1
|
Michalak KP, Michalak AZ, Brenk-Krakowska A. Acute COVID-19 and LongCOVID syndrome - molecular implications for therapeutic strategies - review. Front Immunol 2025; 16:1582783. [PMID: 40313948 PMCID: PMC12043656 DOI: 10.3389/fimmu.2025.1582783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Accepted: 03/28/2025] [Indexed: 05/03/2025] Open
Abstract
Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) has been recognized not only for its acute effects but also for its ability to cause LongCOVID Syndrome (LCS), a condition characterized by persistent symptoms affecting multiple organ systems. This review examines the molecular and immunological mechanisms underlying LCS, with a particular focus on autophagy inhibition, chronic inflammation, oxidative, nitrosative and calcium stress, viral persistence and autoimmunology. Potential pathophysiological mechanisms involved in LCS include (1) autoimmune activation, (2) latent viral persistence, where SARS-CoV-2 continues to influence host metabolism, (3) reactivation of latent pathogens such as Epstein-Barr virus (EBV) or cytomegalovirus (CMV), exacerbating immune and metabolic dysregulation, and (4) possible persistent metabolic and inflammatory dysregulation, where the body fails to restore post-infection homeostasis. The manipulation of cellular pathways by SARS-CoV-2 proteins is a critical aspect of the virus' ability to evade immune clearance and establish long-term dysfunction. Viral proteins such as NSP13, ORF3a and ORF8 have been shown to disrupt autophagy, thereby impairing viral clearance and promoting immune evasion. In addition, mitochondrial dysfunction, dysregulated calcium signaling, oxidative stress, chronic HIF-1α activation and Nrf2 inhibition create a self-sustaining inflammatory feedback loop that contributes to tissue damage and persistent symptoms. Therefore understanding the molecular basis of LCS is critical for the development of effective therapeutic strategies. Targeting autophagy and Nrf2 activation, glycolysis inhibition, and restoration calcium homeostasis may provide novel strategies to mitigate the long-term consequences of SARS-CoV-2 infection. Future research should focus on personalized therapeutic interventions based on the dominant molecular perturbations in individual patients.
Collapse
Affiliation(s)
- Krzysztof Piotr Michalak
- Laboratory of Vision Science and Optometry, Physics and Astronomy Faculty, Adam Mickiewicz University in Poznań, Poznań, Poland
| | | | - Alicja Brenk-Krakowska
- Laboratory of Vision Science and Optometry, Physics and Astronomy Faculty, Adam Mickiewicz University in Poznań, Poznań, Poland
| |
Collapse
|
2
|
Michalak KP, Michalak AZ. Understanding chronic inflammation: couplings between cytokines, ROS, NO, Ca i 2+, HIF-1α, Nrf2 and autophagy. Front Immunol 2025; 16:1558263. [PMID: 40264757 PMCID: PMC12012389 DOI: 10.3389/fimmu.2025.1558263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 03/14/2025] [Indexed: 04/24/2025] Open
Abstract
Chronic inflammation is an important component of many diseases, including autoimmune diseases, intracellular infections, dysbiosis and degenerative diseases. An important element of this state is the mainly positive feedback between inflammatory cytokines, reactive oxygen species (ROS), nitric oxide (NO), increased intracellular calcium, hypoxia-inducible factor 1-alpha (HIF-1α) stabilisation and mitochondrial oxidative stress, which, under normal conditions, enhance the response against pathogens. Autophagy and the nuclear factor erythroid 2-related factor 2 (Nrf2)-mediated antioxidant response are mainly negatively coupled with the above-mentioned elements to maintain the defence response at a level appropriate to the severity of the infection. The current review is the first attempt to build a multidimensional model of cellular self-regulation of chronic inflammation. It describes the feedbacks involved in the inflammatory response and explains the possible pathways by which inflammation becomes chronic. The multiplicity of positive feedbacks suggests that symptomatic treatment of chronic inflammation should focus on inhibiting multiple positive feedbacks to effectively suppress all dysregulated elements including inflammation, oxidative stress, calcium stress, mito-stress and other metabolic disturbances.
Collapse
Affiliation(s)
- Krzysztof Piotr Michalak
- Laboratory of Vision Science and Optometry, Physics and Astronomy Faculty, Adam Mickiewicz University in Poznań, Poznań, Poland
| | | |
Collapse
|
3
|
Silva RCMC. The dichotomic role of cytokines in aging. Biogerontology 2024; 26:17. [PMID: 39621124 DOI: 10.1007/s10522-024-10152-4] [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: 09/04/2024] [Accepted: 10/30/2024] [Indexed: 12/11/2024]
Abstract
The chronic inflammation present in aged individuals is generally depicted as a detrimental player for longevity. Here, it is discussed several beneficial effects associated with the cytokines that are chronically elevated in inflammaging. These cytokines, such as IL-1β, type I interferons, IL-6 and TNF positively regulate macroautophagy, mitochondrial function, anti-tumor immune responses and skeletal muscle biogenesis, possibly contributing to longevity. On the other side, the detrimental and antagonistic role of these cytokines including the induction of sarcopenia, tissue damage and promotion of tumorigenesis are also discussed, underscoring the dichotomy associated with inflammaging and its players. In addition, it is discussed the role of the anti-inflammatory cytokine IL-10 and other cytokines that affect aging in a more linear way, such as IL-11, which promotes senescence, and IL-4 and IL-15, which promotes longevity. It is also discussed more specific regulators of aging that are downstream cytokines-mediated signaling.
