18 injury. The upregulation of Lgr5 expression after exposure to a single stressor suggests that minor injury to IEC-18 induces Lgr5+ ISCs to stimulate repair. Conversely, when IEC-18 cells were exposed to multiple stressors, Lgr5 expression was reduced. We speculate that this finding is similar to what happens in NEC when multiple stressors cause impairment of intestinal epithelium regeneration.
Collapse
Affiliation(s)
- Carol Lee
- Division of General and Thoracic Surgery, Program of Translational Medicine, The Hospital for Sick Children, 1526-555 University Ave, Toronto, ON M5G 1X8, Canada
| | - Adam Minich
- Division of General and Thoracic Surgery, Program of Translational Medicine, The Hospital for Sick Children, 1526-555 University Ave, Toronto, ON M5G 1X8, Canada
| | - Bo Li
- Division of General and Thoracic Surgery, Program of Translational Medicine, The Hospital for Sick Children, 1526-555 University Ave, Toronto, ON M5G 1X8, Canada
| | - Hiromu Miyake
- Division of General and Thoracic Surgery, Program of Translational Medicine, The Hospital for Sick Children, 1526-555 University Ave, Toronto, ON M5G 1X8, Canada
| | - Shogo Seo
- Division of General and Thoracic Surgery, Program of Translational Medicine, The Hospital for Sick Children, 1526-555 University Ave, Toronto, ON M5G 1X8, Canada
| | - Agostino Pierro
- Division of General and Thoracic Surgery, Program of Translational Medicine, The Hospital for Sick Children, 1526-555 University Ave, Toronto, ON M5G 1X8, Canada.
| |
Collapse
|
20
|
Inhibition of corticotropin-releasing hormone receptor 1 and activation of receptor 2 protect against colonic injury and promote epithelium repair. Sci Rep 2017; 7:46616. [PMID: 28492284 PMCID: PMC5425914 DOI: 10.1038/srep46616] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 03/21/2017] [Indexed: 12/16/2022] Open
Abstract
Maternal separation (MS) in neonates can lead to intestinal injury. MS in neonatal mice disrupts mucosal morphology, induces colonic inflammation and increases trans-cellular permeability. Several studies indicate that intestinal epithelial stem cells are capable of initiating gut repair in a variety of injury models but have not been reported in MS. The pathophysiology of MS-induced gut injury and subsequent repair remains unclear, but communication between the brain and gut contribute to MS-induced colonic injury. Corticotropin-releasing hormone (CRH) is one of the mediators involved in the brain–gut axis response to MS-induced damage. We investigated the roles of the CRH receptors, CRHR1 and CRHR2, in MS-induced intestinal injury and subsequent repair. To distinguish their specific roles in mucosal injury, we selectively blocked CRHR1 and CRHR2 with pharmacological antagonists. Our results show that in response to MS, CRHR1 mediates gut injury by promoting intestinal inflammation, increasing gut permeability, altering intestinal morphology, and modulating the intestinal microbiota. In contrast, CRHR2 activates intestinal stem cells and is important for gut repair. Thus, selectively blocking CRHR1 and promoting CRHR2 activity could prevent the development of intestinal injuries and enhance repair in the neonatal period when there is increased risk of intestinal injury such as necrotizing enterocolitis.
Collapse
|