1
|
Oz B, Gunduz I, Karatas A, Koca SS. Hypouricemia in Behçet's Syndrome: Prevalence and Clinical Outcomes. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:739. [PMID: 40283030 PMCID: PMC12029136 DOI: 10.3390/medicina61040739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2025] [Revised: 04/07/2025] [Accepted: 04/15/2025] [Indexed: 04/29/2025]
Abstract
Background and Objectives: Behçet's syndrome (BS) is a systemic inflammatory disorder characterized by recurrent oral and genital ulcers, uveitis, and vascular involvement. Serum uric acid (SUA) has been implicated in various inflammatory conditions, due to its antioxidant properties and role in oxidative stress. Abnormal SUA levels, particularly hypouricemia, may influence inflammatory processes, but their significance in BS pathophysiology remains unexplored. This study aimed to determine the prevalence of abnormal SUA levels among BS patients and investigate their associations with its clinical manifestations and laboratory parameters. Materials and Methods: A retrospective analysis was conducted on 436 patients with complete data who met the international criteria for Behçet's syndrome, including 420 patients classified as hypouricemic or normouricemic, for detailed evaluation. Patients were classified as hypouricemic (<3 mg/dL), hyperuricemic (>7 mg/dL), or normouricemic (3-7 mg/dL). Data on sociodemographics, laboratory findings, and clinical characteristics were collected. Mortality and malignancy associations were analyzed using logistic regression. Inverse probability weighting (IPW) was employed to adjust for confounding factors. Results: Initial unadjusted analysis showed that hypouricemic BS patients had significantly lower rates of acneiform lesions (7.3% vs. 14.4%, p = 0.020) and vascular involvement (3.8% vs. 11.6%, p = 0.038) compared to normouricemic patients. However, after adjustment for confounding variables using the IPW methodology, these associations lost statistical significance (p = 0.592 and p = 0.519, respectively). Both before and after adjustment, no significant differences were observed between groups regarding major organ involvement, disease severity, or activity markers. Conclusions: After controlling for confounding factors, hypouricemia in BS patients did not demonstrate significant associations with specific clinical manifestations or disease outcomes. While the unadjusted data initially suggested potential relationships with acneiform lesions and vascular involvement, these associations were not supported by comprehensive statistical analysis. Further prospective studies are warranted to elucidate the complex relationship between uric acid metabolism and BS pathophysiology.
Collapse
Affiliation(s)
- Burak Oz
- Department of Rheumatology, Firat University Faculty of Medicine, 23200 Elazig, Turkey; (B.O.); (S.S.K.)
| | - Ibrahım Gunduz
- Department of Rheumatology, Selahaddin Eyyubi State Hospital, 21100 Diyarbakir, Turkey;
| | - Ahmet Karatas
- Department of Rheumatology, Firat University Faculty of Medicine, 23200 Elazig, Turkey; (B.O.); (S.S.K.)
| | - Suleyman S. Koca
- Department of Rheumatology, Firat University Faculty of Medicine, 23200 Elazig, Turkey; (B.O.); (S.S.K.)
| |
Collapse
|
2
|
Jalali A, Jafari F, Behnamrad S, Zarshenas MM, Zhang X, Kashkooe A. The Genus Paeonia: A Review of the Targeted Signaling Pathways and Underlying Mechanisms of Pharmacological and Clinical Properties. Curr Drug Discov Technol 2025; 22:e100724231842. [PMID: 38988165 DOI: 10.2174/0115701638318395240703115522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 05/15/2024] [Accepted: 05/29/2024] [Indexed: 07/12/2024]
Abstract
INTRODUCTION The Paeoniaceae family contains only the Paeonia genus and is considered a major group of flowering plants. Several traditional and pharmacological applications of Paeoniaceae herbs have been described. This paper aimes to determine the pharmacological activities of the most prevalent herbs from the genus Paeonia by focusing on their underlying mechanism of action and signaling pathways, providing insight for further in-depth research on the medicinal resources of Paeonia. METHODS The "Paeoniaceae" keyword was searched from 1st January 1995 to 15th May 2024 through the PubMed and Scopus databases. Only papers related to pharmacology, pharmaceutics, and toxicology were extracted. The possible pharmacological activity of the Paeonia plants, including their underlying mechanisms of action and signaling pathways, was subsequently discussed. RESULTS Following our venture, only 15 Paeonia herbs were adequately evaluated for their pharmacological applications. Paeonia lactiflora Pall., Paeonia suffruticosa Andrews, and Paeonia emodi Royle are among the most prevalent Paeonia plants that have attracted increased attention in modern pharmacological studies. Paeonia herbs possess various pharmacological applications, such as antiinflammatory, anti-allergic, anticancer, antimicrobial, cardiovascular protective, cosmetic and skincare, radical scavenging, hepatoprotective and anti-ulcerative, anti-diabetic, musculoskeletal, and neuroprotective effects, and can be used as alternative therapies under critical medical conditions. CONCLUSION Among the applications of Paeonia herbs, anti-inflammatory and antioxidant activities are critical, as most other pharmacological effects are attributed to them. In other words, nuclear factor (NF)-κB and nuclear factor erythroid 2-related factor 2 (Nrf2) can be considered the most important signaling pathways involved in the pharmacological activity of Paeonia herbs.
Collapse
Affiliation(s)
- Atefeh Jalali
- Medicinal Plants Processing Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Phytopharmaceuticals (Traditional Pharmacy), School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fereshteh Jafari
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Shima Behnamrad
- Medicinal Plants Processing Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad M Zarshenas
- Medicinal Plants Processing Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Phytopharmaceuticals (Traditional Pharmacy), School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
- Epilepsy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Xiuxin Zhang
- Institute of Vegetables and Flowers, Key Laboratory of Biology and Genetic Improvement of Horticultural Crops, Chinese Academy of Agricultural Sciences, Ministry of Agriculture and Rural Affairs, Beijing, China
| | - Ali Kashkooe
- Medicinal Plants Processing Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
3
|
Walsh C, Lane JA, van Sinderen D, Hickey RM. Tailored Combinations of Human Milk Oligosaccharides Modulate the Immune Response in an In Vitro Model of Intestinal Inflammation. Biomolecules 2024; 14:1481. [PMID: 39766188 PMCID: PMC11727556 DOI: 10.3390/biom14121481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 11/15/2024] [Accepted: 11/19/2024] [Indexed: 01/15/2025] Open
Abstract
Infants rely on their developing immune system and the protective components of breast milk to defend against bacterial and viral pathogens, as well as immune disorders such as food allergies, prior to the introduction of solid foods. When breastfeeding is not feasible, fortified infant formula will most frequently be offered, usually based on a cow's milk-based substitute. The current study aimed to explore the immunomodulatory effects of combinations of commercially available human milk oligosaccharides (HMOs). An in vitro co-culture model of Caco-2 intestinal epithelial cells and THP-1 macrophages was established to replicate the hallmarks of intestinal inflammation and to evaluate the direct effects of different synthetic HMO combinations. Notably, a blend of the most prevalent fucosylated and sialylated HMOs, 2'-fucosyllactose (2'-FL) and 6'-siallylactose (6'-SL), respectively, resulted in decreased pro-inflammatory cytokine levels. These effects were dependent on the HMO concentration and on the HMO ratio resembling those in breastmilk. Interestingly, adding additional HMO structures did not enhance the anti-inflammatory effects. This research highlights the importance of carefully selecting HMO combinations in nutritional products, particularly for infant milk formulations, to effectively mimic the benefits associated with breastmilk.
Collapse
Affiliation(s)
- Clodagh Walsh
- Teagasc Food Research Centre, Moorepark, Fermoy, P61 C996 Cork, Ireland;
- Health and Happiness Group, H&H Research, P61 K202 Cork, Ireland;
- APC Microbiome Ireland and School of Microbiology, University College Cork, T12 YT20 Cork, Ireland;
| | - Jonathan A. Lane
- Health and Happiness Group, H&H Research, P61 K202 Cork, Ireland;
| | - Douwe van Sinderen
- APC Microbiome Ireland and School of Microbiology, University College Cork, T12 YT20 Cork, Ireland;
| | - Rita M. Hickey
- Teagasc Food Research Centre, Moorepark, Fermoy, P61 C996 Cork, Ireland;
- APC Microbiome Ireland and School of Microbiology, University College Cork, T12 YT20 Cork, Ireland;
| |
Collapse
|
4
|
Sallam M, Breuer R, Wrotniak B, Alibrahim O. Necrotizing Enterocolitis Complicating Severe RSV Bronchiolitis in PICU Settings. Clin Pediatr (Phila) 2024; 63:1544-1550. [PMID: 38303673 DOI: 10.1177/00099228241227763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2024]
Abstract
This retrospective study aims to analyze the baseline characteristics and factors associated with poor outcomes in patients with necrotizing enterocolitis (NEC) complicating respiratory syncytial virus (RSV) infection. Using the Virtual Pediatric Systems data registry, patients under 2 years admitted to the pediatric intensive care unit (PICU) were screened. Patients with documented RSV infection and NEC, intestinal perforation, noninfectious gastroenteritis/colitis, or pneumatosis intestinalis occurring around the timing of RSV bronchiolitis diagnosis were included. Out of the screened patients, 41 were analyzed. Most patients (93%) were aged 30 days to 2 years, one-third had baseline anatomical cardiac defects, and 20% history of prematurity. Median PICU length of stay was 11.7 days. Seven patients died before hospital discharge. While not statistically significant, nonsurvivors tended to exhibit higher PRISM-3 scores, more acidemia, and lower systolic blood pressure. These findings emphasize the need for cautious assessment of gastrointestinal symptoms in critically ill patients with RSV infection.
Collapse
Affiliation(s)
- Mohammad Sallam
- University at Buffalo, Buffalo, NY, USA
- Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
| | | | | | | |
Collapse
|
5
|
Nofi CP, Prince JM, Brewer MR, Aziz M, Wang P. An anti-eCIRP strategy for necrotizing enterocolitis. Mol Med 2024; 30:156. [PMID: 39304832 PMCID: PMC11414128 DOI: 10.1186/s10020-024-00935-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 09/11/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND Necrotizing enterocolitis (NEC) is a severe gastrointestinal disease characterized by intestinal inflammation and injury, with high mortality risk. Extracellular cold-inducible RNA-binding protein (eCIRP) is a recently discovered damage-associated molecular pattern that propagates inflammation and tissue injury; however, the role of eCIRP in NEC remains unknown. We hypothesize that eCIRP exacerbates NEC pathogenesis and the novel eCIRP-scavenging peptide, milk fat globule-epidermal growth factor-factor VIII (MFG-E8)-derived oligopeptide 3 (MOP3), attenuates NEC severity, serving as a new therapeutic strategy to treat NEC. METHODS Stool samples from premature neonates were collected prospectively and eCIRP levels were measured. Wild-type (WT) and CIRP-/- mouse pups were subjected to NEC utilizing a combination of hypoxia and hypercaloric formula orogastric gavage with lipopolysaccharide supplementation. In parallel, WT pups were treated with MOP3 or vehicle. Endpoints including NEC severity, intestinal injury, barrier dysfunction, lung injury, and overall survival were determined. RESULTS Stool samples from NEC neonates had elevated eCIRP levels compared to healthy age-matched controls (p < 0.05). CIRP-/- pups were significantly protected from NEC severity, intestinal injury, bowel inflammation, intestinal barrier dysfunction, lung injury, and systemic inflammation. NEC survival was 100% for CIRP-/- pups compared to 65% for WT (p < 0.05). MOP3 treatment recapitulated the benefits afforded by CIRP-knockdown, preventing NEC severity, improving inflammatory profiles, and attenuating organ injury. MOP3 treatment improved NEC survival to 80% compared to 50% for vehicle treatment (p < 0.05). CONCLUSIONS eCIRP exacerbates NEC evidenced by protection with CIRP-deficiency and administration of MOP3, a CIRP-directed therapeutic, in a murine model. Thus, eCIRP is a novel target with human relevance, and MOP3 is a promising treatment for lethal NEC.
Collapse
Affiliation(s)
- Colleen P Nofi
- Center for Immunology and Inflammation, the Feinstein Institutes for Medical Research, 350 Community Dr., Manhasset, NY, 11030, USA
- Elmezzi Graduate School of Molecular Medicine, Manhasset, NY, 11030, USA
- Department of Surgery, Zucker School of Medicine, Manhasset, NY, 11030, USA
| | - Jose M Prince
- Center for Immunology and Inflammation, the Feinstein Institutes for Medical Research, 350 Community Dr., Manhasset, NY, 11030, USA
- Department of Surgery, Zucker School of Medicine, Manhasset, NY, 11030, USA
| | - Mariana R Brewer
- Department of Pediatrics, Zucker School of Medicine, Manhasset, NY, 11030, USA
| | - Monowar Aziz
- Center for Immunology and Inflammation, the Feinstein Institutes for Medical Research, 350 Community Dr., Manhasset, NY, 11030, USA.
- Elmezzi Graduate School of Molecular Medicine, Manhasset, NY, 11030, USA.
- Department of Surgery, Zucker School of Medicine, Manhasset, NY, 11030, USA.
- Department of Molecular Medicine, Zucker School of Medicine, Manhasset, NY, 11030, USA.
| | - Ping Wang
- Center for Immunology and Inflammation, the Feinstein Institutes for Medical Research, 350 Community Dr., Manhasset, NY, 11030, USA.
- Elmezzi Graduate School of Molecular Medicine, Manhasset, NY, 11030, USA.
- Department of Surgery, Zucker School of Medicine, Manhasset, NY, 11030, USA.
- Department of Molecular Medicine, Zucker School of Medicine, Manhasset, NY, 11030, USA.
| |
Collapse
|
6
|
Zhang Y, Tian Y, Zhong X, Zhang R, Yang S, Jin J, Lyu C, Fan J, Shi B, Zhu K, Xiao Y, Lin N, Ma D, Tou J, Shu Q, Lai D. RNF31-mediated IKKα ubiquitination aggravates inflammation and intestinal injury through regulating NF-κB activation in human and mouse neonates. Life Sci 2024; 352:122893. [PMID: 38971367 DOI: 10.1016/j.lfs.2024.122893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 06/22/2024] [Accepted: 07/03/2024] [Indexed: 07/08/2024]
Abstract
AIMS Neonatal necrotizing enterocolitis (NEC) is a leading cause of intestine inflammatory disease, and macrophage is significantly activated during NEC development. Posttranslational modifications (PTMs) of proteins, particularly ubiquitination, play critical roles in immune response. This study aimed to investigate the effects of ubiquitin-modified proteins on macrophage activation and NEC, and discover novel NEC-related inflammatory proteins. MATERIALS AND METHODS Proteomic and ubiquitin proteomic analyses of intestinal macrophages in NEC/healthy mouse pups were carried out. In vitro macrophage inflammation model and in vivo NEC mouse model, as well as clinical human samples were used for further verification the inhibitor of nuclear factor-κB kinase α (IKKα) ubiquitination on NEC development through Western blot, immunofluorescence, quantitative real-time polymerase chain reaction (qRT-PCR) and flow cytometry. KEY FINDINGS We report here that IKKα was a new ubiquitin-modified protein during NEC through ubiquitin proteomics, and RING finger protein 31 (RNF31) acted as an E3 ligase to be involved in IKKα degradation. Inhibition of IKKα ubiquitination and degradation with siRNF31 or proteasome inhibitor decreased nuclear factor-κB (NF-κB) activation, thereby decreasing the expression of pro-inflammatory factors and M1 macrophage polarization, resulting in reliving the severity of NEC. SIGNIFICANCE Our study suggests the activation of RNF31-IKKα-NF-κB axis triggering NEC development and suppressing RNF31-mediated IKKα degradation may be therapeutic strategies to be developed for NEC treatment.