Collapse
|
4
|
Bravo Iniguez A, Sun Q, Cui Q, Du M, Zhu MJ. Cannabidiol Enhances Mitochondrial Metabolism and Antioxidant Defenses in Human Intestinal Epithelial Caco-2 Cells. Nutrients 2024; 16:3843. [PMID: 39599629 PMCID: PMC11597683 DOI: 10.3390/nu16223843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 11/05/2024] [Accepted: 11/07/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND The reintroduction of hemp production has resulted in increased consumption of cannabidiol (CBD) products, particularly CBD oil, yet their effects on intestinal health are not fully understood. Proper mitochondrial function and antioxidant defenses are vital for maintaining the intestinal epithelial barrier. AMP-activated protein kinase (AMPK) and peroxisome proliferator-activated receptor gamma coactivator (PGC)1α are key mediators of mitochondrial metabolism. METHODS & RESULTS Using Caco-2 cells, we found that CBD oil promoted AMPK phosphorylation, upregulated differentiation markers, and enhanced PGC1α/SIRT3 mitochondrial signaling. CBD oil reduced reactive oxygen species production and increased antioxidant enzymes. Moreover, CBD oil also increased levels of citrate, malate, and succinate-key metabolites of the tricarboxylic acid cycle-alongside upregulation of pyruvate dehydrogenase and isocitrate dehydrogenase 1. Similarly, pure CBD induced metabolic and antioxidant signaling. CONCLUSIONS CBD enhances mitochondrial metabolic activity and antioxidant defense in Caco-2 cells, making it a promising candidate for treating intestinal dysfunction.
Collapse
Affiliation(s)
- Alejandro Bravo Iniguez
- School of Food Science, Washington State University, Pullman, WA 99164, USA; (A.B.I.); (Q.S.); (Q.C.)
| | - Qi Sun
- School of Food Science, Washington State University, Pullman, WA 99164, USA; (A.B.I.); (Q.S.); (Q.C.)
| | - Qiaorong Cui
- School of Food Science, Washington State University, Pullman, WA 99164, USA; (A.B.I.); (Q.S.); (Q.C.)
| | - Min Du
- Department of Animal Sciences, Washington State University, Pullman, WA 99164, USA;
| | - Mei-Jun Zhu
- School of Food Science, Washington State University, Pullman, WA 99164, USA; (A.B.I.); (Q.S.); (Q.C.)
| |
Collapse
|
5
|
Zhu L, Zhu M, Li X, Shen Y, Duan S, Yang J. Functional Characterization of Ao4g24: An Uncharacterized Gene Involved in Conidiation, Trap Formation, Stress Response, and Secondary Metabolism in Arthrobotrys oligospora. Microorganisms 2024; 12:1532. [PMID: 39203374 PMCID: PMC11356499 DOI: 10.3390/microorganisms12081532] [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: 07/18/2024] [Revised: 07/23/2024] [Accepted: 07/24/2024] [Indexed: 09/03/2024] Open
Abstract
Arthrobotrys oligospora is a typical nematode-trapping (NT) fungus, which can secrete food cues to lure, capture, and digest nematodes by triggering the production of adhesive networks (traps). Based on genomic and proteomic analyses, multiple pathogenic genes and proteins involved in trap formation have been characterized; however, there are numerous uncharacterized genes that play important roles in trap formation. The functional studies of these unknown genes are helpful in systematically elucidating the complex interactions between A. oligospora and nematode hosts. In this study, we screened the gene AOL_s00004g24 (Ao4g24). This gene is similar to the SWI/SNF chromatin remodeling complex, which was found to play a potential role in trap formation in our previous transcriptome analysis. Here, we characterized the function of Ao4g24 by gene disruption, phenotypic analysis, and metabolomics. The deletion of Ao4g24 led to a remarkable decrease in conidia yield, trap formation, and secondary metabolites. Meanwhile, the absence of Ao4g24 influenced the mitochondrial membrane potential, ATP content, autophagy, ROS level, and stress response. These results indicate that Ao4g24 has crucial functions in sporulation, trap formation, and pathogenicity in NT fungi. Our study provides a reference for understanding the role of unidentified genes in mycelium growth and trap formation in NT fungi.
