1
|
Cai D, Tian B, Liang S, Cen Y, Fang J, Ma X, Zhong Z, Ren Z, Shen L, Gou L, Wang Y, Zuo Z. More Active Intestinal Immunity Developed by Obese Mice Than Non-Obese Mice After Challenged by Escherichia coli. Front Vet Sci 2022; 9:851226. [PMID: 35720836 PMCID: PMC9205201 DOI: 10.3389/fvets.2022.851226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Accepted: 03/21/2022] [Indexed: 11/13/2022] Open
Abstract
Obese mice presented lower mortality to non-fatal pneumonia induced by Escherichia coli (E. coli) than the non-obese mice. However, it remained obscure whether the intestine contributed to the protective effect of obese mice with infection. The 64 non-obese (NOB) mice were divided into NOB-uninfected and NOB-E. coli groups, while 64 high-fat diet-induced obesity (DIO) mice were divided into DIO-uninfected and DIO-E. coli groups. Mice in E. coli groups were intranasally instilled with 40 μl E. coli (4.0 ×109 colony-forming units [CFUs]), while uninfected groups with the same volume of phosphate buffer saline (PBS). The T subsets of Intraepithelial lymphocytes (IELs) and lamina propria lymphocytes (LPLs) in the intestine were collected for flow cytometry analysis at 0, 12, 24, and 72 h post-infection, also the duodenum and colon were harvested to survey histopathological change. The results showed that the percentage of CD3+T cells in LPLs in DIO-E. coli group was significantly lower than that in the DIO-uninfected group after infection (p < 0.05). The percentage of CD4+T cells in IELs in NOB-E. coli was significantly lower than that in DIO-E. coli after infection (p < 0.05). The percentage of CD8+T cells in LPLs in NOB-E. coli was significantly lower than that in DIO-E. coli at 12 and 24 h (p < 0.05). The immunoglobulin A (IgA)+ cells in DIO-uninfected were higher than that in NOB-uninfected at all time points (p < 0.05). The IgA+ cells in DIO-E. coli were higher than that in DIO-uninfected at 12, 24, and 72 h (p < 0.05). The results revealed that the level of intestinal mucosal immunity in obese mice was more active than that in non-obese mice.
Collapse
Affiliation(s)
- Dongjie Cai
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Bin Tian
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Shuang Liang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Yao Cen
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Jing Fang
- Key Laboratory of Animal Disease and Human Health of Sichuan, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Xiaoping Ma
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Zhijun Zhong
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Zhihua Ren
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Liuhong Shen
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Liping Gou
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Ya Wang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Zhicai Zuo
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
- *Correspondence: Zhicai Zuo
| |
Collapse
|
2
|
Sarasola MDLP, Táquez Delgado MA, Nicoud MB, Medina VA. Histamine in cancer immunology and immunotherapy. Current status and new perspectives. Pharmacol Res Perspect 2021; 9:e00778. [PMID: 34609067 PMCID: PMC8491460 DOI: 10.1002/prp2.778] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 03/25/2021] [Indexed: 12/11/2022] Open
Abstract
Cancer is the second leading cause of death globally and its incidence and mortality are rapidly increasing worldwide. The dynamic interaction of immune cells and tumor cells determines the clinical outcome of cancer. Immunotherapy comes to the forefront of cancer treatments, resulting in impressive and durable responses but only in a fraction of patients. Thus, understanding the characteristics and profiles of immune cells in the tumor microenvironment (TME) is a necessary step to move forward in the design of new immunomodulatory strategies that can boost the immune system to fight cancer. Histamine produces a complex and fine-tuned regulation of the phenotype and functions of the different immune cells, participating in multiple regulatory responses of the innate and adaptive immunity. Considering the important actions of histamine-producing immune cells in the TME, in this review we first address the most important immunomodulatory roles of histamine and histamine receptors in the context of cancer development and progression. In addition, this review highlights the current progress and foundational developments in the field of cancer immunotherapy in combination with histamine and pharmacological compounds targeting histamine receptors.