Collapse
Affiliation(s)
- Yuebai Zhang
- Department of Thoracic and Cardiovascular Surgery, Children's hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Yangfan Tian
- Department of Thoracic and Cardiovascular Surgery, Children's hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Xiaohui Zhong
- Department of Thoracic and Cardiovascular Surgery, Children's hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Ruoyang Zhang
- Department of Thoracic and Cardiovascular Surgery, Children's hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Sisi Yang
- Department of Neonatal Surgery, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Jingyi Jin
- Department of Thoracic and Cardiovascular Surgery, Children's hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Chengjie Lyu
- Department of Neonatal Surgery, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Jiajie Fan
- Department of Thoracic and Cardiovascular Surgery, Children's hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Bo Shi
- Department of Pediatric Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Kun Zhu
- Department of Pathology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Yi Xiao
- Department of Thoracic and Cardiovascular Surgery, Children's hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Nan Lin
- Department of Neonatal Surgery, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Daqing Ma
- Perioperative and Systems Medicine, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Zhejiang, China; Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, UK
| | - Jinfa Tou
- Department of Neonatal Surgery, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China.
| | - Qiang Shu
- Department of Thoracic and Cardiovascular Surgery, Children's hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China.
| | - Dengming Lai
- Department of Neonatal Surgery, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China.
| |
Collapse
|
7
|
Sharda D, Attri K, Choudhury D. Greener healing: sustainable nanotechnology for advanced wound care. DISCOVER NANO 2024; 19:127. [PMID: 39136798 PMCID: PMC11322481 DOI: 10.1186/s11671-024-04061-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 06/29/2024] [Indexed: 08/16/2024]
Abstract
Wound healing involves a carefully regulated sequence of events, encompassing pro-inflammatory and anti-inflammatory stages, tissue regeneration, and remodeling. However, in individuals with diabetes, this process gets disrupted due to dysregulation caused by elevated glucose levels and pro-inflammatory cytokines in the bloodstream. Consequently, the pro-inflammatory stage is prolonged, while the anti-inflammatory phase is delayed, leading to impaired tissue regeneration and remodeling with extended healing time. Furthermore, the increased glucose levels in open wounds create an environment conducive to microbial growth and tissue sepsis, which can escalate to the point of limb amputation. Managing diabetic wounds requires meticulous care and monitoring due to the lack of widely available preventative and therapeutic measures. Existing clinical interventions have limitations, such as slow recovery rates, high costs, and inefficient drug delivery methods. Therefore, exploring alternative avenues to develop effective wound-healing treatments is essential. Nature offers a vast array of resources in the form of secondary metabolites, notably polyphenols, known for their antimicrobial, anti-inflammatory, antioxidant, glucose-regulating, and cell growth-promoting properties. Additionally, nanoparticles synthesized through environmentally friendly methods hold promise for wound healing applications in diabetic and non-diabetic conditions. This review provides a comprehensive discussion and summary of the potential wound-healing abilities of specific natural polyphenols and their nanoparticles. It explores the mechanisms of action underlying their efficacy and presents effective formulations for promoting wound-healing activity.
Collapse
Affiliation(s)
- Deepinder Sharda
- School of Chemistry and Biochemistry, Thapar Institute of Engineering and Technology, Patiala, Punjab, 147004, India
| | - Komal Attri
- School of Chemistry and Biochemistry, Thapar Institute of Engineering and Technology, Patiala, Punjab, 147004, India
- Thapar Institute of Engineering and Technology-Virginia Tech (USA) Centre of Excellence in Emerging Materials, Thapar Institute of Engineering and Technology, Patiala, Punjab, 147004, India
| | - Diptiman Choudhury
- School of Chemistry and Biochemistry, Thapar Institute of Engineering and Technology, Patiala, Punjab, 147004, India.
- Thapar Institute of Engineering and Technology-Virginia Tech (USA) Centre of Excellence in Emerging Materials, Thapar Institute of Engineering and Technology, Patiala, Punjab, 147004, India.
| |
Collapse
|
8
|
Filliaux S, Bertelsen C, Baughman H, Komives E, Lyubchenko Y. The Interaction of NF-κB Transcription Factor with Centromeric Chromatin. J Phys Chem B 2024; 128:5803-5813. [PMID: 38860885 DOI: 10.1021/acs.jpcb.3c08388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2024]
Abstract
Centromeric chromatin is a subset of chromatin structure and governs chromosome segregation. The centromere is composed of both CENP-A nucleosomes (CENP-Anuc) and H3 nucleosomes (H3nuc) and is enriched with alpha-satellite (α-sat) DNA repeats. These CENP-Anuc have a different structure than H3nuc, decreasing the base pairs (bp) of wrapped DNA from 147 bp for H3nuc to 121 bp for CENP-Anuc. All these factors can contribute to centromere function. We investigated the interaction of H3nuc and CENP-Anuc with NF-κB, a crucial transcription factor in regulating immune response and inflammation. We utilized atomic force microscopy (AFM) to characterize complexes of both types of nucleosomes with NF-κB. We found that NF-κB unravels H3nuc, removing more than 20 bp of DNA, and that NF-κB binds to the nucleosomal core. Similar results were obtained for the truncated variant of NF-κB comprised only of the Rel homology domain and missing the transcription activation domain (TAD), suggesting that RelATAD is not critical in unraveling H3nuc. By contrast, NF-κB did not bind to or unravel CENP-Anuc. These findings with different affinities for two types of nucleosomes to NF-κB may have implications for understanding the mechanisms of gene expression in bulk and centromere chromatin.
Collapse
Affiliation(s)
- Shaun Filliaux
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska 68198-6025, United States
| | - Chloe Bertelsen
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska 68198-6025, United States
| | - Hannah Baughman
- Department of Chemistry and Biochemistry, UC San Diego, La Jolla, California 92093-0378, United States
| | - Elizabeth Komives
- Department of Chemistry and Biochemistry, UC San Diego, La Jolla, California 92093-0378, United States
| | - Yuri Lyubchenko
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska 68198-6025, United States
| |
Collapse
|
9
|
Nofi CP, Prince JM, Wang P, Aziz M. Chromatin as alarmins in necrotizing enterocolitis. Front Immunol 2024; 15:1403018. [PMID: 38881893 PMCID: PMC11176418 DOI: 10.3389/fimmu.2024.1403018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 05/20/2024] [Indexed: 06/18/2024] Open
Abstract
Necrotizing enterocolitis (NEC) is a severe gastrointestinal disease primarily affecting premature neonates, marked by poorly understood pro-inflammatory signaling cascades. Recent advancements have shed light on a subset of endogenous molecular patterns, termed chromatin-associated molecular patterns (CAMPs), which belong to the broader category of damage-associated molecular patterns (DAMPs). CAMPs play a crucial role in recognizing pattern recognition receptors and orchestrating inflammatory responses. This review focuses into the realm of CAMPs, highlighting key players such as extracellular cold-inducible RNA-binding protein (eCIRP), high mobility group box 1 (HMGB1), cell-free DNA, neutrophil extracellular traps (NETs), histones, and extracellular RNA. These intrinsic molecules, often perceived as foreign, have the potential to trigger immune signaling pathways, thus contributing to NEC pathogenesis. In this review, we unravel the current understanding of the involvement of CAMPs in both preclinical and clinical NEC scenarios. We also focus on elucidating the downstream signaling pathways activated by these molecular patterns, providing insights into the mechanisms that drive inflammation in NEC. Moreover, we scrutinize the landscape of targeted therapeutic approaches, aiming to mitigate the impact of tissue damage in NEC. This in-depth exploration offers a comprehensive overview of the role of CAMPs in NEC, bridging the gap between preclinical and clinical insights.
Collapse
Affiliation(s)
- Colleen P. Nofi
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
- Elmezzi Graduate School of Molecular Medicine, Manhasset, NY, United States
- Department of Surgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, United States
| | - Jose M. Prince
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
- Department of Surgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, United States
| | - Ping Wang
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
- Elmezzi Graduate School of Molecular Medicine, Manhasset, NY, United States
- Department of Surgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, United States
- Department of Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, United States
| | - Monowar Aziz
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
- Elmezzi Graduate School of Molecular Medicine, Manhasset, NY, United States
- Department of Surgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, United States
- Department of Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, United States
| |
Collapse
|
10
|
Dicks LMT. Gut Bacteria Provide Genetic and Molecular Reporter Systems to Identify Specific Diseases. Int J Mol Sci 2024; 25:4431. [PMID: 38674014 PMCID: PMC11050607 DOI: 10.3390/ijms25084431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 04/15/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
With genetic information gained from next-generation sequencing (NGS) and genome-wide association studies (GWAS), it is now possible to select for genes that encode reporter molecules that may be used to detect abnormalities such as alcohol-related liver disease (ARLD), cancer, cognitive impairment, multiple sclerosis (MS), diabesity, and ischemic stroke (IS). This, however, requires a thorough understanding of the gut-brain axis (GBA), the effect diets have on the selection of gut microbiota, conditions that influence the expression of microbial genes, and human physiology. Bacterial metabolites such as short-chain fatty acids (SCFAs) play a major role in gut homeostasis, maintain intestinal epithelial cells (IECs), and regulate the immune system, neurological, and endocrine functions. Changes in butyrate levels may serve as an early warning of colon cancer. Other cancer-reporting molecules are colibactin, a genotoxin produced by polyketide synthetase-positive Escherichia coli strains, and spermine oxidase (SMO). Increased butyrate levels are also associated with inflammation and impaired cognition. Dysbiosis may lead to increased production of oxidized low-density lipoproteins (OX-LDLs), known to restrict blood vessels and cause hypertension. Sudden changes in SCFA levels may also serve as a warning of IS. Early signs of ARLD may be detected by an increase in regenerating islet-derived 3 gamma (REG3G), which is associated with changes in the secretion of mucin-2 (Muc2). Pro-inflammatory molecules such as cytokines, interferons, and TNF may serve as early reporters of MS. Other examples of microbial enzymes and metabolites that may be used as reporters in the early detection of life-threatening diseases are reviewed.
Collapse
Affiliation(s)
- Leon M T Dicks
- Department of Microbiology, Stellenbosch University, Stellenbosch 7600, South Africa
| |
Collapse
|
11
|
Malik S, Fatima B, Hussain D, Imran M, Chohan TA, Khan MS, Majeed S, Najam-Ul-Haq M. Synthesis of novel nonsteroidal anti-inflammatory galloyl β-sitosterol-loaded lignin-capped Ag-based drug. Inflammopharmacology 2024; 32:1333-1351. [PMID: 37994993 DOI: 10.1007/s10787-023-01390-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 10/27/2023] [Indexed: 11/24/2023]
Abstract
Biocompatible anti-inflammatory lignin-capped Ag (LCAg) nanoparticles (NPs) were synthesized for the delivery of galloyl β-sitosterol (Galloyl-BS). β-Sitosterol (BS) is effective against inflammatory responses, like cancer-induced inflammations. BS was modified via gallic acid esterification to enhance its anti-inflammatory potential. LCAg NPs were synthesized by a green method and loaded with galloyl-BS. For comparison, pure BS was also loaded onto LCAg NPs in a separate assembly. The antioxidant potential of Galloyl-BS was greater (IC50 177 µM) than pure BS. Materials were characterized by FT-IR, SEM, XRD, and Zeta potential. Using UV-Vis spectroscopy, drug release experiments were performed by varying pH, time, concentration, and temperature. Maximum drug release was observed after 18 h at pH 6 and 40 °C. Galloyl-BS showed improved drug loading efficiency, release %age, and antioxidant activity compared to pure BS when loaded onto LCAg NPs. DLCAg exhibited excellent anti-inflammatory activity in rat models. These findings indicate that galloyl-BS (drug)-loaded LCAg (DLCAg) NPs have the potential as an anti-inflammatory agent without any prior release and scavenging in normal cells.
Collapse
Affiliation(s)
- Sana Malik
- Department of Biochemistry, Bahauddin Zakariya University, Multan, 60800, Pakistan
| | - Batool Fatima
- Department of Biochemistry, Bahauddin Zakariya University, Multan, 60800, Pakistan.
| | - Dilshad Hussain
- HEJ Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Muhammad Imran
- Biochemistry Section Institute of Chemical Sciences, University of Peshawar, Peshawar, Pakistan
| | - Tahir Ali Chohan
- Department of Biochemistry, University of Veterinary and Animal Sciences, Lahore, Pakistan
| | | | - Saadat Majeed
- Institute of Chemical Sciences, Bahauddin Zakariya University, Multan, 60800, Pakistan
| | - Muhammad Najam-Ul-Haq
- Institute of Chemical Sciences, Bahauddin Zakariya University, Multan, 60800, Pakistan.
| |
Collapse
|
12
|
Filliaux S, Bertelsen C, Baughman H, Komives E, Lyubchenko YL. The Interaction of NF-κB Transcription Factor with Centromeric Chromatin. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.13.580208. [PMID: 38405937 PMCID: PMC10888803 DOI: 10.1101/2024.02.13.580208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Centromeric chromatin is a subset of chromatin structure and governs chromosome segregation. The centromere is composed of both CENP-A nucleosomes (CENP-A nuc ) and H3 nucleosomes (H3 nuc ) and is enriched with alpha-satellite (α-sat) DNA repeats. These CENP-A nuc have a different structure than H3 nuc , decreasing the base pairs (bp) of wrapped DNA from 147 bp for H3 nuc to 121 bp for CENP-A nuc . All these factors can contribute to centromere function. We investigated the interaction of H3 nuc and CENP-A nuc with NF-κB, a crucial transcription factor in regulating immune response and inflammation. We utilized Atomic Force Microscopy (AFM) to characterize complexes of both types of nucleosomes with NF-κB. We found that NF-κB unravels H3 nuc , removing more than 20 bp of DNA, and that NF-κB binds to the nucleosomal core. Similar results were obtained for the truncated variant of NF-κB comprised only of the Rel Homology domain and missing the transcription activation domain (TAD), suggesting the RelA TAD is not critical in unraveling H3 nuc . By contrast, NF-κB did not bind to or unravel CENP- A nuc . These findings with different affinities for two types of nucleosomes to NF-κB may have implications for understanding the mechanisms of gene expression in bulk and centromere chromatin.
Collapse
|
13
|
Sobotka AA, Tempera I. PARP1 as an Epigenetic Modulator: Implications for the Regulation of Host-Viral Dynamics. Pathogens 2024; 13:131. [PMID: 38392869 PMCID: PMC10891851 DOI: 10.3390/pathogens13020131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 01/23/2024] [Accepted: 01/24/2024] [Indexed: 02/25/2024] Open
Abstract
The principal understanding of the Poly(ADP-ribose) polymerase (PARP) regulation of genomes has been focused on its role in DNA repair; however, in the past few years, an additional role for PARPs and PARylation has emerged in regulating viral-host interactions. In particular, in the context of DNA virus infection, PARP1-mediated mechanisms of gene regulations, such as the involvement with cellular protein complexes responsible for the folding of the genome into the nucleus, the formation of chromatin loops connecting distant regulatory genomic regions, and other methods of transcriptional regulation, provide additional ways through which PARPs can modulate the function of both the host and the viral genomes during viral infection. In addition, potential viral amplification of the activity of PARPs on the host genome can contribute to the pathogenic effect of viral infection, such as viral-driven oncogenesis, opening the possibility that PARP inhibition may represent a potential therapeutic approach to target viral infection. This review will focus on the role of PARPs, particularly PARP1, in regulating the infection of DNA viruses.