Collapse
Affiliation(s)
| | | | | | | | | | - Jinkui Yang
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Key Laboratory for Southwest Microbial Diversity of the Ministry of Education, and School of Life Science, Yunnan University, Kunming 650032, China; (L.Z.); (M.Z.); (X.L.); (Y.S.); (S.D.)
| |
Collapse
|
6
|
Pan X, Caillon A, Fan S, Khan S, Tomatsu S, Pshezhetsky AV. Heterologous HSPC Transplantation Rescues Neuroinflammation and Ameliorates Peripheral Manifestations in the Mouse Model of Lysosomal Transmembrane Enzyme Deficiency, MPS IIIC. Cells 2024; 13:877. [PMID: 38786099 PMCID: PMC11120110 DOI: 10.3390/cells13100877] [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: 04/02/2024] [Revised: 05/13/2024] [Accepted: 05/15/2024] [Indexed: 05/25/2024] Open
Abstract
Mucopolysaccharidosis III type C (MPS IIIC) is an untreatable neuropathic lysosomal storage disease caused by a genetic deficiency of the lysosomal N-acetyltransferase, HGSNAT, catalyzing a transmembrane acetylation of heparan sulfate. HGSNAT is a transmembrane enzyme incapable of free diffusion between the cells or their cross-correction, which limits development of therapies based on enzyme replacement and gene correction. Since our previous work identified neuroinflammation as a hallmark of the CNS pathology in MPS IIIC, we tested whether it can be corrected by replacement of activated brain microglia with neuroprotective macrophages/microglia derived from a heterologous HSPC transplant. Eight-week-old MPS IIIC (HgsnatP304L) mice were transplanted with HSPC from congenic wild type mice after myeloablation with Busulfan and studied using behavior test battery, starting from the age of 6 months. At the age of ~8 months, mice were sacrificed to study pathological changes in the brain, heparan sulfate storage, and other biomarkers of the disease. We found that the treatment corrected several behavior deficits including hyperactivity and reduction in socialization, but not memory decline. It also improved several features of CNS pathology such as microastroglyosis, expression of pro-inflammatory cytokine IL-1β, and accumulation of misfolded amyloid aggregates in cortical neurons. At the periphery, the treatment delayed development of terminal urinary retention, potentially increasing longevity, and reduced blood levels of heparan sulfate. However, we did not observe correction of lysosomal storage phenotype in neurons and heparan sulfate brain levels. Together, our results demonstrate that neuroinflammation in a neurological lysosomal storage disease, caused by defects in a transmembrane enzyme, can be effectively ameliorated by replacement of microglia bearing the genetic defect with cells from a normal healthy donor. They also suggest that heterologous HSPC transplant, if used together with other methods, such as chaperone therapy or substrate reduction therapy, may constitute an effective combination therapy for MPS IIIC and other disorders with a similar etiology.
Collapse
Affiliation(s)
- Xuefang Pan
- Department of Pediatrics and Centre Hospitalier Universitaire Sainte-Justine Research Centre, University of Montreal, Montreal, QC H3T 1C5, Canada; (X.P.); (A.C.); (S.F.)
| | - Antoine Caillon
- Department of Pediatrics and Centre Hospitalier Universitaire Sainte-Justine Research Centre, University of Montreal, Montreal, QC H3T 1C5, Canada; (X.P.); (A.C.); (S.F.)
| | - Shuxian Fan
- Department of Pediatrics and Centre Hospitalier Universitaire Sainte-Justine Research Centre, University of Montreal, Montreal, QC H3T 1C5, Canada; (X.P.); (A.C.); (S.F.)
- Department of Anatomy and Cell Biology, McGill University, Montreal, QC H3A 0C7, Canada
| | - Shaukat Khan
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE 19803, USA; (S.K.); (S.T.)
| | - Shunji Tomatsu
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE 19803, USA; (S.K.); (S.T.)
| | - Alexey V. Pshezhetsky
- Department of Pediatrics and Centre Hospitalier Universitaire Sainte-Justine Research Centre, University of Montreal, Montreal, QC H3T 1C5, Canada; (X.P.); (A.C.); (S.F.)