Collapse
Affiliation(s)
- María de la Paz Sarasola
- Laboratory of Tumor Biology and Inflammation, Institute for Biomedical Research (BIOMED), School of Medical Sciences, Pontifical Catholic University of Argentina (UCA), and the National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina
| | - Mónica A Táquez Delgado
- Laboratory of Tumor Biology and Inflammation, Institute for Biomedical Research (BIOMED), School of Medical Sciences, Pontifical Catholic University of Argentina (UCA), and the National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina
| | - Melisa B Nicoud
- Laboratory of Tumor Biology and Inflammation, Institute for Biomedical Research (BIOMED), School of Medical Sciences, Pontifical Catholic University of Argentina (UCA), and the National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina
| | - Vanina A Medina
- Laboratory of Tumor Biology and Inflammation, Institute for Biomedical Research (BIOMED), School of Medical Sciences, Pontifical Catholic University of Argentina (UCA), and the National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina
| |
Collapse
|
3
|
Ghosh R, Chatterjee S, Dubey S, Lavie CJ. Famotidine Against SARS-CoV2: A Hope or Hype? Mayo Clin Proc 2020; 95:1797-1799. [PMID: 32753153 PMCID: PMC7275146 DOI: 10.1016/j.mayocp.2020.05.027] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 05/27/2020] [Indexed: 12/17/2022]
Affiliation(s)
- Ritwik Ghosh
- Department of General Medicine, Burdwan Medical College and Hospital, Burdwan, West Bengal, India
| | - Subhankar Chatterjee
- Department of General Medicine, Rajendra Institute of Medical Sciences, Ranchi, Jharkhand, India
| | - Souvik Dubey
- Department of Neuromedicine, Bangur Institute of Neurosciences, Institute of Post Graduate Medical Education and Research and SSKM Hospital, Kolkata, West Bengal, India
| | - Carl J Lavie
- Department of Cardiovascular Diseases, John Ochsner Heart and Vascular Institute, Ochsner Clinical School, The University of Queensland School of Medicine, New Orleans, LA
| |
Collapse
|
4
|
Elieh Ali Komi D, Ribatti D. Mast cell-mediated mechanistic pathways in organ transplantation. Eur J Pharmacol 2019; 857:172458. [PMID: 31202799 DOI: 10.1016/j.ejphar.2019.172458] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 06/11/2019] [Accepted: 06/12/2019] [Indexed: 02/02/2023]
Abstract
Adaptive immunity has gained importance in transplant immunology for years, based on models in which T-cells orchestrate the immune responses during rejection. Most recently, researches revealed that innate immune cells, including mast cells (MCs) also play a pivotal role in allograft rejection. MC mediated immunoregulatory responses influence the innate and adaptive immune responses. Their capability to produce an array of both pro-inflammatory and anti-inflammatory mediators, expressing a wide range of costimulatory molecules in addition to acting as antigen-presenting cells (APCs), make them effective immune cells far beyond their classical role as primary orchestrator cells of allergy. Activated regulatory Tcells (Treg) cells contribute to MC recruitment into grafts by releasing interleukin (IL)-9. Tregs are capable of stabilizing MCs and suppressing IgE mediated degranulation through interaction of Treg expressing OX40 with MCs expressing OX40L. MCs in turn release transforming growth factor (TGF)-β and IL-10 which possess suppressive properties. Thus, these cells can suppress the proliferation of T-cells and support the generation of Tregs. MCs in addition to orchestrating immune responses in grafts by cell-to-cell interactions with variety of immune cells, cause histologic changes, mainly fibrosis by releasing mediators such as histamine, fibroblast growth factor-2 (FGF-2), TGF-β, chymase, and cathepsin G. The role of MCs in transplant rejection remains controversial. The accumulation of MCs in rejected grafts suggests that they play a role in preventing graft tolerance, and contribute to the progression of chronic rejection of allografts. However, high expression of MC-related gene products in tolerant grafts and their known interaction with Tregs on the other hand, support the notion that they are an integral component in achieving peripheral tolerance.
Collapse
Affiliation(s)
- Daniel Elieh Ali Komi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Domenico Ribatti
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy.
| |
Collapse
|
5
|
Santos VS, Freire MS, Santana RNS, Martins-Filho PRS, Cuevas LE, Gurgel RQ. Association between histamine-2 receptor antagonists and adverse outcomes in neonates: A systematic review and meta-analysis. PLoS One 2019; 14:e0214135. [PMID: 30947259 PMCID: PMC6448909 DOI: 10.1371/journal.pone.0214135] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 03/07/2019] [Indexed: 12/25/2022] Open
Abstract
Background The use of histamine-2 receptor antagonists (H2RA) in neonates is still debated because of possible risk of infection, necrotizing enterocolitis (NEC) and increased mortality. Aim To review whether the use of H2RA in neonates admitted to neonatal intensive care units (NICU) is associated with infection, NEC or mortality. Materials and method We performed a systematic search in PubMed, Web of Science and SCOPUS databases using the terms “histamine-2 receptor antagonists”, “infection”, “necrotizing enterocolitis”, “mortality”, “neonates” and related terms to identify studies published up to April 30, 2017. We included studies conducted in hospitalized neonates and exposed to H2RA. The primary outcomes were infection, NEC and mortality. We included reports of infections with clinical signs and positive culture, and NEC according to Bell stages (stage ≥II) based on standardised clinical and radiologic criteria. Among 1,144 studies identified, 10 fulfilled the selection criteria. Information extracted included study design, sample size and number of participants, along with the outcomes of interest. We conducted a meta-analysis of adjusted data and pooled estimates of infection, NEC and mortality are reported as odds ratios (OR) and 95% confidence intervals (95%CI). Results Ten studies were analysed. There were substantial associations between H2RA and infection (pooled OR: 2.09; 95%CI: 1.35–3.24; P = 0.001) and NEC (pooled OR: 2.81, 95%CI: 1.19–6.64; P = 0.02) but not with the mortality risk (pooled OR: 1.76; 95%CI: 0.50–6.16; P: 0.38). Conclusion Current evidence suggests that H2RA is associated with an increased risk of infection and NEC, but not with mortality in neonates admitted to NICU. The use of H2RA in neonates must be stringently considered when necessary.