Collapse
Affiliation(s)
- Asher A. Sobotka
- Wistar Institute, Philadelphia, PA 19104, USA
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | |
Collapse
|
14
|
Donadio JLS, Fabi JP, Sztein MB, Salerno-Gonçalves R. Dietary fiber pectin: challenges and potential anti-inflammatory benefits for preterms and newborns. Front Nutr 2024; 10:1286138. [PMID: 38283907 PMCID: PMC10811139 DOI: 10.3389/fnut.2023.1286138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 12/29/2023] [Indexed: 01/30/2024] Open
Abstract
Pectins, a class of dietary fibers abundant in vegetables and fruits, have drawn considerable interest due to their potential anti-inflammatory properties. Numerous studies have indicated that incorporating pectins into infant formula could be a safe strategy for alleviating infant regurgitation and diarrhea. Moreover, pectins have been shown to modulate cytokine production, macrophage activity, and NF-kB expression, all contributing to their anti-inflammatory effects. Despite this promising evidence, the exact mechanisms through which pectins exert these functions and how their structural characteristics influence these processes remain largely unexplored. This knowledge is particularly significant in the context of gut inflammation in developing preterm babies, a critical aspect of necrotizing enterocolitis (NEC), and in children and adults dealing with inflammatory bowel disease (IBD). Our mini review aims to provide an up-to-date compilation of relevant research on the effects of pectin on gut immune responses, specifically focusing on preterms and newborns. By shedding light on the underlying mechanisms and implications of pectin-mediated anti-inflammatory properties, this review seeks to advance our knowledge in this area and pave the way for future research and potential therapeutic interventions.
Collapse
Affiliation(s)
- Janaina L. S. Donadio
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Food Science and Experimental Nutrition, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
- Food Research Center (FoRC), CEPID-FAPESP (Research, Innovation and Dissemination Centers, São Paulo Research Foundation), São Paulo, Brazil
| | - João Paulo Fabi
- Department of Food Science and Experimental Nutrition, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
- Food Research Center (FoRC), CEPID-FAPESP (Research, Innovation and Dissemination Centers, São Paulo Research Foundation), São Paulo, Brazil
| | - Marcelo B. Sztein
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Rosângela Salerno-Gonçalves
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
15
|
Manohar K, Mesfin FM, Liu J, Shelley WC, Brokaw JP, Markel TA. Effect of Oral Chondroitin Sulfate Supplementation on Acute Brain Injury in a Murine Necrotizing Enterocolitis Model. J Am Coll Surg 2024; 238:82-98. [PMID: 37870229 DOI: 10.1097/xcs.0000000000000896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2023]
Abstract
BACKGROUND Necrotizing enterocolitis (NEC) is a devastating condition where inflammatory changes and necrosis in the gut results in activation of brain microglia and subsequent neurodevelopmental impairment. Chondroitin sulfate (CS) is a glycosaminoglycan in human breast milk that is absent in conventional formulas. We hypothesized that oral formula supplementation with CS during a murine model of experimental NEC would not only attenuate intestinal injury, but also brain injury. STUDY DESIGN NEC was induced in mouse pups on postnatal days (PNDs) 5 to 8. Three conditions were studied: (1) breastfed controls, (2) NEC, and (3) NEC+enteral CS (formula+200 mg/kg/d of CS). Pups were euthanized on PND 9 or reunited with dams by the evening of PND 8. Intestinal segments were H&E stained, and immunohistochemistry was performed on brain tissue for Iba-1 to assess for microglial morphology and cortical changes. Neurodevelopmental assays were performed on mice reunited with foster dams on PND 9. Single-cell RNA-sequencing analysis was performed on human intestinal epithelial cells exposed to (1) nothing, (2) hydrogen peroxide (H 2 O 2 ) alone, or (3) H 2 O 2 + CS to look at the differential gene expression between groups. Groups were compared with ANOVA or Kruskal-Wallis tests as appropriate with p < 0.05 considered significant. RESULTS Compared with NEC, mice treated with oral CS showed improved clinical outcomes, decreased intestinal injury, and attenuated microglial activation and deleterious cortical change. Mice with CS performed better on early neurodevelopmental assays when compared with NEC alone. Single-cell analysis of HIEC-6 cells demonstrated that CS treatment down regulated several inflammatory pathways including nuclear factor κB-suggesting an explanation for the improved Th17 intestinal cytokine profile. CONCLUSIONS Oral CS supplementation improved both physiological, clinical, and developmental outcomes. These data suggest that CS is a safe compound for formula supplementation for the prevention of NEC.
Collapse
Affiliation(s)
- Krishna Manohar
- From the Department of Surgery, Indiana University School of Medicine, Indianapolis, IN (Manohar, Mesfin, Liu, Shelley, Brokaw, Markel)
- Riley Hospital for Children at Indiana University Health, Indianapolis, IN (Manohar, Mesfin, Liu, Shelley, Brokaw, Markel)
| | - Fikir M Mesfin
- From the Department of Surgery, Indiana University School of Medicine, Indianapolis, IN (Manohar, Mesfin, Liu, Shelley, Brokaw, Markel)
- Riley Hospital for Children at Indiana University Health, Indianapolis, IN (Manohar, Mesfin, Liu, Shelley, Brokaw, Markel)
| | - Jianyun Liu
- From the Department of Surgery, Indiana University School of Medicine, Indianapolis, IN (Manohar, Mesfin, Liu, Shelley, Brokaw, Markel)
- Riley Hospital for Children at Indiana University Health, Indianapolis, IN (Manohar, Mesfin, Liu, Shelley, Brokaw, Markel)
| | - W Christopher Shelley
- From the Department of Surgery, Indiana University School of Medicine, Indianapolis, IN (Manohar, Mesfin, Liu, Shelley, Brokaw, Markel)
- Riley Hospital for Children at Indiana University Health, Indianapolis, IN (Manohar, Mesfin, Liu, Shelley, Brokaw, Markel)
| | - John P Brokaw
- From the Department of Surgery, Indiana University School of Medicine, Indianapolis, IN (Manohar, Mesfin, Liu, Shelley, Brokaw, Markel)
- Riley Hospital for Children at Indiana University Health, Indianapolis, IN (Manohar, Mesfin, Liu, Shelley, Brokaw, Markel)
| | - Troy A Markel
- From the Department of Surgery, Indiana University School of Medicine, Indianapolis, IN (Manohar, Mesfin, Liu, Shelley, Brokaw, Markel)
- Riley Hospital for Children at Indiana University Health, Indianapolis, IN (Manohar, Mesfin, Liu, Shelley, Brokaw, Markel)
| |
Collapse
|
16
|
Abdel-Gawad DRI, Ibrahim MA, Moawad UK, Kamel S, El-Banna HA, El-Banna AH, Hassan WH, El-Ela FIA. Effectiveness of natural biomaterials in the protection and healing of experimentally induced gastric mucosa Ulcer in rats. Mol Biol Rep 2023; 50:9085-9098. [PMID: 37741810 PMCID: PMC10635934 DOI: 10.1007/s11033-023-08776-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 08/22/2023] [Indexed: 09/25/2023]
Abstract
BACKGROUND A gastric ulcer is a painful lesion of the gastric mucosa that can be debilitating or even fatal. The effectiveness of several plant extracts in the therapy of this illness has been demonstrated in traditional pharmacopoeias. AIM this study was aimed to see if propolis, ginseng in normal or nano form, and amygdalin might help in preventing the ulcerative effects of absolute ethanol. METHODS Gastroprotective properties of pretreatments before ethanol gavage in rats were compared to omeprazole. The ulcer and stomach parameters (ulcerated regions) were measured (mm2), ulcer inhibition percentage, the stomachs were assessed macroscopically with gastric biopsy histological examinations. RESULTS Amygdalin, normal and nano ginseng, nano propolis followed by propolis all showed great efficacy in protecting the cyto-architecture and function of the gastric mucosa. The number of ulcerated sites was greatly reduced, and the percentage of stomach protection was increased. Histopathological examination had confirmed great protective effects of the nanoformulations followed by amygdalin. The protection and healing rate was completed to about 100% in all tested materials while ulcer areas were still partially unhealed in normal propolis and omeprazole. Quantitative assay of the m-RNA levels Enothelin 1(ET-1), leukotriene4 (LT-4), and caspase 3(Cas-3) genes and Histamine were done and revealed significant up-regulations in ethanol group and the maximum protective effect was reported with ginseng nano, moreover the histamine content was significantly decreased with nano- formulated extracts. CONCLUSION Amygdalin and the nanoformulated ginseng and propolis had exhibited a marked protective effect against the ulcerative toxic effects of ethanol.
Collapse
Affiliation(s)
- Doaa R I Abdel-Gawad
- Lecturer of Toxicology and Forensic Medicine, Faculty of Veterinary Medicine, Beni-Suef University, Beni Suef, Egypt
| | - Marwa A Ibrahim
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt.
| | - Usama K Moawad
- Department of Histology and Cytology, Faculty of Veterinary Medicine, Beni-Suef University, Beni-Suef, Egypt
| | - Shaimaa Kamel
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt
| | | | - Ahmed H El-Banna
- Michael Sayegh Faculty of Pharmacy, Aqaba University of Technology, Aqaba, Jordan
| | - Walid Hamdy Hassan
- Mycology and Immunology Department, Faculty of Veterinary Medicine, Beni-Suef University, Beni-Suef, 62511, Egypt
| | - Fatma I Abo El-Ela
- Department of Pharmacology, Faculty of Veterinary Medicine, Beni-Suef University, 62511, Beni-Suef, Egypt
| |
Collapse
|
17
|
Buga AM, Padureanu V, Riza AL, Oancea CN, Albu CV, Nica AD. The Gut-Brain Axis as a Therapeutic Target in Multiple Sclerosis. Cells 2023; 12:1872. [PMID: 37508537 PMCID: PMC10378521 DOI: 10.3390/cells12141872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 07/14/2023] [Accepted: 07/15/2023] [Indexed: 07/30/2023] Open
Abstract
The CNS is very susceptible to oxidative stress; the gut microbiota plays an important role as a trigger of oxidative damage that promotes mitochondrial dysfunction, neuroinflammation, and neurodegeneration. In the current review, we discuss recent findings on oxidative-stress-related inflammation mediated by the gut-brain axis in multiple sclerosis (MS). Growing evidence suggests targeting gut microbiota can be a promising strategy for MS management. Intricate interaction between multiple factors leads to increased intra- and inter-individual heterogeneity, frequently painting a different picture in vivo from that obtained under controlled conditions. Following an evidence-based approach, all proposed interventions should be validated in clinical trials with cohorts large enough to reach significance. Our review summarizes existing clinical trials focused on identifying suitable interventions, the suitable combinations, and appropriate timings to target microbiota-related oxidative stress. Most studies assessed relapsing-remitting MS (RRMS); only a few studies with very limited cohorts were carried out in other MS stages (e.g., secondary progressive MS-SPMS). Future trials must consider an extended time frame, perhaps starting with the perinatal period and lasting until the young adult period, aiming to capture as many complex intersystem interactions as possible.
Collapse
Affiliation(s)
- Ana Maria Buga
- Department of Biochemistry, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania; (A.M.B.); (C.N.O.)
| | - Vlad Padureanu
- Department of Internal Medicine, University of Medicine and Pharmacy of Craiova, 200638 Craiova, Romania;
| | - Anca-Lelia Riza
- Laboratory of Human Genomics, University of Medicine and Pharmacy of Craiova, 200638 Craiova, Romania;
- Regional Center for Medical Genetics Dolj, Emergency County Hospital Craiova, 200638 Craiova, Romania
| | - Carmen Nicoleta Oancea
- Department of Biochemistry, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania; (A.M.B.); (C.N.O.)
| | - Carmen Valeria Albu
- Department of Neurology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Alexandru Dan Nica
- Department of Biochemistry, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania; (A.M.B.); (C.N.O.)
| |
Collapse
|
18
|
Snyder KB, Hunter CJ. Bugs and the barrier: A review of the gut microbiome and intestinal barrier in necrotizing enterocolitis. Semin Pediatr Surg 2023; 32:151310. [PMID: 37290337 DOI: 10.1016/j.sempedsurg.2023.151310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Necrotizing enterocolitis (NEC) is a devastating gastrointestinal disease that affects premature neonates. It frequently results in significant morbidity and mortality for those affected. Years of research into the pathophysiology of NEC have revealed it to be a variable and multifactorial disease. However, there are risk factors associated with NEC including low birth weight, prematurity, intestinal immaturity, alterations in microbial colonization, and history of rapid or formula based enteral feeds (Fig. 1).1-3 An accepted generalization of the pathogenesis of NEC includes a hyperresponsive immune reaction to insults such as ischemia, starting formula feeds, or alterations in the microbiome with pathologic bacterial colonization and translocation. This reaction causes a hyperinflammatory response disrupting the normal intestinal barrier, allowing abnormal bacterial translocation and ultimately sepsis.1,2,4 This review will focus specifically on the interactions with the microbiome and intestinal barrier function in NEC.
Collapse
Affiliation(s)
- K Brooke Snyder
- Division of Pediatric Surgery, Oklahoma Children's Hospital, 1200 Everett Drive, ET NP 2320 Oklahoma City, OK 73104, United States; The University of Oklahoma Health Sciences Center, Department of Surgery, 800 Research Parkway, Suite 449, Oklahoma City, OK 73104, United States
| | - Catherine J Hunter
- Division of Pediatric Surgery, Oklahoma Children's Hospital, 1200 Everett Drive, ET NP 2320 Oklahoma City, OK 73104, United States; The University of Oklahoma Health Sciences Center, Department of Surgery, 800 Research Parkway, Suite 449, Oklahoma City, OK 73104, United States.
| |
Collapse
|
19
|
Golubkova A, Hunter CJ. Development of the Neonatal Intestinal Barrier, Microbiome, and Susceptibility to NEC. Microorganisms 2023; 11:1247. [PMID: 37317221 PMCID: PMC10221463 DOI: 10.3390/microorganisms11051247] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 04/28/2023] [Accepted: 05/04/2023] [Indexed: 06/16/2023] Open
Abstract
The function of the intestinal barrier is partially dependent on host maturity and the colonization patterns of the microbiome to which it is exposed. Premature birth and stressors of neonatal intensive care unit (NICU)-related support (e.g., antibiotics, steroids, etc.) can alter the host internal environment resulting in changes in the intestinal barrier. Pathogenic microbial proliferation and breach of the immature intestinal barrier are proposed to be crucial steps in the development of neonatal diseases such as necrotizing enterocolitis. This article will review the current literature on the intestinal barrier in the neonatal gut, the consequences of microbiome development for this defense system, and how prematurity can influence neonatal susceptibility to gastrointestinal infection.