- Department of Anatomy and Cell Biology, McGill University, Montreal, QC H3A 0C7, Canada
| |
Collapse
|
7
|
Ling XC, Huang PH, Chen HC, Hsueh YJ, Lee CW, Lien R, Lee CC, Chu SM, Chen KJ, Hwang YS, Lai CC, Chiang MC, Wu WC. Association of serum levels of inflammatory cytokines with retinopathy of prematurity in preterm infants. Front Pediatr 2024; 11:1195904. [PMID: 38259597 PMCID: PMC10800500 DOI: 10.3389/fped.2023.1195904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 12/19/2023] [Indexed: 01/24/2024] Open
Abstract
Introduction Retinopathy of prematurity (ROP) is a retinal vascular developmental disease associated with risks factors such as supplementary oxygen use or low birth weight/early gestational age. Multiple studies have reported associations between ROP and systemic inflammation. In this study, we investigated serum cytokines associated with ROP development and severity and assessed their applicability as potential biomarkers of ROP. Methods This prospective study was conducted at an institutional referral center between 2019 and 2021. To measure the serum levels of 40 inflammatory cytokines in eligible premature patients, we collected their serum samples during the enrollment of patients or the intravitreal injection of anti-vascular endothelial growth factor (VEGF) agents and after 2 and 4 weeks. Results Fifty patients were enrolled. In patients with type 1 ROP who received anti-VEGF agents (n = 22), the levels of serum intercellular adhesion molecule-1 decreased significantly (p < 0.05) at 4 weeks compared with the baseline level, whereas those of serum granulocyte-macrophage colony-stimulating factor increased significantly (p < 0.05). In patients with ROP who did not require any treatment (n = 14), no significant change was noted in the level of any of the 40 inflammatory cytokines. In control infants without ROP (n = 14), the serum levels of tumor necrosis factor-α, interleukin (IL)-15, and IL-12p40 increased significantly (p < 0.05) at 4 weeks. The changes in the levels of serum inflammatory cytokines did not vary significantly among the aforementioned three groups. A generalized estimating equation indicated that zone 1 ROP, stage 3 ROP, older postmenstrual age, respiratory distress syndrome, necrotizing enterocolitis, and sepsis were associated with the changes in serum cytokine levels. Conclusions Although significant changes (compared with baseline) were observed in the serum levels of certain inflammatory cytokines in patients with type 1 ROP and infants without ROP, no significant difference in cytokine level fluctuations were noted among the three groups. Changes in serum inflammatory cytokine levels may not predict ROP development or severity. Additional comprehensive studies are warranted to establish their definitive role and significance in ROP, emphasizing the need for continued research in this area.
Collapse
Affiliation(s)
- Xiao Chun Ling
- Department of Ophthalmology, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Pin-Hsuan Huang
- Center for Big Data Analytics and Statistics, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Hung-Chi Chen
- Department of Ophthalmology, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yi-Jen Hsueh
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Center for Tissue Engineering, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan, Taiwan
| | - Chia-Wen Lee
- Department of Ophthalmology, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Reyin Lien
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Division of Neonatology, Department of Pediatrics, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Chien-Chung Lee
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Division of Neonatology, Department of Pediatrics, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Shih-Ming Chu
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Division of Neonatology, Department of Pediatrics, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Kuan-Jen Chen
- Department of Ophthalmology, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yih-Shiou Hwang
- Department of Ophthalmology, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chi-Chun Lai
- Department of Ophthalmology, Chang Gung Memorial Hospital, Keelung, Taiwan
| | - Ming-Chou Chiang
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Division of Neonatology, Department of Pediatrics, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Wei-Chi Wu
- Department of Ophthalmology, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|
8
|
Yang S, Wei X, Zhong Y, Guo C, Liu X, Wang Z, Tu Y. Programmed death of intestinal epithelial cells in neonatal necrotizing enterocolitis: a mini-review. Front Pediatr 2023; 11:1199878. [PMID: 37342533 PMCID: PMC10277470 DOI: 10.3389/fped.2023.1199878] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 05/23/2023] [Indexed: 06/23/2023] Open
Abstract
Necrotizing enterocolitis (NEC) is one of the most fatal diseases in premature infants. Damage to the intestinal epithelial barrier (IEB) is an important event in the development of intestinal inflammation and the evolution of NEC. The intestinal epithelial monolayer formed by the tight arrangement of intestinal epithelial cells (IECs) constitutes the functional IEB between the organism and the extra-intestinal environment. Programmed death and regenerative repair of IECs are important physiological processes to maintain the integrity of IEB function in response to microbial invasion. However, excessive programmed death of IECs leads to increased intestinal permeability and IEB dysfunction. Therefore, one of the most fundamental questions in the field of NEC research is to reveal the pathological death process of IECs, which is essential to clarify the pathogenesis of NEC. This review focuses on the currently known death modes of IECs in NEC mainly including apoptosis, necroptosis, pyroptosis, ferroptosis, and abnormal autophagy. Furthermore, we elaborate on the prospect of targeting IECs death as a treatment for NEC based on exciting animal and clinical studies.