Collapse
Affiliation(s)
- Victor S. Santos
- Centre for Epidemiology and Public Health. Federal University of Alagoas, Arapiraca, Brazil
| | - Marina S. Freire
- Department of Medicine. Federal University of Sergipe, Aracaju, Brazil
| | | | - Paulo R. S. Martins-Filho
- Postgraduate Programme in Health Science. Federal University of Sergipe, Aracaju, Brazil
- Investigative Pathology Laboratory, Federal University of Sergipe, Brazil
| | - Luis E. Cuevas
- Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Ricardo Q. Gurgel
- Department of Medicine. Federal University of Sergipe, Aracaju, Brazil
- Postgraduate Programme in Health Science. Federal University of Sergipe, Aracaju, Brazil
- * E-mail:
| |
Collapse
|
6
|
Jafarzadeh A, Nemati M, Khorramdelazad H, Hassan ZM. Immunomodulatory properties of cimetidine: Its therapeutic potentials for treatment of immune-related diseases. Int Immunopharmacol 2019; 70:156-166. [PMID: 30802678 DOI: 10.1016/j.intimp.2019.02.026] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 02/06/2019] [Accepted: 02/13/2019] [Indexed: 12/27/2022]
Abstract
Histamine exerts potent modulatory impacts on the cells of innate- [including neutrophils, monocytes, macrophages, dendritic cells (DCs), natural killer (NK) cells and NKT cells] and adaptive immunity (such as Th1-, Th2-, Th17-, regulatory T-, CD8+ cytotoxic T cells, and B cells) through binding to histamine receptor 2 (H2R). Cimetidine, as an H2R antagonist, reverses the histamine-mediated immunosuppression, as it has powerful stimulatory effects on the effector functions of neutrophils, monocytes, macrophages, DCs, NK cells, NKT cells, Th1-, Th2-, Th17-, and CD8+ cytotoxic T cells. However, cimetidine reduces the regulatory/suppressor T cell-mediated immunosuppression. Experimentally, cimetidine potentiate some immunologic activities in vitro and in vivo. The therapeutic potentials of cimetidine as an immunomodulatory agent were also investigated in a number of human diseases (such as cancers, viral warts, allergic disorders, burn, and bone resorption) and vaccination. This review aimed to provide a concise summary regarding the impacts of cimetidine on the immune system and highlight the cellular mechanisms of action and the immunomodulatory effects of this drug in various diseases to give novel insights regarding the therapeutic potentials of this drug for treatment of immune-related disorders. The review encourages more investigations to consider the immunomodulatory characteristic of cimetidine for managing of immune-related disorders.
Collapse
Affiliation(s)
- Abdollah Jafarzadeh
- Department of Immunology, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran; Molecular Medicine Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.
| | - Maryam Nemati
- Department of Hematology and Laboratory Sciences, School of Para-Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Hossain Khorramdelazad
- Molecular Medicine Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran; Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | | |
Collapse
|
7
|
Manzoni P, García Sánchez R, Meyer M, Stolfi I, Pugni L, Messner H, Cattani S, Betta PM, Memo L, Decembrino L, Bollani L, Rinaldi M, Fioretti M, Quercia M, Maule M, Tavella E, Mussa A, Tzialla C, Laforgia N, Mosca F, Magaldi R, Mostert M, Farina D. Exposure to Gastric Acid Inhibitors Increases the Risk of Infection in Preterm Very Low Birth Weight Infants but Concomitant Administration of Lactoferrin Counteracts This Effect. J Pediatr 2018; 193:62-67.e1. [PMID: 29198543 DOI: 10.1016/j.jpeds.2017.09.080] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 09/26/2017] [Accepted: 09/28/2017] [Indexed: 01/08/2023]
Abstract
OBJECTIVE To investigate whether exposure to inhibitors of gastric acidity, such as H2 blockers or proton pump inhibitors, can independently increase the risk of infections in very low birth weight (VLBW) preterm infants in the neonatal intensive care unit. STUDY DESIGN This is a secondary analysis of prospectively collected data from a multicenter, randomized controlled trial of bovine lactoferrin (BLF) supplementation (with or without the probiotic Lactobacillus rhamnosus GG) vs placebo in prevention of late-onset sepsis (LOS) and necrotizing enterocolitis (NEC) in preterm infants. Inhibitors of gastric acidity were used at the recommended dosages/schedules based on the clinical judgment of attending physicians. The distribution of days of inhibitors of gastric acidity exposure between infants with and without LOS/NEC was assessed. The mutually adjusted effects of birth weight, gestational age, duration of inhibitors of gastric acidity treatment, and exposure to BLF were controlled through multivariable logistic regression. Interaction between inhibitors of gastric acidity and BLF was tested; the effects of any day of inhibitors of gastric acidity exposure were then computed for BLF-treated vs -untreated infants. RESULTS Two hundred thirty-five of 743 infants underwent treatment with inhibitors of gastric acidity, and 86 LOS episodes occurred. After multivariate analysis, exposure to inhibitors of gastric acidity remained significantly and independently associated with LOS (OR, 1.03; 95% CI, 1.008-1.067; P = .01); each day of inhibitors of gastric acidity exposure conferred an additional 3.7% odds of developing LOS. Risk was significant for Gram-negative (P < .001) and fungal (P = .001) pathogens, but not for Gram-positive pathogens (P = .97). On the test for interaction, 1 additional day of exposure to inhibitors of gastric acidity conferred an additional 7.7% risk for LOS (P = .003) in BLF-untreated infants, compared with 1.2% (P = .58) in BLF-treated infants. CONCLUSION Exposure to inhibitors of gastric acidity is significantly associated with the occurrence of LOS in preterm VLBW infants. Concomitant administration of BLF counteracts this selective disadvantage. TRIAL REGISTRATION isrctn.org: ISRCTN53107700.