Collapse
Affiliation(s)
| | - Catherine J. Hunter
- Division of Pediatric Surgery, Department of Surgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
20
|
Anderson J, Bender G, Minh Thang C, Quang Thanh L, Thi Trang Dai V, Van Thanh P, Thi Hong Nhu B, Ngoc Xuan Trang D, Thi Phuong Trinh P, Vu Thuong N, Trong Toan N, Mulholland K, Pellicci DG, Anh Ha Do L, Licciardi PV. TLR Responses in Preterm and Term Infant Cord Blood Mononuclear Cells. Pathogens 2023; 12:pathogens12040596. [PMID: 37111482 PMCID: PMC10145848 DOI: 10.3390/pathogens12040596] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/12/2023] [Accepted: 04/13/2023] [Indexed: 04/29/2023] Open
Abstract
Preterm infants are more susceptible to severe bacterial and viral infectious diseases than their full-term counterparts. A major contributor to this increased susceptibility may be due to differences in their ability to respond to pathogens. While studies have demonstrated altered bacterial Toll-like receptor (TLR) responses, there is limited data on viral TLR responses in preterm infants. In this study, cord blood mononuclear cells (CBMCs) from 10 moderately preterm (30.4-34.1 wGA), 10 term (37-39.5 wGA) infants, and 5 adults were stimulated with TLR2 (lipoteichoic acid), TLR3 (poly I:C), TLR4 (lipopolysaccharide), TLR7/8 (R848), and TLR9 (CpG-ODN 2216) agonists. Following stimulation, the cellular response was measured by intracellular flow cytometry to detect cell-specific NF-κB (as a marker of the inflammatory response), and multiplex assays were used to measure the cytokine response. This study found that preterm and term infants exhibit very similar baseline TLR expression. In response to both bacterial and viral TLR agonists comparing cell-specific NF-κB activation, preterm infants exhibited increased monocyte activation following LTA stimulation; however, no other differences were observed. Similarly, no difference in cytokine response was observed following stimulation with TLRs. However, a stronger correlation between NF-κB activation and cytokine responses was observed in term infants following poly I:C and R848 stimulation compared to preterm infants. In contrast, despite similar TLR expression, adults produced higher levels of IFN-α following R848 stimulation compared to preterm and term infants. These findings suggest preterm and term infants have a similar capacity to respond to both bacterial and viral TLR agonists. As preterm infants are more likely to develop severe infections, further research is required to determine the immunological factors that may be driving this and develop better interventions for this highly vulnerable group.
Collapse
Affiliation(s)
- Jeremy Anderson
- Murdoch Children's Research Institute, Melbourne, VIC 3052, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Georgia Bender
- Murdoch Children's Research Institute, Melbourne, VIC 3052, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Cao Minh Thang
- Pasteur Institute of Ho Chi Minh City, Ho Chi Minh City 72408, Vietnam
| | | | - Vo Thi Trang Dai
- Pasteur Institute of Ho Chi Minh City, Ho Chi Minh City 72408, Vietnam
| | - Phan Van Thanh
- Pasteur Institute of Ho Chi Minh City, Ho Chi Minh City 72408, Vietnam
| | | | | | | | | | | | - Kim Mulholland
- Murdoch Children's Research Institute, Melbourne, VIC 3052, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, VIC 3010, Australia
- Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, London WC1E 7HT, UK
| | - Daniel G Pellicci
- Murdoch Children's Research Institute, Melbourne, VIC 3052, Australia
- Department of Microbiology and Immunology, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Lien Anh Ha Do
- Murdoch Children's Research Institute, Melbourne, VIC 3052, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Paul V Licciardi
- Murdoch Children's Research Institute, Melbourne, VIC 3052, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, VIC 3010, Australia
| |
Collapse
|
21
|
Zhang X, Luo Y, Gu R, Jiang Z. Astaxanthin Alleviates Inflammatory Response in Neonatal Necrotizing Enterocolitis Rats by Regulating NOD2/TLR4 Pathway. Gastroenterol Res Pract 2023; 2023:6078308. [PMID: 37021078 PMCID: PMC10070044 DOI: 10.1155/2023/6078308] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 02/09/2023] [Accepted: 03/11/2023] [Indexed: 04/07/2023] Open
Abstract
Background Necrotizing enterocolitis (NEC) is often associated with exaggerated activation of inflammatory response. Astaxanthin has been shown in studies to have a positive and advantageous effect on anti-inflammatory response. Hence, it is of great significance to study the protective effect of astaxanthin in NEC disease and its molecular mechanism. Objective The present study was to investigate whether astaxanthin attenuates NEC rats and to explore its potential mechanism. Material and Methods. Hematoxylin-eosin staining was used to observe the pathological change of the intestinal tissue in NEC rats. Subsequently, we determined the anti-oxidative stress, anti-apoptosis, and anti-inflammation in astaxanthin with enzyme-linked immunosorbent assay kits, TUNEL staining, western blot, and immunohistochemistry assay. Furthermore, we added nucleotide-binding oligomerization domain 2 (NOD2) inhibitor to certify the molecular pathway of the astaxanthin in NEC rats. Results Astaxanthin improved the pathological changes of the intestinal tissues. It restrained inflammation, oxidative stress, and protected cells from apoptosis in the intestinal tissue and serum of the NEC rats. Moreover, astaxanthin enhanced NOD2, whereas it suppressed toll-like receptor 4 (TLR4), nuclear factor-κB (NF-κB) pathway-related proteins. Apart from that, the NOD2 inhibitor offset the protective effect of the astaxanthin towards the NEC rats. Conclusion The present study indicated that astaxanthin alleviated oxidative stress, inflammatory response, and apoptosis in NEC rats by enhancing NOD2 and inhibiting TLR4 pathway.
Collapse
Affiliation(s)
- Xuandong Zhang
- Department of Neonatology, Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Yujia Luo
- Department of Neonatology, Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Rui Gu
- Department of Neonatology, Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Zhou Jiang
- Department of Neonatology, Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| |
Collapse
|
22
|
Therapeutic Potential of Gut Microbiota and Its Metabolite Short-Chain Fatty Acids in Neonatal Necrotizing Enterocolitis. Life (Basel) 2023; 13:life13020561. [PMID: 36836917 PMCID: PMC9959300 DOI: 10.3390/life13020561] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/31/2023] [Accepted: 02/15/2023] [Indexed: 02/19/2023] Open
Abstract
Short chain fatty acids (SCFAs), the principle end-products produced by the anaerobic gut microbial fermentation of complex carbohydrates (CHO) in the colon perform beneficial roles in metabolic health. Butyrate, acetate and propionate are the main SCFA metabolites, which maintain gut homeostasis and host immune responses, enhance gut barrier integrity and reduce gut inflammation via a range of epigenetic modifications in DNA/histone methylation underlying these effects. The infant gut microbiota composition is characterized by higher abundances of SCFA-producing bacteria. A large number of in vitro/vivo studies have demonstrated the therapeutic implications of SCFA-producing bacteria in infant inflammatory diseases, such as obesity and asthma, but the application of gut microbiota and its metabolite SCFAs to necrotizing enterocolitis (NEC), an acute inflammatory necrosis of the distal small intestine/colon affecting premature newborns, is scarce. Indeed, the beneficial health effects attributed to SCFAs and SCFA-producing bacteria in neonatal NEC are still to be understood. Thus, this literature review aims to summarize the available evidence on the therapeutic potential of gut microbiota and its metabolite SCFAs in neonatal NEC using the PubMed/MEDLINE database.
Collapse
|
23
|
Schiller EA, Cohen K, Lin X, El-Khawam R, Hanna N. Extracellular Vesicle-microRNAs as Diagnostic Biomarkers in Preterm Neonates. Int J Mol Sci 2023; 24:2622. [PMID: 36768944 PMCID: PMC9916767 DOI: 10.3390/ijms24032622] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 01/24/2023] [Accepted: 01/25/2023] [Indexed: 02/03/2023] Open
Abstract
Neonates born prematurely (<37 weeks of gestation) are at a significantly increased risk of developing inflammatory conditions associated with high mortality rates, including necrotizing enterocolitis, bronchopulmonary dysplasia, and hypoxic-ischemic brain damage. Recently, research has focused on characterizing the content of extracellular vesicles (EVs), particularly microRNAs (miRNAs), for diagnostic use. Here, we describe the most recent work on EVs-miRNAs biomarkers discovery for conditions that commonly affect premature neonates.
Collapse
Affiliation(s)
- Emily A. Schiller
- Department of Foundational Medicine, New York University Long Island School of Medicine, Mineola, NY 11501, USA
| | - Koral Cohen
- Department of Foundational Medicine, New York University Long Island School of Medicine, Mineola, NY 11501, USA
| | - Xinhua Lin
- Department of Foundational Medicine, New York University Long Island School of Medicine, Mineola, NY 11501, USA
| | - Rania El-Khawam
- Department of Pediatrics, Division of Neonatology, New York University Langone Long Island Hospital, Mineola, NY 11501, USA
| | - Nazeeh Hanna
- Department of Foundational Medicine, New York University Long Island School of Medicine, Mineola, NY 11501, USA
- Department of Pediatrics, Division of Neonatology, New York University Langone Long Island Hospital, Mineola, NY 11501, USA
| |
Collapse
|
24
|
Kaplina A, Kononova S, Zaikova E, Pervunina T, Petrova N, Sitkin S. Necrotizing Enterocolitis: The Role of Hypoxia, Gut Microbiome, and Microbial Metabolites. Int J Mol Sci 2023; 24:2471. [PMID: 36768793 PMCID: PMC9917134 DOI: 10.3390/ijms24032471] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 01/15/2023] [Accepted: 01/17/2023] [Indexed: 02/01/2023] Open
Abstract
Necrotizing enterocolitis (NEC) is a life-threatening disease that predominantly affects very low birth weight preterm infants. Development of NEC in preterm infants is accompanied by high mortality. Surgical treatment of NEC can be complicated by short bowel syndrome, intestinal failure, parenteral nutrition-associated liver disease, and neurodevelopmental delay. Issues surrounding pathogenesis, prevention, and treatment of NEC remain unclear. This review summarizes data on prenatal risk factors for NEC, the role of pre-eclampsia, and intrauterine growth retardation in the pathogenesis of NEC. The role of hypoxia in NEC is discussed. Recent data on the role of the intestinal microbiome in the development of NEC, and features of the metabolome that can serve as potential biomarkers, are presented. The Pseudomonadota phylum is known to be associated with NEC in preterm neonates, and the role of other bacteria and their metabolites in NEC pathogenesis is also discussed. The most promising approaches for preventing and treating NEC are summarized.
Collapse
Affiliation(s)
- Aleksandra Kaplina
- Research Laboratory for Physiology and Diseases of Newborns, Almazov National Medical Research Centre, St. Petersburg 197341, Russia
| | - Svetlana Kononova
- Group of Protein Synthesis Regulation, Institute of Protein Research, Russian Academy of Sciences, Pushchino 142290, Russia
| | - Ekaterina Zaikova
- Research Laboratory of Autoimmune and Autoinflammatory Diseases, Almazov National Medical Research Centre, St. Petersburg 197341, Russia
| | - Tatiana Pervunina
- Institute of Perinatology and Pediatrics, Almazov National Medical Research Centre, St. Petersburg 197341, Russia
| | - Natalia Petrova
- Research Laboratory for Physiology and Diseases of Newborns, Almazov National Medical Research Centre, St. Petersburg 197341, Russia
| | - Stanislav Sitkin
- Epigenetics and Metagenomics Group, Institute of Perinatology and Pediatrics, Almazov National Medical Research Centre, St. Petersburg 197341, Russia
- Department of Internal Diseases, Gastroenterology and Dietetics, North-Western State Medical University Named after I.I. Mechnikov, St. Petersburg 191015, Russia
| |
Collapse
|
25
|
Akduman H, Tayman C, Korkmaz V, Akduman F, Fettah ND, Gürsoy BK, Turkmenoglu TT, Çağlayan M. Astaxanthin Reduces the Severity of Intestinal Damage in a Neonatal Rat Model of Necrotizing Enterocolitis. Am J Perinatol 2022; 39:1820-1827. [PMID: 33853144 DOI: 10.1055/s-0041-1727156] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
OBJECTIVE This study aimed to ascertain the effects of astaxanthin (ASX) in an experimental necrotizing enterocolitis (NEC) model using rat pups. STUDY DESIGN Forty-two pups born from five Wistar albino rats were randomly divided into three groups as the control group, NEC + placebo (saline), and NEC + ASX. Pups in the NEC + ASX group were given 100 mg/kg/day oral ASX from day 1 to day 4 of the study. Saline of 2 mL/kg was given to the NEC + placebo group. Histopathological, immunohistochemical (caspase-3), and biochemical evaluations including the total antioxidant status (TAS), total oxidant status (TOS), superoxide dismutase (SOD), glutathione (GSH), lipid hydroperoxide (LPO), 8-hydroxydeoxyguanosine (8-OHdG), advanced oxidation protein products (AOPP), myeloperoxidase (MPO), tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), and nuclear factor erythroid 2-related factor 2 (Nfr-2) activities were all performed. RESULTS A better survival rate and weight gain were demonstrated in the NEC + ASX group (p < 0.05). In the histopathological evaluation, the severity of intestinal damage was significantly reduced in the NEC + ASX group, as well as decreased apoptosis (enzyme-linked immunosorbent assay [ELISA] for caspase-3; p = 0.001). The biochemical analyses of intestinal tissue TOS, oxidative stress index (OSI; TOS/TAS), IL-1β, LPO, 8-OHdG, AOPP, caspase-3 (p < 0.001 for all), and TNF-α and MPO (p = 0.001 for both parameters) levels were lower in the NEC + ASX group than in the NEC + placebo group. Nrf-2, TAS, GSH, and SOD levels were higher in the NEC + ASX group than in the NEC + placebo group (p = 0.001, 0.001, <0.001, and 0.01, respectively). CONCLUSION ASX treatment has been shown to effectively reduce the severity of intestinal damage in NEC due to its antioxidant, anti-inflammatory, and antiapoptotic properties. KEY POINTS · NEC causes extremely high morbidity and mortality, as well as many complications.. · We investigated the effectiveness of ASX in the experimental NEC model created in rat pups.. · First study examining the effect of ASX on the experimental NEC rat model..
Collapse
Affiliation(s)
- Hasan Akduman
- Division of Neonatology, Department of Pediatrics, SBU Ankara Dr. Sami Ulus Maternity Child Health and Diseases Training and Research Hospital, Ankara, Turkey
| | - Cuneyt Tayman
- Department of Neonatology, Ankara City Hospital, Cankaya, Ankara, Turkey
| | - Veli Korkmaz
- Department of Pediatrics, Lokman Hekim University, Ankara, Turkey
| | - Filiz Akduman
- Department of Pediatrics, Beypazarı State Hospital, Ankara, Turkey
| | - Nurdan D Fettah
- Department of Neonatology, SBU Ankara Dr. Sami Ulus Maternity Child Health and Diseases Training and Research Hospital, Ankara, Turkey
| | - Başak K Gürsoy
- Department of Neonatology, SBU Ankara Dr. Sami Ulus Maternity Child Health and Diseases Training and Research Hospital, Ankara, Turkey
| | - Tugba T Turkmenoglu
- Department of Pathology, Ankara Diskapi Yildirim Beyzat Training and Research Hospital, Ankara, Turkey
| | - Murat Çağlayan
- Department of Medicinal Biochemistry, University of Health Sciences Gülhane Health Sciences Institute, Ankara, Turkey
| |
Collapse
|
26
|
Bermudez-Lekerika P, Crump KB, Tseranidou S, Nüesch A, Kanelis E, Alminnawi A, Baumgartner L, Muñoz-Moya E, Compte R, Gualdi F, Alexopoulos LG, Geris L, Wuertz-Kozak K, Le Maitre CL, Noailly J, Gantenbein B. Immuno-Modulatory Effects of Intervertebral Disc Cells. Front Cell Dev Biol 2022; 10:924692. [PMID: 35846355 PMCID: PMC9277224 DOI: 10.3389/fcell.2022.924692] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 05/20/2022] [Indexed: 11/29/2022] Open
Abstract
Low back pain is a highly prevalent, chronic, and costly medical condition predominantly triggered by intervertebral disc degeneration (IDD). IDD is often caused by structural and biochemical changes in intervertebral discs (IVD) that prompt a pathologic shift from an anabolic to catabolic state, affecting extracellular matrix (ECM) production, enzyme generation, cytokine and chemokine production, neurotrophic and angiogenic factor production. The IVD is an immune-privileged organ. However, during degeneration immune cells and inflammatory factors can infiltrate through defects in the cartilage endplate and annulus fibrosus fissures, further accelerating the catabolic environment. Remarkably, though, catabolic ECM disruption also occurs in the absence of immune cell infiltration, largely due to native disc cell production of catabolic enzymes and cytokines. An unbalanced metabolism could be induced by many different factors, including a harsh microenvironment, biomechanical cues, genetics, and infection. The complex, multifactorial nature of IDD brings the challenge of identifying key factors which initiate the degenerative cascade, eventually leading to back pain. These factors are often investigated through methods including animal models, 3D cell culture, bioreactors, and computational models. However, the crosstalk between the IVD, immune system, and shifted metabolism is frequently misconstrued, often with the assumption that the presence of cytokines and chemokines is synonymous to inflammation or an immune response, which is not true for the intact disc. Therefore, this review will tackle immunomodulatory and IVD cell roles in IDD, clarifying the differences between cellular involvements and implications for therapeutic development and assessing models used to explore inflammatory or catabolic IVD environments.