Collapse
Affiliation(s)
- Shuo Yang
- Department of Pharmacy, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
- Department of Critical Care Medicine, School of Anesthesiology, Naval Medical University, Shanghai, China
| | - Xin Wei
- Department of Clinical Pharmacy, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yuting Zhong
- Department of Critical Care Medicine, School of Anesthesiology, Naval Medical University, Shanghai, China
| | - Conglu Guo
- Department of Critical Care Medicine, School of Anesthesiology, Naval Medical University, Shanghai, China
| | - Xinzhu Liu
- Department of Clinical Pharmacy, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zhibin Wang
- Department of Pharmacy, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
- Department of Critical Care Medicine, School of Anesthesiology, Naval Medical University, Shanghai, China
| | - Ye Tu
- Department of Pharmacy, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
9
|
Gao Y, Wang C, Jiang D, An G, Jin F, Zhang J, Han G, Cui C, Jiang P. New insights into the interplay between autophagy and oxidative and endoplasmic reticulum stress in neuronal cell death and survival. Front Cell Dev Biol 2022; 10:994037. [PMID: 36187470 PMCID: PMC9524158 DOI: 10.3389/fcell.2022.994037] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 08/30/2022] [Indexed: 12/03/2022] Open
Abstract
Autophagy is a dynamic process that maintains the normal homeostasis of cells by digesting and degrading aging proteins and damaged organelles. The effect of autophagy on neural tissue is still a matter of debate. Some authors suggest that autophagy has a protective effect on nerve cells, whereas others suggest that autophagy also induces the death of nerve cells and aggravates nerve injury. In mammals, oxidative stress, autophagy and endoplasmic reticulum stress (ERS) constitute important defense mechanisms to help cells adapt to and survive the stress conditions caused by physiological and pathological stimuli. Under many pathophysiological conditions, oxidative stress, autophagy and ERS are integrated and amplified in cells to promote the progress of diseases. Over the past few decades, oxidative stress, autophagy and ERS and their interactions have been a hot topic in biomedical research. In this review, we summarize recent advances in understanding the interactions between oxidative stress, autophagy and ERS in neuronal cell death and survival.
Collapse
Affiliation(s)
- Yahao Gao
- Clinical Medical School, Jining Medical University, Jining, China
| | - Changshui Wang
- Department of Neurosurgery, Affiliated Hospital of Jining Medical University, Jining, China
| | - Di Jiang
- Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Gang An
- Clinical Medical School, Jining Medical University, Jining, China
| | - Feng Jin
- Department of Neurosurgery, Affiliated Hospital of Jining Medical University, Jining, China
| | - Junchen Zhang
- Department of Neurosurgery, Affiliated Hospital of Jining Medical University, Jining, China
| | - Guangkui Han
- Department of Neurosurgery, Affiliated Hospital of Jining Medical University, Jining, China
| | - Changmeng Cui
- Department of Neurosurgery, Affiliated Hospital of Jining Medical University, Jining, China
- *Correspondence: Changmeng Cui, ; Pei Jiang,
| | - Pei Jiang
- Department of Clinical Pharmacy, Jining First People’s Hospital, Jining Medical University, Jining, China
- *Correspondence: Changmeng Cui, ; Pei Jiang,
| |
Collapse
|
10
|
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
|
11
|
Sampath C, Raju AV, Freeman ML, Srinivasan S, Gangula PR. Nrf2 attenuates hyperglycemia-induced nNOS impairment in adult mouse primary enteric neuronal crest cells and normalizes stomach function. Am J Physiol Gastrointest Liver Physiol 2022; 322:G368-G382. [PMID: 35084215 PMCID: PMC8897013 DOI: 10.1152/ajpgi.00323.2021] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Enteric neuronal cells play a vital role in gut motility in humans and experimental rodent models. Patients with diabetes are more vulnerable to gastrointestinal dysfunction due to enteric neuronal degeneration. In this study, we examined the mechanistic role and regulation of nuclear factor-erythroid 2-related factor 2 (Nrf2) in hyperglycemia-induced enteric neuronal cell apoptosis in vitro by using adult mouse primary enteric neuronal crest cells (pENCs). Our data show that hyperglycemia (HG) or inhibition of Nrf2 induces apoptosis by elevating proinflammatory cytokines, reactive oxygen species (ROS) and suppresses neuronal nitric oxide synthase (nNOS-α) via PI3K/Nrf2-mediated signaling. Conversely, treating pENCs with cinnamaldehyde (CNM), a naturally occurring Nrf2 activator, prevented HG-induced apoptosis. These novel data reveal a negative feedback mechanism for GSK-3 activation. To further demonstrate that loss of Nrf2 leads to inflammation, oxidative stress, and reduces nNOS-mediated gastric function, we have used streptozotocin (STZ)-induced diabetic and Nrf2 null female mice. In vivo activation of Nrf2 with CNM (50 mg/kg, 3 days a week, ip) attenuated impaired nitrergic relaxation and delayed gastric emptying (GE) in conventional type 1 diabetic but not in Nrf2 null female mice. Supplementation of CNM normalized diabetes-induced altered gastric antrum protein expression of 1) p-AKT/p-p38MAPK/p-GSK-3β, 2) BH4 (cofactor of nNOS) biosynthesis enzyme GCH-1, 3) nNOSα, 4) TLR4, NF-κB, and 5) inflammatory cytokines (TNF-α, IL-1β, IL-6). We conclude that activation of Nrf2 prevents hyperglycemia-induced apoptosis in pENCs and restores nitrergic-mediated gastric motility and GE in STZ-induced diabetes female mice.NEW & NOTEWORTHY Primary neuronal cell crust (pENCs) in the intestine habitats nNOS and Nrf2, which was suppressed in diabetic gastroparesis. Activation of Nrf2 restored nNOS by suppressing inflammatory markers in pENCs cells. Inhibition of Nrf2 reveals a negative feedback mechanism for the activation of GSK-3. Activation of Nrf2 alleviates STZ-induced delayed gastric emptying and nitrergic relaxation in female mice. Activation of Nrf2 restored impaired gastric BH4 biosynthesis enzyme GCH-1, nNOSα expression thus regulating nitric oxide levels.