Collapse
Affiliation(s)
- Paolo Manzoni
- Neonatology and NICU, S Anna Hospital, AOU Città della Salute e della Scienza, Torino, Italy.
| | - Ruben García Sánchez
- Neonatology and NICU, Complejo Asistencial Universitario de Salamanca, Salamanca, Spain
| | - Michael Meyer
- Neonatology and NICU, Middlemore Hospital, Auckland, New Zealand
| | - Ilaria Stolfi
- Neonatology, Azienda Ospedaliera Universitaria Policlinico Umberto I, Rome, Italy
| | - Lorenza Pugni
- NICU, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Università degli Studi di Milano, Milan, Italy
| | - Hubert Messner
- Neonatology and NICU, Ospedale Regionale, Bolzano/Bozen, Italy
| | - Silvia Cattani
- NICU, University of Modena and Reggio Emilia, Modena, Italy
| | | | - Luigi Memo
- UOC di Pediatria e Patologia Neonatale, Ospedale San Martino, Belluno, Italy
| | - Lidia Decembrino
- Patologia Neonatale, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Lina Bollani
- Patologia Neonatale, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | | | - Maria Fioretti
- Neonatology and NICU, S Anna Hospital, AOU Città della Salute e della Scienza, Torino, Italy
| | - Michele Quercia
- Cancer Epidemiology Unit, University of Turin, Department of Medical Sciences, Torino, Italy
| | - Milena Maule
- Cancer Epidemiology Unit, University of Turin, Torino, Italy
| | - Elena Tavella
- Neonatology and NICU, S Anna Hospital, AOU Città della Salute e della Scienza, Torino, Italy
| | - Alessandro Mussa
- Neonatology and NICU, S Anna Hospital, AOU Città della Salute e della Scienza, Torino, Italy
| | - Chryssoula Tzialla
- Patologia Neonatale, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Nicola Laforgia
- Cancer Epidemiology Unit, University of Turin, Department of Medical Sciences, Torino, Italy
| | - Fabio Mosca
- NICU, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Università degli Studi di Milano, Milan, Italy
| | | | - Michael Mostert
- Department of Pediatrics, University of Turin, Torino, Italy
| | - Daniele Farina
- Neonatology and NICU, S Anna Hospital, AOU Città della Salute e della Scienza, Torino, Italy
| | | |
Collapse
|
8
|
Santana RNS, Santos VS, Ribeiro-Júnior RF, Freire MS, Menezes MAS, Cipolotti R, Gurgel RQ. Use of ranitidine is associated with infections in newborns hospitalized in a neonatal intensive care unit: a cohort study. BMC Infect Dis 2017; 17:375. [PMID: 28558748 PMCID: PMC5450121 DOI: 10.1186/s12879-017-2482-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 05/22/2017] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND The inhibition of gastric acid secretion with ranitidine is frequently prescribed off-label to newborns admitted to neonatal intensive care units (NICU). Some studies show that the use of inhibitors of gastric acid secretion (IGAS) may predispose to infections and necrotising enterocolitis (NEC), but there are few data to confirm this association. This study aimed to compare the rates of neonatal infections and NEC among preterm infants (<37 weeks gestation) hospitalised in a NICU exposed or not to treatment with ranitidine. METHODS A retrospective cohort study was conducted with all consecutive preterm newborns admitted to a NICU between August-2014 and October-2015. The rates of infection, NEC, and death of newborns exposed or not to ranitidine were recorded. RESULTS A total of 300 newborns were enrolled, of which 115 had received ranitidine and 185 had not. The two groups were similar with regard to the main demographic and clinical characteristics. Forty-eight (41.7%) of the 115 infants exposed to ranitidine and 49 (26.5%) of the 185 infants not exposed were infected (RR = 1.6, 95%CI 1.1-2.2, p = 0.006). The late onset (>48 h) blood culture positive infection rate was higher in the group exposed to ranitidine than in the untreated group (13.0% vs. 3.8%, p = 0.001). There was no significant association between the use of ranitidine and NEC (Bell stage >II) (p = 0.36). The mortality rate risk was 4-fold higher in infants receiving ranitidine (16.5% vs. 8.6%, p < 0.001). CONCLUSION Ranitidine use in neonates was associated with an increased risk of infections and mortality, but not with NEC.