Collapse
Affiliation(s)
- Paola Bermudez-Lekerika
- Tissue Engineering for Orthopaedics and Mechanobiology, Bone and Joint Program, Department for BioMedical Research (DBMR), Faculty of Medicine, University of Bern, Bern, Switzerland.,Department of Orthopaedic Surgery and Traumatology, Inselspital, Bern University Hospital, Medical Faculty, University of Bern, Bern, Switzerland
| | - Katherine B Crump
- Tissue Engineering for Orthopaedics and Mechanobiology, Bone and Joint Program, Department for BioMedical Research (DBMR), Faculty of Medicine, University of Bern, Bern, Switzerland.,Department of Orthopaedic Surgery and Traumatology, Inselspital, Bern University Hospital, Medical Faculty, University of Bern, Bern, Switzerland
| | | | - Andrea Nüesch
- Biomolecular Sciences Research Centre, Sheffield Hallam University, Sheffield, United Kingdom
| | - Exarchos Kanelis
- ProtATonce Ltd., Athens, Greece.,School of Mechanical Engineering, National Technical University of Athens, Zografou, Greece
| | - Ahmad Alminnawi
- GIGA In Silico Medicine, University of Liège, Liège, Belgium.,Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium
| | | | | | - Roger Compte
- Twin Research and Genetic Epidemiology, St Thomas' Hospital, King's College London, London, United Kingdom
| | - Francesco Gualdi
- Institut Hospital Del Mar D'Investigacions Mèdiques (IMIM), Barcelona, Spain
| | - Leonidas G Alexopoulos
- ProtATonce Ltd., Athens, Greece.,School of Mechanical Engineering, National Technical University of Athens, Zografou, Greece
| | - Liesbet Geris
- GIGA In Silico Medicine, University of Liège, Liège, Belgium.,Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium.,Biomechanics Research Unit, KU Leuven, Leuven, Belgium
| | - Karin Wuertz-Kozak
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY, United States.,Spine Center, Schön Klinik München Harlaching Academic Teaching Hospital and Spine Research Institute of the Paracelsus Private Medical University Salzburg (Austria), Munich, Germany
| | - Christine L Le Maitre
- Biomolecular Sciences Research Centre, Sheffield Hallam University, Sheffield, United Kingdom
| | | | - Benjamin Gantenbein
- Tissue Engineering for Orthopaedics and Mechanobiology, Bone and Joint Program, Department for BioMedical Research (DBMR), Faculty of Medicine, University of Bern, Bern, Switzerland.,Department of Orthopaedic Surgery and Traumatology, Inselspital, Bern University Hospital, Medical Faculty, University of Bern, Bern, Switzerland
| |
Collapse
|
27
|
Zhang X, Zhang X, Luo H, Shu R, Guo L, Zhou J, Tan B, Guo X, Wang Y, Tian Y. Platelet-To-Lymphocyte and Neutrophil-To-Lymphocyte Ratios Predict Intestinal Injury in Male Heroin Addicts. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:2195330. [PMID: 35880090 PMCID: PMC9308521 DOI: 10.1155/2022/2195330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 06/24/2022] [Accepted: 06/30/2022] [Indexed: 11/24/2022]
Abstract
Objective To explore the potential link between gut damage and proinflammatory cytokines in heroin-dependent patients. Methods We retrospectively analyzed and compared partial blood counts and biomarkers of intestinal injury and their potential correlations in 38 male heroin abuse patients and 29 healthy male participants. In addition, we compared and assessed proinflammatory cytokines and immune cells in 10 heroin abuse patients and 10 healthy participants. Results Neutrophil counts, platelets/lymphocytes (PLR), neutrophils/lymphocytes (NLR), gut injury biomarkers, and proinflammatory cytokines, CD19+B in patients compared with healthy subjects' cells increased significantly. The number of lymphocytes, CD3 CD4 T cells, and CD3 CD8 T cells decreased in patients compared to healthy individuals. When distinguishing between heroin addicts and healthy people, ROC/AUC analysis showed that a cutoff of 142.42 for PLR and 2.18 for NLR yielded a sensitivity of 65% and 85% and a specificity of 96.5% and 89.7%, respectively (p = 0.001, p < 0.001). For predicting intestinal injury, ROC/AUC analysis showed that a cutoff of 135.7 for PLR and 0.15 for NLR yielded a sensitivity of 52% and 60% and a specificity of 82% and 86.4%, respectively (p = 0.003, p = 0.009). Male heroin addicts are subject to intestinal injury and present with increased proinflammatory cytokine levels. Conclusion NLR and PLR are possible indirect biomarkers for heroin dependence based on intestinal injury.
Collapse
Affiliation(s)
- Xinfeng Zhang
- Department of Gastrointestinal and Hernia Surgery, The First Affiliated Hospital of Kunming Medical University, China Kunming, Yunnan 650032
| | - Xiaoli Zhang
- Department of Gastrointestinal and Hernia Surgery, The First Affiliated Hospital of Kunming Medical University, China Kunming, Yunnan 650032
| | - Huayou Luo
- Department of Gastrointestinal and Hernia Surgery, The First Affiliated Hospital of Kunming Medical University, China Kunming, Yunnan 650032
| | - Ruo Shu
- Department of Gastrointestinal and Hernia Surgery, The First Affiliated Hospital of Kunming Medical University, China Kunming, Yunnan 650032
| | - Li Guo
- Department of Gastrointestinal and Hernia Surgery, The First Affiliated Hospital of Kunming Medical University, China Kunming, Yunnan 650032
| | - Jinghong Zhou
- Department of Gastrointestinal and Hernia Surgery, The First Affiliated Hospital of Kunming Medical University, China Kunming, Yunnan 650032
| | - Bowen Tan
- Department of Gastrointestinal and Hernia Surgery, The First Affiliated Hospital of Kunming Medical University, China Kunming, Yunnan 650032
| | - Xiao Guo
- Department of Gastrointestinal and Hernia Surgery, The First Affiliated Hospital of Kunming Medical University, China Kunming, Yunnan 650032
| | - Yuhan Wang
- Department of Gastrointestinal and Hernia Surgery, The First Affiliated Hospital of Kunming Medical University, China Kunming, Yunnan 650032
| | - Yan Tian
- Department of Gastrointestinal and Hernia Surgery, The First Affiliated Hospital of Kunming Medical University, China Kunming, Yunnan 650032
| |
Collapse
|
28
|
Neonatal intermittent hypoxia, fish oil, and/or antioxidant supplementation on gut microbiota in neonatal rats. Pediatr Res 2022; 92:109-117. [PMID: 34455420 PMCID: PMC8882692 DOI: 10.1038/s41390-021-01707-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 06/22/2021] [Accepted: 08/09/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND Preterm infants frequently experience intermittent hypoxia (IH) episodes, rendering them susceptible to oxidative stress and gut dysbiosis. We tested the hypothesis that early supplementation with antioxidants and/or fish oil promotes gut biodiversity and mitigates IH-induced gut injury. METHODS Newborn rats were exposed to neonatal IH from birth (P0) to P14 during which they received daily oral supplementation with: (1) coenzyme Q10 (CoQ10) in olive oil, (2) fish oil, (3) glutathione nanoparticles (nGSH), (4) CoQ10 + fish oil, or (5) olive oil (placebo control). Pups were placed in room air (RA) from P14 to P21 with no further treatment. RA controls were similarly treated. Stool samples were assessed for microbiota and terminal ileum for histopathology and morphometry, total antioxidant capacity, lipid peroxidation, and biomarkers of gut injury. RESULTS Neonatal IH induced histopathologic changes consistent with necrotizing enterocolitis, which were associated with increased lipid peroxidation, toll-like receptor, transforming growth factor, and nuclear factor kappa B. Combination of CoQ10 + fish oil and nGSH were most effective for preserving gut integrity, reducing biomarkers of gut injury, and increasing commensal organisms. CONCLUSIONS Combination of antioxidants and fish oil may confer synergistic benefits to mitigate IH-induced injury in the terminal ileum. IMPACT Antioxidant and fish oil (PUFA) co-treatment was most beneficial for reducing neonatal IH-induced gut injury. The synergistic effects of antioxidant and fish oil is likely due to prevention of IH-induced ROS attack on lipids, thus preserving and augmenting its therapeutic benefits. Combination treatment was also effective for increasing the abundance of the non-pathogenic Firmicutes phylum, which is associated with a healthy gastrointestinal system of the newborn. Extremely low gestational age neonates who are at high risk for frequent, repetitive neonatal IH and oxidative stress-induced diseases may benefit from this combination therapy.
Collapse
|
29
|
Adams JM, Valentine CJ, Karns RA, Rogers LK, Murase M, Fowler GN, Nommsen-Rivers LA. DHA Supplementation Attenuates Inflammation-Associated Gene Expression in the Mammary Gland of Lactating Mothers Who Deliver Preterm. J Nutr 2022; 152:1404-1414. [PMID: 35199834 PMCID: PMC9178958 DOI: 10.1093/jn/nxac043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 01/25/2022] [Accepted: 02/21/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND In a randomized trial of DHA supplementation to lactating mothers who delivered preterm, there were significant increases in DHA status in the mother and her infant. OBJECTIVES Our objective here was to characterize the mammary gland transcriptomes from the above study. We hypothesized that proinflammatory gene expression would be attenuated in the increased DHA group compared with the standard DHA group. METHODS In the original trial, mothers delivering at <29 wk gestation at the University of Cincinnati Medical Center and intending to express their milk were randomly assigned to supplementation with 200 mg/d DHA (standard group: STD) or 1000 mg/d DHA (experimental group: EXP) within 7 d of delivery. Here, we conducted RNA-seq transcriptome analysis of n = 5 EXP and n = 4 STD extracellular mammary mRNA samples extracted from the fat layer of milk samples obtained 4 wk postenrollment. Transcripts were assessed for differential expression (false discovery rate adjusted P value <0.05) and clustering between EXP compared with STD groups. Ontological analysis of all differentially expressed genes (DEGs) was performed with Toppcluster. RESULTS There were 409 DEGs. We observed 5 main groups of biological processes that were upregulated, including those associated with improved immune regulation and management of oxidative stress; and 3 main groups of biological processes that were downregulated, including 1 associated with immune dysregulation. For example, we observed upregulation of inflammation-inhibiting genes including NFKB inhibitor alpha (NFKBIA; fold-change (FC), adjusted P value: FC = 1.70, P = 0.007) and interleukin-18 binding protein (IL18BP: FC = 2.2, adjusted P = 0.02); and downregulation of proinflammatory genes including interleukin 7 receptor (IL7R: FC = -1.9, adjusted P = 0.02) and interleukin 1 receptor like 1 (IL1RL1: FC = -13.0, adjusted P = 0.02). CONCLUSIONS Increased DHA supplementation during lactation can modulate the expression of inflammation-related genes within the mammary gland. This might translate to milk composition with a more optimal inflammasome profile. Future research with a larger clinical trial and greater interrogation of clinical outcomes is warranted.
Collapse
Affiliation(s)
- Joselyn M Adams
- Department of Rehabilitation, Exercise, and Nutritional Sciences, University of Cincinnati College of Allied Health Sciences, Cincinnati, OH, USA
| | - Christina J Valentine
- Department of Neonatology, Banner University Medical Center, The University of Arizona, Tucson, AZ, USA
| | - Rebekah A Karns
- Division of Biostatistics and Epidemiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Lynette K Rogers
- Center for Perinatal Research, Nationwide Children's Hospital, Columbus, OH, USA
| | - Masahiko Murase
- Department of Neonatology, Showa University Hospital, Shinagawa City, Tokyo, Japan
| | - Grace N Fowler
- Department of Obstetrics and Gynecology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Laurie A Nommsen-Rivers
- Department of Rehabilitation, Exercise, and Nutritional Sciences, University of Cincinnati College of Allied Health Sciences, Cincinnati, OH, USA
| |
Collapse
|
30
|
Li D, Yao X, Yue J, Fang Y, Cao G, Midgley AC, Nishinari K, Yang Y. Advances in Bioactivity of MicroRNAs of Plant-Derived Exosome-Like Nanoparticles and Milk-Derived Extracellular Vesicles. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:6285-6299. [PMID: 35583385 DOI: 10.1021/acs.jafc.2c00631] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
MicroRNA (miRNA) is a class of small noncoding RNA involved in physiological and pathological processes via the regulation of gene expression. Naked miRNAs are unstable and liable to degradation by RNases. Exosome-like nanoparticles (ELNs) secreted by plants and extracellular vesicles (EVs) found in milk are abundant in miRNAs, which can be carried by ELNs and EVs to target cells to exert their bioactivities. In this review, we describe the current understanding of miRNAs in plant ELNs and milk EVs, summarize their important roles in regulation of inflammation, intestinal barrier, tumors, and infantile immunological functions, and also discuss the adverse effect of EV miRNAs on human health. Additionally, we prospect recent challenges centered around ELN and EV miRNAs for interventional applications and provide insights of grain-derived ELNs and miRNAs interventional use in human health. Overall, plant ELNs and milk EVs can transfer miRNAs to mitigate the pathological status of recipient cells by mediating the expression of target genes but may also exert some side effects. More studies are required to elucidate the in-depth understanding of potential interventional effects of ELN and EV miRNAs on human health.