Collapse
Affiliation(s)
- Chethan Sampath
- 1Department of ODS and Research, School of Dentistry, Meharry Medical College, Nashville, Tennessee
| | - Abhinav V. Raju
- 2Division of Digestive Diseases, Department of Medicine, Emory University, Atlanta, Georgia
| | - Michael L. Freeman
- 4Department of Radiation Oncology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Shanthi Srinivasan
- 2Division of Digestive Diseases, Department of Medicine, Emory University, Atlanta, Georgia,3Atlanta Veterans Affairs Health Care System, Atlanta, Georgia
| | - Pandu R. Gangula
- 1Department of ODS and Research, School of Dentistry, Meharry Medical College, Nashville, Tennessee
| |
Collapse
|
12
|
Lee PT, Yamamoto FY, Low CF, Loh JY, Chong CM. Gut Immune System and the Implications of Oral-Administered Immunoprophylaxis in Finfish Aquaculture. Front Immunol 2022; 12:773193. [PMID: 34975860 PMCID: PMC8716388 DOI: 10.3389/fimmu.2021.773193] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 11/23/2021] [Indexed: 12/13/2022] Open
Abstract
The gastrointestinal immune system plays an important role in immune homeostasis regulation. It regulates the symbiotic host-microbiome interactions by training and developing the host's innate and adaptive immunity. This interaction plays a vital role in host defence mechanisms and at the same time, balancing the endogenous perturbations of the host immune homeostasis. The fish gastrointestinal immune system is armed with intricate diffused gut-associated lymphoid tissues (GALTs) that establish tolerance toward the enormous commensal gut microbiome while preserving immune responses against the intrusion of enteric pathogens. A comprehensive understanding of the intestinal immune system is a prerequisite for developing an oral vaccine and immunostimulants in aquaculture, particularly in cultured fish species. In this review, we outline the remarkable features of gut immunity and the essential components of gut-associated lymphoid tissue. The mechanistic principles underlying the antigen absorption and uptake through the intestinal epithelial, and the subsequent immune activation through a series of molecular events are reviewed. The emphasis is on the significance of gut immunity in oral administration of immunoprophylactics, and the different potential adjuvants that circumvent intestinal immune tolerance. Comprehension of the intestinal immune system is pivotal for developing effective fish vaccines that can be delivered orally, which is less labour-intensive and could improve fish health and facilitate disease management in the aquaculture industry.
Collapse
Affiliation(s)
- Po-Tsang Lee
- Department of Aquaculture, National Taiwan Ocean University, Keelung, Taiwan
| | - Fernando Y Yamamoto
- Thad Cochran National Warmwater Aquaculture Center, Mississippi Agriculture and Forestry Experiment Station, Mississippi State University, Stoneville, MS, United States
| | - Chen-Fei Low
- Institute of Systems Biology, Universiti Kebangsaan Malaysia, Bangi, Malaysia
| | - Jiun-Yan Loh
- Centre of Research for Advanced Aquaculture (CORAA), UCSI University, Cheras, Malaysia
| | - Chou-Min Chong
- Aquatic Animal Health and Therapeutics Laboratory (AquaHealth), Institute of Bioscience, Universiti Putra Malaysia, Serdang, Malaysia
| |
Collapse
|
13
|
Hu K, Xiao L, Li L, Shen Y, Yang Y, Huang J, Wang Y, Zhang L, Wen S, Tang L. The mitochondria-targeting antioxidant MitoQ alleviated lipopolysaccharide/ d-galactosamine-induced acute liver injury in mice. Immunol Lett 2021; 240:24-30. [PMID: 34525396 DOI: 10.1016/j.imlet.2021.09.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 08/02/2021] [Accepted: 09/10/2021] [Indexed: 01/06/2023]
Abstract
The mitochondria are the primary source of reactive oxygen species (ROS) under pathological condition, but the significance of mitochondrial ROS in the development of Lipopolysaccharide (LPS)/D-galactosamine (D-Gal)-induced acute liver injury remains unclear. In the present study, the level of mitochondrial ROS in LPS/D-Gal has been determined by MitoSox staining and the potential roles of mitochondrial ROS in LPS/D-Gal-induced liver injury have been investigated by using the mitochondria-targeting antioxidant MitoQ. The results indicated that LPS/D-Gal exposure induced the generation of mitochondrial ROS while treatment with MitoQ reduced the level of mitochondrial ROS. Treatment with MitoQ ameliorated LPS/D-Gal-induced histopathologic abnormalities, suppressed the elevation of AST and ALT, and increased the survival rate of the experimental animals. Treatment with MitoQ also suppressed LPS/D-Gal-induced production of tumor necrosis factor α (TNF-α), inhibited the activities of caspase-3, caspase-8 and caspase-9, decreased the level of cleaved caspase-3 and reduced the counts of TUNEL positive cells. These results indicate that mitochondrial ROS is involved in the development of LPS-induced acute liver injury and the mitochondria-targeting antioxidant MitoQ might have potential value for the treatment of inflammation-based acute liver injury.