Collapse
Affiliation(s)
- Ruth N S Santana
- Department of Medicine, Federal University of Sergipe, R. Cláudio Batista, s/n - Cidade Nova, Aracaju, 49060-108, Brazil
| | - Victor S Santos
- Postgraduate Program in Health Sciences, Federal University of Sergipe, R. Cláudio Batista, s/n - Cidade Nova, Aracaju, 49060-108, Brazil
| | - Ruy F Ribeiro-Júnior
- Department of Medicine, Federal University of Sergipe, R. Cláudio Batista, s/n - Cidade Nova, Aracaju, 49060-108, Brazil
| | - Marina S Freire
- Department of Medicine, Federal University of Sergipe, R. Cláudio Batista, s/n - Cidade Nova, Aracaju, 49060-108, Brazil
| | - Maria A S Menezes
- Postgraduate Program in Health Sciences, Federal University of Sergipe, R. Cláudio Batista, s/n - Cidade Nova, Aracaju, 49060-108, Brazil
| | - Rosana Cipolotti
- Department of Medicine, Federal University of Sergipe, R. Cláudio Batista, s/n - Cidade Nova, Aracaju, 49060-108, Brazil.,Postgraduate Program in Health Sciences, Federal University of Sergipe, R. Cláudio Batista, s/n - Cidade Nova, Aracaju, 49060-108, Brazil
| | - Ricardo Q Gurgel
- Department of Medicine, Federal University of Sergipe, R. Cláudio Batista, s/n - Cidade Nova, Aracaju, 49060-108, Brazil. .,Postgraduate Program in Health Sciences, Federal University of Sergipe, R. Cláudio Batista, s/n - Cidade Nova, Aracaju, 49060-108, Brazil.
| |
Collapse
|
9
|
Abstract
BACKGROUND Digoxin, a cardiac glycoside used for the treatment of heart failure, was reported to inhibit the retinoid-related orphan receptor gamma t (RORγt) and attenuate the severity of experimental autoimmune encephalomyelitis and arthritis in mice. However, the effects of digoxin in a mice model of inflammatory bowel disease have not been elucidated. METHODS Colitis was induced in severe combined immunodeficiency mice by adoptive transfer of CD45RB CD4 T cells. Digoxin or a vehicle was injected into mice with colitis intraperitoneally every other day and changes in body weight were evaluated. After 6 to 8 weeks, the treated mice were killed and evaluated for histological score, T-cell subset, and cytokine messenger RNA (mRNA) expression in the colonic tissue. RESULTS Wasting disease and histological damage were significantly attenuated in digoxin-treated mice with colitis compared with those in the vehicle-treated mice. In addition, the mRNAs of Th17-related cytokines were downregulated, whereas those of interleukin-10 were upregulated in the colonic mucosa of digoxin-treated mice. However, unexpectedly, the mRNA expression level of tumor necrosis factor alpha did not decrease in the colonic mucosa of digoxin-treated mice with colitis. This observation suggests that digoxin may ameliorate colitis by a tumor necrosis factor alpha-independent pathway. CONCLUSIONS This study has shown for the first time that treatment with digoxin can ameliorate murine experimental colitis. This finding suggests that the suppression of Th17 using reagents such as digoxin could be effective in treating Crohn's disease refractory to anti-tumor necrosis factor alpha therapy.
Collapse
|
10
|
Lalic-Popovic M, Paunkovic J, Grujic Z, Golocorbin-Kon S, Vasovic V, Al-Salami H, Mikov M. The Effect of Diabetes and Hypertension on the Placental Permeation of the Hydrophilic Drug, Ranitidine. Placenta 2016; 48:144-150. [PMID: 27871467 DOI: 10.1016/j.placenta.2016.11.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Revised: 10/26/2016] [Accepted: 11/01/2016] [Indexed: 10/20/2022]
Abstract
INTRODUCTION Ranitidine is a hydrophilic weak base and an H2-receptor antagonist which is commonly used for gastroesophageal reflux, including during pregnancy. It has limited placental permeation and can be used as a pre-anesthetic antacid to prevent aspiration of acidic stomach contents. Recent studies suggest that diabetes and hypertension may influence placental permeation of hydrophilic drugs. Thus, this study aimed to investigate the influence of diabetes and hypertension on ranitidine's placental permeation in pregnant women. METHODS Forty one pregnant women all scheduled for elective cesarean section entered the study: healthy control (n = 15), with hypertension (n = 16) and with gestational diabetes (n = 10). All women received 50 mg of ranitidine intravenously. Three samples of maternal plasma (after ranitidine application, at delivery and after delivery), and two umbilical cord samples (arterial and venous blood) were collected and analyzed for ranitidine concentrations. Maternal pharmacokinetic parameter were calculated as well as feto:maternal and umbilical cord arterial to venous concentration ratios. RESULTS Ranitidine maternal and umbilical cord (arterial and venous) concentrations were similar in all three groups and there were no difference between feto:maternal ratios nor volume of distribution, clearance and half life between the groups. DISCUSSION Fetal concentrations are dependent on maternal concentrations in healthy and hypertensive women but not in diabetic women. Hypertension and diabetes did not affect fetal handling of ranitidine. Though hypertension and diabetes did not influence ranitidine placental permeation, it appears they altered time needed to achieve unity between maternal and fetal plasma.