Collapse
Affiliation(s)
- Dan Li
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi'an, Shaanxi 710021, P. R. China
| | - Xiaolin Yao
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi'an, Shaanxi 710021, P. R. China
| | - Jianxiong Yue
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi'an, Shaanxi 710021, P. R. China
| | - Yapeng Fang
- Department of Food Science and Engineering, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Guifang Cao
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi'an, Shaanxi 710021, P. R. China
| | - Adam C Midgley
- Key Laboratory of Bioactive Materials (MoE), College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Katsuyoshi Nishinari
- Glyn O. Phillips Hydrocolloid Research Centre, School of Bioengineering and Food Science, Hubei University of Technology, Wuhan 430068, China
| | - Yongli Yang
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi'an, Shaanxi 710021, P. R. China
| |
Collapse
|
31
|
Zhang Y, Wang O, Mi H, Yi J, Cai S. Rhus chinensis Mill. fruits prevent necrotizing enterocolitis in rat pups via regulating the expressions of key proteins involved in multiple signaling pathways. JOURNAL OF ETHNOPHARMACOLOGY 2022; 290:115103. [PMID: 35157955 DOI: 10.1016/j.jep.2022.115103] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 02/03/2022] [Accepted: 02/09/2022] [Indexed: 06/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Based on ancient records and previous studies, many parts of Rhus chinensis Mill., including the fruits, have good preventive and therapeutic effects on inflammation, malaria, diarrhea, and gastrointestinal diseases. Rhus plants and Galla chinensis produced from R. chinensis leaves can also prevent or cure intestinal diseases. However, the preventive effect and molecular mechanisms of R. chinensis fruits on necrotizing enterocolitis (NEC) have not been comprehensively studied. AIM OF THE STUDY This article aims to estimate the effect of the 80% ethanol extract of R. chinensis fruits (RM) on alleviating NEC in rat pups and illustrate the potential molecular mechanisms. MATERIALS AND METHODS Rat pups were subjected to formula feeding, intermittent hypoxic, and cold stresses to establish the NEC model. The preventive effects of RM on NEC were evaluated through survival rate; clinical sickness index; macroscopic conditions; histopathology; and expression levels of inflammatory markers (i.e., tumor necrosis factor-α [TNF-α], interleukin-6 [IL-6]), oxidative stress indicators (i.e., total antioxidant status [TAS], total oxidant status [TOS], superoxide dismutase [SOD], glutathione peroxidase [GSH-Px], myeloperoxidase [MPO], malondialdehyde [MDA]), and tight junction proteins (i.e., Zonula Occludens 1 [ZO-1], Occludin). Moreover, the expression levels of several key proteins involved in oxidative stress (i.e., nuclear factor erythroid 2-related factor 2 [Nrf2], NAD(P)H-quinone oxidoreductase-1 [NQO1]), inflammation (i.e., Toll-like receptor 4 [TLR4], phosphorylated-nuclear factor kappa-B [p-NF-κB], inducible nitric oxide synthase [iNOS]), and apoptosis (i.e., cleaved cysteinyl aspartate specific proteinase-3 [cleaved Caspase-3], Bcl-2-associated X [Bax], B-cell lymphoma-2 [Bcl-2]) in intestinal tissues were analyzed to clarify the molecular mechanisms. RESULTS The extract particularly high doses (400 mg RM/kg body weight) could remarkably reduce the mortality and clinical sickness score and improve the macroscopic condition and histopathological injury of the intestine in NEC pups. After RM administration, the levels of TOS, TNF-α, IL-6, MPO, and MDA in the bowel tissue decreased, whereas the levels of TAS, SOD, and GSH-Px were significantly enhanced. The expression levels of ZO-1 and Occludin proteins were dramatically augmented in RM-treated groups to maintain intestinal barrier integrity. Further analyses revealed that RM might prevent NEC pups by improving some pivotal proteins involved in oxidative stress, inflammation, and apoptosis of enterocytes, namely, by down-regulating the levels of TLR4, p-NF-κB, iNOS, cleaved Caspase-3, and Bax and up-regulating the levels of Bcl-2, NQO1, and Nrf2. CONCLUSIONS The RM prevented the intestinal inflammation and damage caused by NEC by regulating the expression of several pivotal proteins involved in oxidative stress, inflammation, and apoptosis. This study might provide a scientific basis for R. chinensis fruits as a traditional herbal medicine to prevent and/or alleviate NEC.
Collapse
Affiliation(s)
- Yi Zhang
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming, 650500, Yunnan Province, People's Republic of China
| | - Ou Wang
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing, 100050, People's Republic of China
| | - Hongying Mi
- The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650032, Yunnan Province, People's Republic of China
| | - Junjie Yi
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming, 650500, Yunnan Province, People's Republic of China
| | - Shengbao Cai
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming, 650500, Yunnan Province, People's Republic of China.
| |
Collapse
|
32
|
Melatonin Alleviates Neonatal Necrotizing Enterocolitis by Repressing the Activation of the NLRP3 Inflammasome. Gastroenterol Res Pract 2022; 2022:6920577. [PMID: 35340691 PMCID: PMC8947927 DOI: 10.1155/2022/6920577] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 02/22/2022] [Accepted: 02/26/2022] [Indexed: 02/08/2023] Open
Abstract
Objective Necrotizing enterocolitis (NEC) is one of the commonest gastrointestinal critical diseases in newborns. Several researches have proven the efficacy of melatonin (MEL) on NEC, but the latent mechanisms were ambiguous. We designed the current research to evaluate the function and mechanism of MEL on NEC in a neonatal mouse model. Methods The newborn mice were subjected to formula milk containing LPS and hypoxia to establish a NEC model and also intraperitoneally injected with MEL. During the experiment, all mice were closely monitored and weighed. The effect of MEL on the histopathological injury of the terminal ileum tissues, inflammation, and oxidative stress of serum in NEC mice was examined by hematoxylin-eosin (H&E) staining and ELISA. The effect of MEL on the NOD-like receptor family pyrin domain containing 3 (NLRP3) inflammasome was assessed via quantitative real-time PCR and Western blot. Results MEL intensified the survival rate and body weight in NEC mice. The H&E staining illustrated that MEL improved the histopathological injury in NEC mice. Moreover, MEL repressed the IL-1β, TNF-α, and MDA levels of serum and enhanced the SOD and GSH-Px levels of serum in NEC mice. We also discovered that MEL attenuated the mRNA and protein levels of NLRP3, Toll-like Receptor 4 (TLR4), NF-κB, and caspase-1 of the terminal ileum tissues in NEC mice. Conclusion Our research illuminated that MEL attenuated the severity of NEC via weakening the activation of the NLRP3 inflammasome.
Collapse
|
33
|
Advanced drug delivery systems containing herbal components for wound healing. Int J Pharm 2022; 617:121617. [PMID: 35218900 DOI: 10.1016/j.ijpharm.2022.121617] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Revised: 02/12/2022] [Accepted: 02/21/2022] [Indexed: 12/18/2022]
Abstract
Management of chronic wound has an immense impact on social and economic conditions in the world. Healthcare costs, aging population, physical trauma, and comorbidities of diabetes and obesity seem to be the major factors of this increasing incidence of chronic wounds. Conditions of chronic wound could not restore functional epidermis; thus, delaying the closure of the wound opening in an expected manner. Failures in restoration of skin integrity delay healing due to changes in skin pathology, such as chronic ulceration or nonhealing. The role of different traditional medicines has been explored for use in the healing of cutaneous wounds, where several phytochemicals, such as flavonoids, alkaloids, phenolic acids, tannins are known to provide potential wound healing properties. However, the delivery of plant-based therapeutics could be improved by the novel platform of nanotechnology. Thus, the objectives of novel delivery strategies of principal bioactive from plant sources are to accelerate the wound healing process, avoid wound complications and enhance patient compliance. Therefore, the opportunities of nanotechnology-based drug delivery of natural wound healing therapeutics have been included in the present discussion with special emphasis on nanofibers, vesicular structures, nanoparticles, nanoemulsion, and nanogels.
Collapse
|
34
|
Xiong NX, Ou J, Li SY, Zhao JH, Huang JF, Li KX, Luo SW, Liu SJ, Wen M, Wu C, Wang S, Luo KK, Hu FZ, Liu QF. A novel ferritin L (FerL) in hybrid crucian carp could participate in host defense against Aeromonas hydrophila infection and diminish inflammatory signals. FISH & SHELLFISH IMMUNOLOGY 2022; 120:620-632. [PMID: 34968709 DOI: 10.1016/j.fsi.2021.12.040] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 11/12/2021] [Accepted: 12/23/2021] [Indexed: 06/14/2023]
Abstract
FerL, a multifunctional iron-storage polypeptide, not only exhibited a regulatory role in iron metabolism, but also participated in the regulation of fish immunity. In this study, ORF sequence of WR-FerL was 522 bp, encoding 173 amino acid residues. Tissue-specific analysis revealed that the highest expression of WR-FerL was detected in spleen. A. hydrophila challenge and LPS stimulation could sharply enhance WR-FerL mRNA expression in tissues and fish cells, respectively. Purified WR-FerL fusion peptide exhibited in vitro binding activity to A. hydrophila and endotoxin, limited bacterial dissemination to tissues as well as attenuated A. hydrophila-induced production of pro-inflammatory cytokines. Moreover, WR-FerL overexpression could abrogate NF-κB and TNFα promoter activity in fish cells. These results indicated that WR-FerL could play an important role in host defense against A. hydrophila infection.
Collapse
Affiliation(s)
- Ning-Xia Xiong
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, PR China
| | - Jie Ou
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, PR China
| | - Shi-Yun Li
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, PR China
| | - Jia-Hui Zhao
- Foreign Studies College, Hunan Normal University, Changsha, 410081, PR China
| | - Jin-Fang Huang
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, PR China
| | - Ke-Xin Li
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, PR China
| | - Sheng-Wei Luo
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, PR China.
| | - Shao-Jun Liu
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, PR China.
| | - Ming Wen
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, PR China
| | - Chang Wu
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, PR China
| | - Shi Wang
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, PR China
| | - Kai-Kun Luo
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, PR China
| | - Fang-Zhou Hu
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, PR China
| | - Qing-Feng Liu
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, PR China
| |
Collapse
|
35
|
Xiong NX, Luo SW, Mao ZW, Fan LF, Luo KK, Wang S, Hu FZ, Wen M, Liu QF, Liu SJ. Ferritin H can counteract inflammatory response in hybrid fish and its parental species after Aeromonas hydrophila infection. Comp Biochem Physiol C Toxicol Pharmacol 2021; 250:109174. [PMID: 34461291 DOI: 10.1016/j.cbpc.2021.109174] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Revised: 08/06/2021] [Accepted: 08/22/2021] [Indexed: 12/18/2022]
Abstract
Ferritin H can participate in the regulation of fish immunity. Tissue-specific analysis revealed that the highest expressions of Ferritin H in parental species were observed in spleen, while peaked level of Ferritin H mRNA in hybrid fish was observed in liver. In addition, A. hydrophila challenge could sharply enhance their Ferritin H mRNA expression in liver, kidney and spleen. To further investigate their roles in immune regulation, their Ferritin H fusion proteins were produced in vitro. Ferritin H fusion proteins could exhibit a direct binding activity to A. hydrophila and endotoxin in a dose-dependent manner, restrict dissemination of A. hydrophila to tissues and abrogate inflammatory cascades. Moreover, treatment with Ferritin H fusion proteins could reduce A. hydrophila-induced lipid peroxidation. These results indicated that Ferritin H in hybrid fish elicited a similar immune regulation of A. hydrophila-induced inflammatory signals in comparison with those of its parents.
Collapse
Affiliation(s)
- Ning-Xia Xiong
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha 410081, PR China
| | - Sheng-Wei Luo
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha 410081, PR China.
| | - Zhuang-Wen Mao
- Hunan Provincial Key Laboratory of Nutrition and Quality Control of Aquatic Animals, Department of Biological and Environmental Engineering, Changsha University, Changsha 410022, PR China
| | - Lan-Fen Fan
- College of Marine Sciences, South China Agricultural University, Guangzhou 510642, PR China
| | - Kai-Kun Luo
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha 410081, PR China
| | - Shi Wang
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha 410081, PR China
| | - Fang-Zhou Hu
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha 410081, PR China
| | - Ming Wen
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha 410081, PR China
| | - Qing-Feng Liu
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha 410081, PR China
| | - Shao-Jun Liu
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha 410081, PR China.
| |
Collapse
|
36
|
Chetta KE, Alcorn JL, Baatz JE, Wagner CL. Cytotoxic Lactalbumin-Oleic Acid Complexes in the Human Milk Diet of Preterm Infants. Nutrients 2021; 13:4336. [PMID: 34959888 PMCID: PMC8707396 DOI: 10.3390/nu13124336] [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] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 11/23/2021] [Accepted: 11/28/2021] [Indexed: 12/13/2022] Open
Abstract
Frozen storage is necessary to preserve expressed human milk for critically ill and very preterm infants. Milk pasteurization is essential for donor milk given to this special population. Due to these storage and processing conditions, subtle changes occur in milk nutrients. These changes may have clinical implications. Potentially, bioactive complexes of unknown significance could be found in human milk given to preterm infants. One such complex, a cytotoxic α-lactalbumin-oleic acid complex named "HAMLET," (Human Alpha-Lactalbumin Made Lethal to Tumor cells) is a folding variant of alpha-lactalbumin that is bound to oleic acid. This complex, isolated from human milk casein, has specific toxicity to both carcinogenic cell lines and immature non-transformed cells. Both HAMLET and free oleic acid trigger similar apoptotic mechanisms in tissue and stimulate inflammation via the NF-κB and MAPK p38 signaling pathways. This protein-lipid complex could potentially trigger various inflammatory pathways with unknown consequences, especially in immature intestinal tissues. The very preterm population is dependent on human milk as a medicinal and broadly bioactive nutriment. Therefore, HAMLET's possible presence and bioactive role in milk should be addressed in neonatal research. Through a pediatric lens, HAMLET's discovery, formation and bioactive benefits will be reviewed.
Collapse
Affiliation(s)
- Katherine E. Chetta
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, Medical University of South Carolina, Shawn Jenkins Children’s Hospital, 10 McClennan Banks Drive, MSC 915, Charleston, SC 29425, USA; (J.E.B.); (C.L.W.)
| | - Joseph L. Alcorn
- Department of Pediatrics, Division of Neonatology and Pediatric Research Center, The University of Texas Health & Science Center at Houston, 6631 Fannin Street MSB 3.252, Houston, TX 77030, USA;
| | - John E. Baatz
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, Medical University of South Carolina, Shawn Jenkins Children’s Hospital, 10 McClennan Banks Drive, MSC 915, Charleston, SC 29425, USA; (J.E.B.); (C.L.W.)
| | - Carol L. Wagner
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, Medical University of South Carolina, Shawn Jenkins Children’s Hospital, 10 McClennan Banks Drive, MSC 915, Charleston, SC 29425, USA; (J.E.B.); (C.L.W.)
| |
Collapse
|
37
|
Yarci E, Tayman C, Ozturk Yarci D, Cakir U, Gonel A, Taskin Turkmenoglu T. Inhibition of Interleukin-6 signaling: A novel therapeutic approach to necrotizing enterocolitis. Int Immunopharmacol 2021; 101:108358. [PMID: 34782273 DOI: 10.1016/j.intimp.2021.108358] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 11/07/2021] [Accepted: 11/07/2021] [Indexed: 10/19/2022]
Abstract
OBJECTIVES This study investigated the effects of tocilizumab on the prevention and treatment of experimental necrotizing enterocolitis (NEC) in newborn rats. METHODS Forty-two newborn Sprague-Dawley rats were randomly separated into three groups: NEC + placebo, NEC + tocilizumab, and the control group. NEC + placebo and NEC + tocilizumab groups were given 1 mg/kg lipopolysaccharide intraperitoneally once only on the first day, were fed with a special rodent formula every 3 h, inhaled 100% CO2 for 10 min, were exposed to cold stress at + 4 °C for 5 min, and 97% O2 for 5 min twice a day for 3 days. NEC + tocilizumab group was treated with 8 mg/kg/day tocilizumab (Actemra®) intraperitoneally, while NEC + placebo group was given intraperitoneal 0.9% saline at a dose of 2 mL/kg/day from the first day to the end of the study. All newborn rats were sacrificed on day 4. Specimens were taken for histopathologic, immunohistochemical and biochemical evaluation from the ileum and proximal colon. RESULTS NEC + tocilizumab group had higher weight gain and survival rate compared to NEC + placebo group and clinical sickness score was reduced in NEC + tocilizumab group (p < 0.05). Lower tissue damage and apoptosis were found in the NEC + tocilizumab group compared to the NEC + placebo group (p < 0.01). Tissue Interleukin-6, Interleukin-1β, TNF-α, myeloperoxidase and caspase-3 levels were significantly decreased in the NEC + tocilizumab group (p < 0.01). CONCLUSIONS Tocilizumab could be a potential option in the prevention and treatment of NEC.