Collapse
Affiliation(s)
- Kai Hu
- Laboratory of Stem cell and Tissue Engineering, Chongqing Medical University, Chongqing, China; Department of Histology and Embryology, Chongqing Medical University, Chongqing, China
| | - Lidan Xiao
- Department of Pathophysiology, Chongqing Medical University, Chongqing, China
| | - Longjiang Li
- Department of Pathophysiology, Chongqing Medical University, Chongqing, China
| | - Yi Shen
- Department of Pathophysiology, Chongqing Medical University, Chongqing, China
| | - Yongqiang Yang
- Department of Pathophysiology, Chongqing Medical University, Chongqing, China
| | - Jiayi Huang
- Department of Pathophysiology, Chongqing Medical University, Chongqing, China
| | - Yaping Wang
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, China
| | - Li Zhang
- Department of Pathophysiology, Chongqing Medical University, Chongqing, China
| | - Sha Wen
- Department of General medicine, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Li Tang
- Laboratory of Stem cell and Tissue Engineering, Chongqing Medical University, Chongqing, China; Department of Pathophysiology, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
14
|
El-Sadek HM, Al-Shorbagy MY, Awny MM, Abdallah DM, El-Abhar HS. Pentoxifylline treatment alleviates kidney ischemia/reperfusion injury: Novel involvement of galectin-3 and ASK-1/JNK & ERK1/2/NF-κB/HMGB-1 trajectories. J Pharmacol Sci 2021; 146:136-148. [PMID: 34030796 DOI: 10.1016/j.jphs.2021.03.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 03/24/2021] [Accepted: 03/29/2021] [Indexed: 02/07/2023] Open
Abstract
Despite the documented renoprotective effect of pentoxifylline (PTX), a non-selective phosphodiesterase-4 inhibitor, the studies appraised only its anti-inflammatory/-oxidant/-apoptotic capacities without assessment of the possible involved trajectories. Here, we evaluated the potential role of galectin-3 and the ASK-1/NF-κB p65 signaling pathway with its upstream/downstream signals in an attempt to unveil part of the cascades involved in the renotherapeutic effect using a renal bilateral ischemia/reperfusion (I/R) model. Rats were randomized into sham-operated, renal I/R (45 min/72 h) and I/R + PTX (100 mg/kg; p.o). Post-treatment with PTX improved renal function and abated serum levels of cystatin C, creatinine, BUN and renal KIM-1 content, effects that were reflected on an improvement of the I/R-induced renal histological changes. On the molecular level, PTX reduced renal contents of galectin-3, ASK-1 with its downstream molecule JNK and ERK1/2, as well as NF-κB p65 and HMGB1. This inhibitory effect extended also to suppress neutrophil infiltration, evidenced by diminishing ICAM-1 and MPO, as well as inflammatory cytokines (TNF-α/IL-18), oxidative stress (MDA/TAC), and caspase-3. The PTX novel renotherapeutic effect involved in part the inhibition of galectin-3 and ASK-1/JNK and ERK1/2/NF-κB/HMGB-1 trajectories to mitigate renal I/R injury and to provide basis for its anti-inflammatory, antioxidant, and anti-apoptotic impacts.
Collapse
Affiliation(s)
- Hagar M El-Sadek
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, October 6 University, Giza, 12585, Egypt
| | - Muhammad Y Al-Shorbagy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt; Department of Pharmaceutical Sciences, College of Pharmacy, Gulf Medical University, Ajman, 4184, United Arab Emirates
| | - Magdy M Awny
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, October 6 University, Giza, 12585, Egypt
| | - Dalaal M Abdallah
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt.