Collapse
Affiliation(s)
| | - Jovana Paunkovic
- Health Department in Novi Sad for Women Health Protection, Serbia
| | - Zorica Grujic
- University of Novi Sad, Faculty of Medicine, Department of Gynecology and Obstetistric, Serbia
| | | | - Velibor Vasovic
- University Novi Sad, Faculty of Medicine, Department of Pharmacology, Toxicology and Clinical Pharmacology, Serbia
| | - Hani Al-Salami
- Biotechnology and Drug Development Research Lab, School of Pharmacy, Curtin Health Innovation Research Institute, Curtin University, Perth WA, Australia
| | - Momir Mikov
- University Novi Sad, Faculty of Medicine, Department of Pharmacology, Toxicology and Clinical Pharmacology, Serbia
| |
Collapse
|
11
|
Deiteren A, De Man JG, Pelckmans PA, De Winter BY. Histamine H₄ receptors in the gastrointestinal tract. Br J Pharmacol 2015; 172:1165-78. [PMID: 25363289 DOI: 10.1111/bph.12989] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Revised: 09/28/2014] [Accepted: 10/20/2014] [Indexed: 12/13/2022] Open
Abstract
Histamine is a well-established mediator involved in a variety of physiological and pathophysiological mechanisms and exerts its effect through activation of four histamine receptors (H1-H₄). The histamine H₄ receptor is the newest member of this histamine receptor family, and is expressed throughout the gastrointestinal tract as well as in the liver, pancreas and bile ducts. Functional studies using a combination of selective and non-selective H₄ receptor ligands have rapidly increased our knowledge of H₄ receptor involvement in gastrointestinal processes both under physiological conditions and in models of disease. Strong evidence points towards a role for H₄ receptors in the modulation of immune-mediated responses in gut inflammation such as in colitis, ischaemia/reperfusion injury, radiation-induced enteropathy and allergic gut reactions. In addition, data have emerged implicating H₄ receptors in gastrointestinal cancerogenesis, sensory signalling, and visceral pain as well as in gastric ulceration. These studies highlight the potential of H₄ receptor targeted therapy in the treatment of various gastrointestinal disorders such as inflammatory bowel disease, irritable bowel syndrome and cancer.
Collapse
Affiliation(s)
- A Deiteren
- Laboratory of Experimental Medicine and Pediatrics, Division of Gastroenterology, University of Antwerp, Antwerp, Belgium
| | | | | | | |
Collapse
|
12
|
Qiu Y, Yang H. Effects of intraepithelial lymphocyte-derived cytokines on intestinal mucosal barrier function. J Interferon Cytokine Res 2013; 33:551-62. [PMID: 23692551 DOI: 10.1089/jir.2012.0162] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
The mucosal surface of the gastrointestinal tract directly interacts with the mucosal lumen, which is continuously exposed to foreign antigens. Specialized intraepithelial lymphocytes (IELs), located between the basolateral surfaces of the epithelial cells, are important as the first line of defense against microbes as well as for their role in the maintenance of epithelial barrier homeostasis. Although IELs are mainly composed of T cells, they are phenotypically and functionally distinct from T cells in peripheral blood or the spleen. Not only are IELs stimulated by the antigens of the intestinal lumen but are they also stimulated by regulatory immune cells. The integrity of the intestinal mucosal barrier is closely tied to the IEL function. Cytokines produced by IELs modulate the cellular functions that trigger the downstream signaling pathways and mediate the barrier homeostasis. In this review, we will address the broad spectrum of cytokines that are derived from IELs and the functional regulation of these cytokines on the intestinal barrier.
Collapse
Affiliation(s)
- Yuan Qiu
- Department of General Surgery, Xinqiao Hospital, Third Military Medical University , Chongqing, China
| | | |
Collapse
|
13
|
Abstract
BACKGROUND Changes in the intestinal and colonic proteome in patients with necrotizing enterocolitis (NEC) may help to characterize the disease pathology and identify new biomarkers and treatment targets for NEC. METHODS Using gel-based proteomics, proteins in NEC-affected intestinal and colonic sections were compared with those in adjacent, near-normal tissue sections within the same patients. Western blot and immunohistochemistry were applied to crossvalidate proteomic data and histological location of some selected proteins. RESULTS Thirty proteins were identified with differential expression between necrotic and vital small-intestine sections and 23 proteins were identified for colon sections. Five proteins were similarly affected in the small intestine and colon: histamine receptors (HRs), actins, globins, immunoglobulin, and antitrypsin. Two heat shock proteins (HSPs) were affected in the small intestine. Furthermore, proteins involved in antioxidation, angiogenesis, cytoskeleton formation, and metabolism were affected. Finally, secretory proteins such as antitrypsin, fatty-acid binding protein 5, and haptoglobin differed between NEC-affected and vital tissues. CONCLUSION NEC progression affects different pathways in the small intestine and colon. HSPs may play an important role, especially in the small intestine. The identified secretory proteins should be investigated as possible circulating markers of NEC progression in different gut regions.