Collapse
Affiliation(s)
- Erbu Yarci
- Department of Neonatology, University of Health Sicences, Zekai Tahir Burak Womens' Health Training and Research Hospital, Ankara, Turkey
| | - Cuneyt Tayman
- Department of Neonatology, University of Health Sicences, Zekai Tahir Burak Womens' Health Training and Research Hospital, Ankara, Turkey.
| | - Didem Ozturk Yarci
- Obstetrics and Gynecology Clinics, Dr. Hulusi Alatas Elmadag Government Hospital, Ankara, Turkey
| | - Ufuk Cakir
- Department of Neonatology, University of Health Sicences, Zekai Tahir Burak Womens' Health Training and Research Hospital, Ankara, Turkey
| | - Ataman Gonel
- Department of Biochemistry, Harran University Faculty of Medicine, Sanliurfa, Turkey
| | - Tugba Taskin Turkmenoglu
- Department of Pathology, University of Health Sicences, Dıskapi Yildirim Beyazit Training and Research Hospital, Ankara, Turkey
| |
Collapse
|
38
|
Aumayer H, Leonard CA, Pesch T, Prähauser B, Wunderlin S, Guscetti F, Borel N. Chlamydia suis is associated with intestinal NF-κB activation in experimentally infected gnotobiotic piglets. Pathog Dis 2021; 78:5893292. [PMID: 32804203 PMCID: PMC8140907 DOI: 10.1093/femspd/ftaa040] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 07/30/2020] [Indexed: 02/06/2023] Open
Abstract
Chlamydia suis intestinal infection of single-animal experimental groups of gnotobiotic newborn piglets was previously reported to cause severe, temporary small intestinal epithelium damage. We investigated archived intestinal samples for pro-inflammatory nuclear factor kappa B (NF-κB) activation, Interleukin (IL)-6 and IL-8 production and immune cell influx. Samples were collected 2, 4 and 7 days post-inoculation with C. suis strain S45/6 or mock inoculum (control). Increased nuclear localization of epithelial NF-κB, representative of activation, in the jejunum and ileum of C. suis-infected animals, compared to uninfected controls, began by 2 days post-infection (dpi) and persisted through 7 dpi. Infected animals showed increased production of IL-8, peaking at 2 dpi, compared to controls. Infection-mediated CD45-positive immune cell influx into the jejunal lamina propria peaked at 7 dpi, when epithelial damage was largely resolved. Activation of NF-κB appears to be a key early event in the innate response of the unprimed porcine immune system challenged with C. suis. This results in an acute phase, coinciding with the most severe clinical symptoms, diarrhea and weight loss. Immune cells recruited shortly after infection remain present in the lamina propria during the recovery phase, which is characterized by reduced chlamydial shedding and restored intestinal epithelium integrity.
Collapse
Affiliation(s)
- Helen Aumayer
- Department of Pathobiology, Institute of Veterinary Pathology, Vetsuisse Faculty, University of Zurich, Winterthurerstrasse 268, CH-8057 Zurich, Switzerland
| | - Cory Ann Leonard
- Department of Pathobiology, Institute of Veterinary Pathology, Vetsuisse Faculty, University of Zurich, Winterthurerstrasse 268, CH-8057 Zurich, Switzerland
| | - Theresa Pesch
- Department of Pathobiology, Institute of Veterinary Pathology, Vetsuisse Faculty, University of Zurich, Winterthurerstrasse 268, CH-8057 Zurich, Switzerland
| | - Barbara Prähauser
- Department of Pathobiology, Institute of Veterinary Pathology, Vetsuisse Faculty, University of Zurich, Winterthurerstrasse 268, CH-8057 Zurich, Switzerland
| | - Sabina Wunderlin
- Department of Pathobiology, Institute of Veterinary Pathology, Vetsuisse Faculty, University of Zurich, Winterthurerstrasse 268, CH-8057 Zurich, Switzerland
| | - Franco Guscetti
- Department of Pathobiology, Institute of Veterinary Pathology, Vetsuisse Faculty, University of Zurich, Winterthurerstrasse 268, CH-8057 Zurich, Switzerland
| | - Nicole Borel
- Department of Pathobiology, Institute of Veterinary Pathology, Vetsuisse Faculty, University of Zurich, Winterthurerstrasse 268, CH-8057 Zurich, Switzerland
| |
Collapse
|
39
|
Wu YZ, Chan KYY, Leung KT, Lam HS, Tam YH, Lee KH, Li K, Ng PC. The miR-223/nuclear factor I-A axis regulates inflammation and cellular functions in intestinal tissues with necrotizing enterocolitis. FEBS Open Bio 2021; 11:1907-1920. [PMID: 33932136 PMCID: PMC8255851 DOI: 10.1002/2211-5463.13164] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 02/05/2021] [Accepted: 04/12/2021] [Indexed: 01/01/2023] Open
Abstract
We previously demonstrated that microRNA(miR)‐223 is overexpressed in intestinal tissue of infants with necrotizing enterocolitis (NEC). The objective of the current study was to identify the target gene of miR‐223 and to investigate the role of the miR‐223/nuclear factor I‐A (NFIA) axis in cellular functions that underpin the pathophysiology of NEC. The target gene of miR‐223 was identified by in silico target prediction bioinformatics, luciferase assay, and western blotting. We investigated downstream signals of miR‐223 and cellular functions by overexpressing the miRNA in Caco‐2 and FHs74 cells stimulated with lipopolysaccharide or lipoteichoic acid (LTA). NFIA was identified as a target gene of miR‐223. Overexpression of miR‐223 significantly induced MYOM1 and inhibited NFIA and RGN in Caco‐2 cells, while costimulation with LTA decreased expression of GNA11, MYLK, and PRKCZ. Expression levels of GNA11, MYLK, IL‐6, and IL‐8 were increased, and levels of NFIA and RGN were decreased in FHs74 cells. These potential downstream genes were significantly correlated with levels of miR‐223 or NFIA in primary NEC tissues. Overexpression of miR‐223 significantly increased apoptosis of Caco‐2 and FHs74 cells, while proliferation of FHs74 was inhibited. These results suggest that upon binding with NFIA, miR‐223 regulates functional effectors in pathways of apoptosis, cell proliferation, G protein signaling, inflammation, and smooth muscle contraction. The miR‐223/NFIA axis may play an important role in the pathophysiology of NEC by enhancing inflammation and tissue damage.
Collapse
Affiliation(s)
- Yu Zheng Wu
- Department of Paediatrics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong
| | - Kathy Yuen Yee Chan
- Department of Paediatrics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong
| | - Kam Tong Leung
- Department of Paediatrics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong
| | - Hugh Simon Lam
- Department of Paediatrics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong
| | - Yuk Him Tam
- Department of Surgery, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Kim Hung Lee
- Department of Surgery, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Karen Li
- Department of Paediatrics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong
| | - Pak Cheung Ng
- Department of Paediatrics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong
| |
Collapse
|
40
|
Son YJ, Shin JM, Ha IJ, Erdenebileg S, Jung DS, Kim YS, Kim SM, Nho CW. Identification of Chemical Compounds from Artemisia gmelinii using UPLC-QTOF-MS/MS and their Regulatory Effects on Immune Responses in DSS-Induced Colitis Mice. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2021; 49:941-963. [PMID: 33827384 DOI: 10.1142/s0192415x21500452] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Artemisia gmelinii Web. ex Stechm. (AG), a popular medicinal herb in Asia, has been used as a common food ingredient in Korea and is traditionally known for its anti-inflammatory properties. Therefore, in this study, we aimed to investigate whether AG relieves IBD, a classic chronic inflammatory disease of the gastrointestinal tract. We identified 35 chemical compounds in AG ethanol extract using ultra-high-performance liquid chromatography-quadrupole time-of-flight tandem mass spectrometry. In mice with DSS-induced IBD, AG administration attenuated the disease activity index and the serum and colonic levels of inflammatory cytokines and chemokines. AG treatment decreased nuclear factor-[Formula: see text]B (NF-[Formula: see text]B) signaling, a key mediator of inflammation, in the mouse colons. Additionally, AG extract enhanced immune responses in lymphoid tissues such as spleen and Peyer's patches. Thus, AG consumption potently ameliorated IBD symptoms and improved immune signaling in lymphoid tissues.
Collapse
Affiliation(s)
- Yang-Ju Son
- Smart Farm Research Center, Gangneung Institute of Natural Products, Korea Institute of Science and Technology (KIST), Gangneung, Gangwon-do 25451, Korea
| | - Ji Min Shin
- Smart Farm Research Center, Gangneung Institute of Natural Products, Korea Institute of Science and Technology (KIST), Gangneung, Gangwon-do 25451, Korea.,Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology, Seoul 02792, Korea
| | - In Jin Ha
- Korean Medicine Clinical Trial Center (K-CTC), Kyung Hee University Korean Medicine Hospital, Seoul 02454, Korea
| | - Saruul Erdenebileg
- Smart Farm Research Center, Gangneung Institute of Natural Products, Korea Institute of Science and Technology (KIST), Gangneung, Gangwon-do 25451, Korea.,Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology, Seoul 02792, Korea
| | - Da Seul Jung
- Smart Farm Research Center, Gangneung Institute of Natural Products, Korea Institute of Science and Technology (KIST), Gangneung, Gangwon-do 25451, Korea.,Department of Biology, College of Natural Sciences, Gangneung-Wonju National University, Gangneung, Gangwon-do 25457, Korea
| | - Young Sik Kim
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul 08826, Korea
| | - Sang Min Kim
- Smart Farm Research Center, Gangneung Institute of Natural Products, Korea Institute of Science and Technology (KIST), Gangneung, Gangwon-do 25451, Korea.,Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology, Seoul 02792, Korea
| | - Chu Won Nho
- Smart Farm Research Center, Gangneung Institute of Natural Products, Korea Institute of Science and Technology (KIST), Gangneung, Gangwon-do 25451, Korea.,Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology, Seoul 02792, Korea
| |
Collapse
|
41
|
Kocamaz H, Özdemir ÖM, Türk NŞ, Enli Y, Şahin B, Ergin H. Dose-dependent effects of adalimumab in neonatal rats with hypoxia/reoxygenation-induced intestinal damage. Bosn J Basic Med Sci 2021; 21:33-38. [PMID: 32651971 PMCID: PMC7861631 DOI: 10.17305/bjbms.2020.4823] [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: 05/08/2020] [Accepted: 07/02/2020] [Indexed: 11/20/2022] Open
Abstract
Tumor necrosis factor-alpha (TNF-α) has an important role in hypoxia/reoxygenation (H/R)-induced intestinal damage. It was shown that blocking TNF-α with infliximab has beneficial effects on experimental necrotizing enterocolitis and hypoxic intestinal injury. However, there is no data about the effect of adalimumab on H/R-induced intestinal damage. Therefore, we aimed to determine potential dose-dependent benefits of adalimumab in such damage in neonatal rats. Wistar albino rat pups were assigned to one of the four groups: control group, hypoxia group, low-dose adalimumab (5 mg/kg/day) treated group (LDAT), and high-dose adalimumab (50 mg/kg/day) treated group (HDAT). On the fourth day of the experiment, all rats except for the control group were exposed to H/R followed by euthanasia. Malondialdehyde (MDA), myeloperoxidase (MPO), TNF-α, total antioxidant capacity (TAC), and total oxidant capacity (TOC) were measured in intestinal tissue. TAC and TOC values were used to calculate the oxidative stress index (OSI). Histopathological injury scores (HIS) were also evaluated in the tissue samples. MDA levels were significantly lower in the LDAT and HDAT groups (p < 0.001). TNF-α levels were significantly lower in the LDAT group (p < 0.001). OSI was significantly higher in the H/R group than in the control and LDAT groups (p < 0.001). Mean HIS values in the LDAT group were significantly lower than those in the H/R and HDAT groups (p < 0.001). This experimental study showed that low-dose adalimumab appears to have a beneficial effect on intestinal injury induced with H/R in neonatal rats.
Collapse
Affiliation(s)
- Halil Kocamaz
- Department of Pediatric Gastroenterology, Faculty of Medicine, Pamukkale University, Denizli, Turkey
| | - Özmert Ma Özdemir
- Division of Neonatology, Department of Pediatrics, Faculty of Medicine, Pamukkale University, Denizli, Turkey
| | - Nilay Şen Türk
- Department of Pathology, Faculty of Medicine, Pamukkale University, Denizli, Turkey
| | - Yaşar Enli
- Department of Biochemistry, Faculty of Medicine, Pamukkale University, Denizli, Turkey
| | - Barbaros Şahin
- Experimental Animal Study Laboratory, Pamukkale University, Denizli, Turkey
| | - Hacer Ergin
- Division of Neonatology, Department of Pediatrics, Faculty of Medicine, Pamukkale University, Denizli, Turkey
| |
Collapse
|
42
|
Gomart A, Vallée A, Lecarpentier Y. Necrotizing Enterocolitis: LPS/TLR4-Induced Crosstalk Between Canonical TGF-β/Wnt/β-Catenin Pathways and PPARγ. Front Pediatr 2021; 9:713344. [PMID: 34712628 PMCID: PMC8547806 DOI: 10.3389/fped.2021.713344] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Accepted: 09/13/2021] [Indexed: 12/13/2022] Open
Abstract
Necrotizing enterocolitis (NEC) represents one of the major causes of morbidity and mortality in premature infants. Several recent studies, however, have contributed to a better understanding of the pathophysiology of this dreadful disease. Numerous intracellular pathways play a key role in NEC, namely: bacterial lipopolysaccharide (LPS), LPS toll-like receptor 4 (TLR4), canonical Wnt/β-catenin signaling and PPARγ. In a large number of pathologies, canonical Wnt/β-catenin signaling and PPARγ operate in opposition to one another, so that when one of the two pathways is overexpressed the other is downregulated and vice-versa. In NEC, activation of TLR4 by LPS leads to downregulation of the canonical Wnt/β-catenin signaling and upregulation of PPARγ. This review aims to shed light on the complex intracellular mechanisms involved in this pathophysiological profile by examining additional pathways such as the GSK-3β, NF-κB, TGF-β/Smads, and PI3K-Akt pathways.