Collapse
|
14
|
Statins directly suppress cytokine production in murine intraepithelial lymphocytes. Cytokine 2013; 61:540-5. [PMID: 23290865 DOI: 10.1016/j.cyto.2012.12.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2012] [Revised: 11/26/2012] [Accepted: 12/06/2012] [Indexed: 01/02/2023]
Abstract
Statins, inhibitors of the enzyme 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase, are known not only as cholesterol-lowering agents but also as anti-inflammatory mediators. However, their regulatory effect on intestinal mucosal immunity remains unclear. The present study examined the possible direct effects of statin on intestinal intraepithelial lymphocytes (IELs), the front line cells of the intestinal mucosal immune system. Murine IELs were isolated from the small intestines of C57BL/6 mice. IELs activated with anti-CD3/CD28 monoclonal antibodies produced interferon (IFN)-γ, tumor necrosis factor (TNF)-α, interleukin (IL)-2, and IL-4 in significant numbers; however, they did not produce IL-5. Both simvastatin and lovastatin suppressed IEL production of IFN-γ, TNF-α, IL-2, and IL-4 in a dose-dependent manner, whereas 48-h treatment with high concentrations (5 × 10(-5)M) of simvastatin and lovastatin did not affect the number of IELs. The suppressive effect of the simvastatin was significantly restored by the addition of mevalonate, farnesyl pyrophosphate ammonium salt, and geranylgeranyl pyrophosphate ammonium salt, which are downstream metabolites of HMG-CoA. These findings suggest that statins have direct suppressive effects on the production of T helper 1-cytokines and IL-4 in IELs; these effects are associated with inhibition of the mevalonate pathway to some extent.
Collapse
|
15
|
Takayanagi Y, Osawa S, Ikuma M, Takagaki K, Zhang J, Hamaya Y, Yamada T, Sugimoto M, Furuta T, Miyajima H, Sugimoto K. Norepinephrine suppresses IFN-γ and TNF-α production by murine intestinal intraepithelial lymphocytes via the β₁ adrenoceptor. J Neuroimmunol 2012; 245:66-74. [PMID: 22398028 DOI: 10.1016/j.jneuroim.2012.02.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2011] [Revised: 01/16/2012] [Accepted: 02/10/2012] [Indexed: 11/28/2022]
Abstract
We examined whether norepinephrine (NE) had direct effects on cytokine production by murine intestinal intraepithelial lymphocytes (IELs), compared with splenocytes. CD3⁺ IELs and CD3⁺ splenocytes expressed α(1B), α(1D), α(2C), β₁, β₂, and β₃ adrenoceptors (ARs). NE significantly suppressed IFN-γ and TNF-α production by IELs and splenocytes ex vivo. The suppressive effects of NE in IELs were reversed by β₁ AR antagonist CGP-20712A, whereas those in splenocytes were reversed by β₂ AR antagonist ICI118,551. In IELs, β₁ AR agonist xamoterol mimicked the suppressive effects of NE. These results indicated NE regulates intestinal mucosal immune responses mediated by IELs via β₁ AR.
Collapse
Affiliation(s)
- Yasuhiro Takayanagi
- First Department of Medicine, Hamamatsu University School of Medicine, Hamamatsu 431-3192, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Terrin G, Passariello A, De Curtis M, Manguso F, Salvia G, Lega L, Messina F, Paludetto R, Canani RB. Ranitidine is associated with infections, necrotizing enterocolitis, and fatal outcome in newborns. Pediatrics 2012; 129:e40-5. [PMID: 22157140 DOI: 10.1542/peds.2011-0796] [Citation(s) in RCA: 159] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND AND OBJECTIVES Gastric acidity is a major nonimmune defense mechanism against infections. The objective of this study was to investigate whether ranitidine treatment in very low birth weight (VLBW) infants is associated with an increased risk of infections, necrotizing enterocolitis (NEC), and fatal outcome. METHODS Newborns with birth weight between 401 and 1500 g or gestational age between 24 and 32 weeks, consecutively observed in neonatal intensive care units, were enrolled in a multicenter prospective observational study. The rates of infectious diseases, NEC, and death in enrolled subjects exposed or not to ranitidine were recorded. RESULTS We evaluated 274 VLBW infants: 91 had taken ranitidine and 183 had not. The main clinical and demographic characteristics did not differ between the 2 groups. Thirty-four (37.4%) of the 91 children exposed to ranitidine and 18 (9.8%) of the 183 not exposed to ranitidine had contracted infections (odds ratio 5.5, 95% confidence interval 2.9-10.4, P < .001). The risk of NEC was 6.6-fold higher in ranitidine-treated VLBW infants (95% confidence interval 1.7-25.0, P = .003) than in control subjects. Mortality rate was significantly higher in newborns receiving ranitidine (9.9% vs 1.6%, P = .003). CONCLUSIONS Ranitidine therapy is associated with an increased risk of infections, NEC, and fatal outcome in VLBW infants. Caution is advocated in the use of this drug in neonatal age.
Collapse
Affiliation(s)
- Gianluca Terrin
- Department of Women’s Health and Territorial Medicine, University La Sapienza, Rome, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
de Vries VC, Noelle RJ. Mast cell mediators in tolerance. Curr Opin Immunol 2010; 22:643-8. [PMID: 20884193 DOI: 10.1016/j.coi.2010.08.015] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2010] [Accepted: 08/31/2010] [Indexed: 01/02/2023]
Abstract
Our knowledge on the function of mast cells (MC) as part of the immune system has expanded from 'key cells in mediating allergy' to 'tunable regulators of the immune response'. Over the past years however, a large body of evidence has been presented indicating a more regulatory role for MC in the immune system by both contact dependent and independent mechanisms. Considering the vast amount of soluble mediators released by MC, it is not surprising that some are involved in the maintenance of peripheral tolerance and the control or even help to resolve ongoing inflammation. In this review we will focus on the immunosuppressive function of some of these mediators produced by MC in a wide variety of disease models.
Collapse
Affiliation(s)
- Victor C de Vries
- Department of Microbiology and Immunology, Dartmouth Medical School, Lebanon, NH, USA
| | | |
Collapse
|
18
|
Bozarov A, Wang YZ, Yu JG, Wunderlich J, Hassanain HH, Alhaj M, Cooke HJ, Grants I, Ren T, Christofi FL. Activation of adenosine low-affinity A3 receptors inhibits the enteric short interplexus neural circuit triggered by histamine. Am J Physiol Gastrointest Liver Physiol 2009; 297:G1147-62. [PMID: 19808660 PMCID: PMC2850084 DOI: 10.1152/ajpgi.00295.2009] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
We tested the novel hypothesis that endogenous adenosine (eADO) activates low-affinity A3 receptors in a model of neurogenic diarrhea in the guinea pig colon. Dimaprit activation of H2 receptors was used to trigger a cyclic coordinated response of contraction and Cl(-) secretion. Contraction-relaxation was monitored by sonomicrometry (via intracrystal distance) simultaneously with short-circuit current (I(sc), Cl(-) secretion). The short interplexus reflex coordinated response was attenuated or abolished by antagonists at H2 (cimetidine), 5-hydroxytryptamine 4 receptor (RS39604), neurokinin-1 receptor (GR82334), or nicotinic (mecamylamine) receptors. The A1 agonist 2-chloro-N(6)-cyclopentyladenosine (CCPA) abolished coordinated responses, and A1 antagonists could restore normal responses. A1-selective antagonists alone [8-cyclopentyltheophylline (CPT), 1,3-dipropyl-8-(2-amino-4-chlorophenyl)xanthine (PACPX), or 8-cyclopentyl-N(3)-[3-(4-(fluorosulfonyl)benzoyloxy)propyl]-xanthine (FSCPX)] caused a concentration-dependent augmentation of crypt cell secretion or contraction and acted at nanomolar concentrations. The A3 agonist N(6)-(3-iodobenzyl)-adenosine-5'-N-methyluronamide (IB-MECA) abolished coordinated responses and the A3 antagonist 3-ethyl-5-benzyl-2-methyl-4-phenylethynyl-6-phenyl-1,4-(+/-)-dihydropyridine-3,5-dicarboxylate (MRS1191) could restore and further augment responses. The IB-MECA effect was resistant to knockdown of adenosine A1 receptor with the irreversible antagonist FSCPX; the IC(50) for IB-MECA was 0.8 microM. MRS1191 alone could augment or unmask coordinated responses to dimaprit, and IB-MECA suppressed them. MRS1191 augmented distension-evoked reflex I(sc) responses. Adenosine deaminase mimicked actions of adenosine receptor antagonists. A3 receptor immunoreactivity was differentially expressed in enteric neurons of different parts of colon. After tetrodotoxin, IB-MECA caused circular muscle relaxation. The data support the novel concept that eADO acts at low-affinity A3 receptors in addition to high-affinity A1 receptors to suppress coordinated responses triggered by immune-histamine H2 receptor activation. The short interplexus circuit activated by histamine involves adenosine, acetylcholine, substance P, and serotonin. We postulate that A3 receptor modulation may occur in gut inflammatory diseases or allergic responses involving mast cell and histamine release.
Collapse
Affiliation(s)
- Andrey Bozarov
- Departments of 1 Anesthesiology and ,2Neuroscience, The Ohio State University, Columbus, Ohio
| | - Yu-Zhong Wang
- 2Neuroscience, The Ohio State University, Columbus, Ohio
| | | | | | | | | | - Helen J. Cooke
- 2Neuroscience, The Ohio State University, Columbus, Ohio
| | | | | | | |
Collapse
|