Collapse
Affiliation(s)
- Alexia Gomart
- Département de Pédiatrie et Médecine de l'adolescent, Centre Hospitalier Intercommunal de Créteil, Créteil, France
| | - Alexandre Vallée
- Department of Clinical Research and Innovation, Foch Hospital, Suresnes, France
| | - Yves Lecarpentier
- Centre de Recherche Clinique, Grand Hôpital de l'Est Francilien, Meaux, France
| |
Collapse
|
43
|
Zerra PE, Josephson CD. Transfusion in Neonatal Patients: Review of Evidence-Based Guidelines. Clin Lab Med 2020; 41:15-34. [PMID: 33494882 DOI: 10.1016/j.cll.2020.10.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Transfusion of red blood cells, platelets, and fresh frozen plasma in neonatal patients has not been well characterized in the literature, with guidelines varying greatly between institutions. However, anemia and thrombocytopenia are highly prevalent, especially in preterm neonates. When transfusing a neonatal patient, clinicians must take into consideration physiologic differences, gestational and postnatal age, congenital disorders, and maternal factors while weighing the risks and benefits of transfusion. This review of existing literature summarizes current evidence-based neonatal transfusion guidelines and highlights areas of current ongoing research and those in need of future studies.
Collapse
Affiliation(s)
- Patricia E Zerra
- Department of Pathology and Laboratory Medicine, Emory University Hospital, 1364 Clifton Road NE, Atlanta, GA 30322, USA; Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Egleston Hospital, 1405 Clifton Rd, Atlanta, GA 30322, USA
| | - Cassandra D Josephson
- Department of Pathology and Laboratory Medicine, Emory University Hospital, 1364 Clifton Road NE, Atlanta, GA 30322, USA; Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Egleston Hospital, 1405 Clifton Rd, Atlanta, GA 30322, USA.
| |
Collapse
|
44
|
Kim YC, Lee SR, Jeon HJ, Kim K, Kim MJ, Choi SD, Lee SE. Acute toxicities of fluorene, fluorene-1-carboxylic acid, and fluorene-9-carboxylic acid on zebrafish embryos (Danio rerio): Molecular mechanisms of developmental toxicities of fluorene-1-carboxylic acid. CHEMOSPHERE 2020; 260:127622. [PMID: 32673875 DOI: 10.1016/j.chemosphere.2020.127622] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 07/03/2020] [Accepted: 07/05/2020] [Indexed: 06/11/2023]
Abstract
In this study, fluorene (FL), FL-1-carboxylic acid (FC-1), and FL-9-carboxylic acid (FC-9) were investigated to understand their acute toxicity by measuring inhibitory effects on hatching rates and developmental processes of zebrafish embryos (Danio rerio). For exposure concentrations up to 3000 μg/L, FC-1 alone showed acute toxicity at 1458 μg/L for LC50 value. FC-1 caused yolk sac and spinal deformities, and pericardial edema. Molecular studies were undertaken to understand FC-1 toxicity examining 61 genes after exposure to 5 μM (equivalent to LC20 value of FC-1) in embryos. In the FC-1-treated embryos, the expression of the cyp7a1 gene, involved in bile acid biosynthesis, was dramatically decreased, while the expression of the Il-1β gene involved in inflammation was remarkably increased. In addition to these findings, in FC-1-treated embryos, the expression of nppa gene related to the differentiation of the myocardium was 3-fold increased. On the other hand, cyp1a, cyp3a, ugt1a1, abcc4, mdr1, and sult1st1 responsible for detoxification of xenobiotics were upregulated in FC-9-treated embryos. Taken together, carboxylation on carbon 1 of FL increased acute toxicity in zebrafish embryos, and its toxicity might be related to morphological changes with modification of normal biological functions and lowered defense ability.
Collapse
Affiliation(s)
- Yong-Chan Kim
- School of Applied Biosciences, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Sang-Ryong Lee
- Department of Biological and Environmental Science, Dongguk University, Goyang-si, Gyeonggi-do, 10326, Republic of Korea
| | - Hwang-Ju Jeon
- School of Applied Biosciences, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Kyeongnam Kim
- School of Applied Biosciences, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Myoung-Jin Kim
- School of Applied Biosciences, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Sung-Deuk Choi
- School of Urban and Environmental Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Sung-Eun Lee
- School of Applied Biosciences, Kyungpook National University, Daegu, 41566, Republic of Korea.
| |
Collapse
|
45
|
Chen W, Wang X, Yan X, Yu Z, Zhang J, Han S. The emerging role of exosomes in the pathogenesis, prognosis and treatment of necrotizing enterocolitis. Am J Transl Res 2020; 12:7020-7033. [PMID: 33312348 PMCID: PMC7724339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Accepted: 08/22/2020] [Indexed: 06/12/2023]
Abstract
Exosomes are a subtype of extracellular vesicles. They contain bioactive molecules, including nucleic acids, proteins and lipids. Among the currently described exosomes, a majority are potential candidates for the diagnosis and treatment of necrotizing enterocolitis (NEC). In this work, we reviewed existing literature reports on exosomes and explored their roles in NEC. Exosomes derived from intestinal epithelial cells (IECs) participates in the development of intestinal diseases, thus can potentially be utilized as biomarkers for NEC. Besides, exosomes of human milk have been demonstrated to protect IECs from oxidative stress, stimulate intestinal stem cells activity, improve the proliferation and migration of IECs, and lower the incidence and severity of experimental NEC. Further, exosomes produced by stem cells can reduce the severity of experimental NEC and protect the intestinal barrier function during NEC. Conclusively, exosomes have been shown to influence the pathogenesis of NEC and exert a protective effect on NEC. However, additional investigations would be urgently necessary to comprehensively elucidate the underlying mechanisms of exosomes in NEC.
Collapse
Affiliation(s)
- Wenjuan Chen
- Department of Pediatrics, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital No. 123 Tian Fei Xiang, Mo Chou Road, Nanjing 210004, Jiangsu Province, China
| | - Xingyun Wang
- Department of Pediatrics, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital No. 123 Tian Fei Xiang, Mo Chou Road, Nanjing 210004, Jiangsu Province, China
| | - Xiangyun Yan
- Department of Pediatrics, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital No. 123 Tian Fei Xiang, Mo Chou Road, Nanjing 210004, Jiangsu Province, China
| | - Zhangbin Yu
- Department of Pediatrics, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital No. 123 Tian Fei Xiang, Mo Chou Road, Nanjing 210004, Jiangsu Province, China
| | - Jun Zhang
- Department of Pediatrics, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital No. 123 Tian Fei Xiang, Mo Chou Road, Nanjing 210004, Jiangsu Province, China
| | - Shuping Han
- Department of Pediatrics, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital No. 123 Tian Fei Xiang, Mo Chou Road, Nanjing 210004, Jiangsu Province, China
| |
Collapse
|
46
|
Yakut HI, Koyuncu E, Cakir U, Tayman C, Koyuncu İ, Taskin Turkmenoglu T, Cakir E, Ozyazici A, Aydogan S, Zenciroglu A. Preventative and therapeutic effects of fennel (Foeniculum vulgare) seed extracts against necrotizing enterocolitis. J Food Biochem 2020; 44:e13284. [PMID: 32510653 DOI: 10.1111/jfbc.13284] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 04/22/2020] [Accepted: 04/27/2020] [Indexed: 02/06/2023]
Abstract
We aimed to understand the efficacy of fennel (Foeniculum vulgare: FV) extract in an experimental necrotizing enterocolitis (NEC) model. Forty-two rat pups were divided into three groups as NEC, NEC treated with fennel extract, and control. At the end of the experiment, tissue samples were taken from the proximal colon and ileum for biochemical and immuno-histological studies including hematoxylin-eosin and Caspase-3-8-9 immunohistochemical staining. Bowel damage and apoptosis were found to be less in the NEC + FV group. Oxidant stress, caspase 3, TNF-α, and IL-6 levels were considerably decreased in the NEC + FV group. Antioxidants were significantly higher in the NEC + FV group more than in the NEC group. Moreover, protein, DNA damage, and lipid peroxidation were found to be decreased in the NEC + FV group compared to the NEC group. PRACTICAL APPLICATIONS: Intense inflammation, oxidant stress, apoptosis, and infection are important in the development of NEC. Fennel has anti-oxidant, anti-inflammatory, antibacterial, antifungal, antiviral, immunomodulatory effects. Fennel extract might be a novel option in the treatment of NEC through its anti-oxidant, anti-inflammatory, anti-apoptotic, and cytoprotective features.
Collapse
Affiliation(s)
- Halil Ibrahim Yakut
- Department of Pediatrics, Ankara Hematology Oncology Children Education and Research Hospital, Health Sciences University, Ankara, Turkey
| | - Ece Koyuncu
- Department of Neonatology, Dr. Sami Ulus Maternity and Children Education and Research Hospital, Health Sciences University, Ankara, Turkey
| | - Ufuk Cakir
- Department of Neonatology, Zekai Tahir Burak Maternity Education and Research Hospital, Health Sciences University, Ankara, Turkey
| | - Cuneyt Tayman
- Department of Neonatology, Zekai Tahir Burak Maternity Education and Research Hospital, Health Sciences University, Ankara, Turkey
| | - İsmail Koyuncu
- Faculty of Medicine, Department of Biochemistry, Harran University, Sanlıurfa, Turkey
| | - Tugba Taskin Turkmenoglu
- Department of Pathology, Ankara Yıldırım Beyazıt Education and Research Hospital, Health Sciences University, Ankara, Turkey
| | - Esra Cakir
- Anesthesiology and Clinical of Critical Care, Ankara Numune Education and Research Hospital, Health Sciences University, Ankara, Turkey
| | - Ahmet Ozyazici
- Department of Neonatology, Dr. Sami Ulus Maternity and Children Education and Research Hospital, Health Sciences University, Ankara, Turkey
| | - Seda Aydogan
- Department of Neonatology, Dr. Sami Ulus Maternity and Children Education and Research Hospital, Health Sciences University, Ankara, Turkey
| | - Aysegul Zenciroglu
- Department of Neonatology, Dr. Sami Ulus Maternity and Children Education and Research Hospital, Health Sciences University, Ankara, Turkey
| |
Collapse
|
47
|
Han SH, Lee NR, Kim HJ, Kang YD, Kim JS, Park JW, Jin HJ. Association between the IL-6, IL-10, and TNFα gene polymorphisms and preterm-birth in Korean women. Genes Genomics 2020; 42:743-750. [DOI: 10.1007/s13258-020-00946-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 05/12/2020] [Indexed: 12/18/2022]
|
48
|
Groer MW, Miller EM, D'Agata A, Ho TTB, Dutra SV, Yoo JY, Yee AL, Gilbert JA, Dishaw LJ. Contributors to Dysbiosis in Very-Low-Birth-Weight Infants. J Obstet Gynecol Neonatal Nurs 2020; 49:232-242. [PMID: 32247727 DOI: 10.1016/j.jogn.2020.02.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/01/2020] [Indexed: 02/08/2023] Open
Abstract
The objective of this commentary was to analyze the causes and outcomes of gut microbiome dysbiosis in preterm infants who are born at very low birth weight (VLBW). The intrauterine development of VLBW infants is interrupted abruptly with preterm birth and followed by extrauterine, health-threatening conditions and sequelae. These infants develop intestinal microbial dysbiosis characterized by low diversity, an overall reduction in beneficial and/or commensal bacteria, and enrichment of opportunistic pathogens of the Gammaproteobacteria class. The origin of VLBW infant dysbiosis is not well understood and is likely the result of a combination of immaturity and medical care. We propose that these factors interact to produce inflammation in the gut, which further perpetuates dysbiosis. Understanding the sources of dysbiosis could result in interventions to reduce gut inflammation, decrease enteric pathology, and improve health outcomes for these vulnerable infants.
Collapse
|
49
|
Tang J, Guo C, Gong F. [Protective effect of Lactobacillus reuteri against oxidative stress in neonatal mice with necrotizing enterocolitis]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2019; 39:1221-1226. [PMID: 31801706 DOI: 10.12122/j.issn.1673-4254.2019.10.14] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE To investigate the protective effect of L. reuteri DSM17938 strain against oxidative stress in a neonatal mouse model of necrotizing enterocolitis (NEC) and explore the possible mechanism. METHODS Ninety-six 10-day-old neonatal C57BL/6J mice were equally randomized into control group, NEC group, and NEC+ L. reuteri group. The pathological changes of the ileocecal intestinal tissue were evaluated with HE staining and double-blind pathological scoring. The mRNA and protein expressions of tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β) in the intestinal tissues were detected using quantitative real-time PCR and ELISA, respectively. Colorimetric assays were used to determine the activity of superoxide dismutase (SOD) and its inhibition rate, malondialdehyde (MDA), glutathione (GSH), oxidized glutathione (GSSG), and GSSG/ GSH ratio. RESULTS Compared with those in the control group, the neonatal mice in NEC group showed significant weight loss (P < 0.05), obvious intestinal injury, increased pathological scores (P < 0.05), increased expressions of TNF-α and IL-1β mRNA and proteins (P < 0.05), decreased SOD activity and inhibition rate, decreased GSH, and significantly increased MDA, GSSG, and GSSG/GSH ratios (P < 0.05). Treatment with L. reuteri obviously decreased the pathological scores, expressions of TNF-α and IL-1β (P < 0.05), MDA, GSSG, and GSSG/GSH ratio (P < 0.05), and significantly increased SOD activity, its inhibition rate, and GSH level in the mice with NEC, but the survival rate was not significantly different between NEC and L. reuteri-treated groups (P > 0.05). CONCLUSIONS L. reuteri DSM17938 can offer protection against NEC in mice by reducing oxidative stress and increasing antioxidant capacity of the intestinal tissue to suppress intestinal inflammations.
Collapse
Affiliation(s)
- Jia Tang
- Department of Pediatrics, Yongchuan Hospital Affiliated to Chongqing Medical University, Chongqing 402160, China
| | - Chunbao Guo
- Department of Hepatobiliary Surgery, Children's Hospital Affiliated to Chongqing Medical University, Chongqing 400016, China
| | - Fang Gong
- Department of Pediatrics, Yongchuan Hospital Affiliated to Chongqing Medical University, Chongqing 402160, China
| |
Collapse
|
50
|
Dithizone-induced Paneth cell disruption significantly decreases intestinal perfusion in the murine small intestine. J Pediatr Surg 2019; 54:2402-2407. [PMID: 30857731 PMCID: PMC6707906 DOI: 10.1016/j.jpedsurg.2019.02.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 02/06/2019] [Accepted: 02/17/2019] [Indexed: 02/06/2023]
Abstract
PURPOSE Necrotizing enterocolitis is associated with decreased intestinal perfusion and ischemia. Paneth cells, specialized epithelial cells, have been shown to regulate the intestinal vasculature and disruption of these cells has been associated with NEC. We hypothesized that Paneth cell disruption in immature mice intestine would decrease the perfusion of the intestinal microvasculature. METHODS Paneth cells were disrupted in P14-16 mice using chemical (dithizone) and transgenic (diphtheria toxin) methodology. Six hours after Paneth cell disruption, Dylight 488 was injected directly into the left ventricle and allowed to perfuse for 5 minutes prior to intestinal harvesting. Tissue samples were evaluated with confocal fluorescence microscopy to quantify intestinal perfusion and samples were quantified by real time RT-PCR for gene expression. RESULTS Dithizone treatment significantly decreased intestinal perfusion compared to controls (p < 0.01). However, diphtheria toxin treatment demonstrated no significant difference in perfusion (p > 0.21). Intestines from all treatment groups had similar PECAM staining, but intestines treated with dithizone had significantly decreased nNOS and iNOS gene expression compared to controls (p < 0.007). CONCLUSIONS Paneth cell disruption significantly decreases the perfusion of the small intestinal microvasculature in a dithizone-specific manner. Dithizone has no effect on the amount of microvasculature, but does impact genes critical to nitric oxide signaling likely contributing to mesenteric vasoconstriction.
Collapse
|