| | - Hanan S El-Abhar
- Department of Pharmacology, Toxicology and Biochemistry, Faculty of Pharmaceutical Sciences and Pharmaceutical Industries, Future University in Egypt, Cairo, 84518, Egypt
| |
Collapse
|
15
|
Superoxide Dismutase 3-Transduced Mesenchymal Stem Cells Preserve Epithelial Tight Junction Barrier in Murine Colitis and Attenuate Inflammatory Damage in Epithelial Organoids. Int J Mol Sci 2021; 22:ijms22126431. [PMID: 34208517 PMCID: PMC8233984 DOI: 10.3390/ijms22126431] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 06/04/2021] [Accepted: 06/11/2021] [Indexed: 01/08/2023] Open
Abstract
Superoxide dismutase 3 (SOD3), also known as extracellular superoxide dismutase, is an enzyme that scavenges reactive oxygen species (ROS). It has been reported that SOD3 exerts anti-inflammatory abilities in several immune disorders. However, the effect of SOD3 and the underlying mechanism in inflammatory bowel disease (IBD) have not been uncovered. Therefore, in the present study, we investigated whether SOD3 can protect intestinal cells or organoids from inflammation-mediated epithelial damage. Cells or mice were treated with SOD3 protein or SOD3-transduced mesenchymal stem cells (MSCs). Caco-2 cells or intestinal organoids stimulated with pro-inflammatory cytokines were used to evaluate the protective effect of SOD3 on epithelial junctional integrity. Dextran sulfate sodium (DSS)-induced colitis mice received SOD3 or SOD3-transduced MSCs (SOD3-MSCs), and were assessed for severity of disease and junctional protein expression. The activation of the mitogen-activated protein kinase (MAPK) pathway and elevated expression of cytokine-encoding genes decreased in TNF-α-treated Caco-2 cells or DSS-induced colitis mice when treated with SOD3 or SOD3-MSCs. Moreover, the SOD3 supply preserved the expression of tight junction (ZO-1, occludin) or adherence junction (E-cadherin) proteins when inflammation was induced. SOD3 also exerted a protective effect against cytokine- or ROS-mediated damage to intestinal organoids. These results indicate that SOD3 can effectively alleviate enteritis symptoms by maintaining the integrity of epithelial junctions and regulating inflammatory- and oxidative stress.
Collapse
|
16
|
Weis VG, Deal AC, Mekkey G, Clouse C, Gaffley M, Whitaker E, Peeler CB, Weis JA, Schwartz MZ, Atala A. Human placental-derived stem cell therapy ameliorates experimental necrotizing enterocolitis. Am J Physiol Gastrointest Liver Physiol 2021; 320:G658-G674. [PMID: 33566727 PMCID: PMC8238163 DOI: 10.1152/ajpgi.00369.2020] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 02/08/2021] [Accepted: 02/08/2021] [Indexed: 02/06/2023]
Abstract
Necrotizing enterocolitis (NEC), a life-threatening intestinal disease, is becoming a larger proportionate cause of morbidity and mortality in premature infants. To date, therapeutic options remain elusive. Based on recent cell therapy studies, we investigated the effect of a human placental-derived stem cell (hPSC) therapy on intestinal damage in an experimental NEC rat pup model. NEC was induced in newborn Sprague-Dawley rat pups for 4 days via formula feeding, hypoxia, and LPS. NEC pups received intraperitoneal (ip) injections of either saline or hPSC (NEC-hPSC) at 32 and 56 h into NEC induction. At 4 days, intestinal macroscopic and histological damage, epithelial cell composition, and inflammatory marker expression of the ileum were assessed. Breastfed (BF) littermates were used as controls. NEC pups developed significant bowel dilation and fragility in the ileum. Further, NEC induced loss of normal villi-crypt morphology, disruption of epithelial proliferation and apoptosis, and loss of critical progenitor/stem cell and Paneth cell populations in the crypt. hPSC treatment improved macroscopic intestinal health with reduced ileal dilation and fragility. Histologically, hPSC administration had a significant reparative effect on the villi-crypt morphology and epithelium. In addition to a trend of decreased inflammatory marker expression, hPSC-NEC pups had increased epithelial proliferation and decreased apoptosis when compared with NEC littermates. Further, the intestinal stem cell and crypt niche that include Paneth cells, SOX9+ cells, and LGR5+ stem cells were restored with hPSC therapy. Together, these data demonstrate hPSC can promote epithelial healing of NEC intestinal damage.NEW & NOTEWORTHY These studies demonstrate a human placental-derived stem cell (hPSC) therapeutic strategy for necrotizing enterocolitis (NEC). In an experimental model of NEC, hPSC administration improved macroscopic intestinal health, ameliorated epithelial morphology, and supported the intestinal stem cell niche. Our data suggest that hPSC are a potential therapeutic approach to attenuate established intestinal NEC damage. Further, we show hPSC are a novel research tool that can be utilized to elucidate critical neonatal repair mechanisms to overcome NEC.
Collapse
Affiliation(s)
- Victoria G Weis
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, North Carolina
| | - Anna C Deal
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, North Carolina
| | - Gehad Mekkey
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, North Carolina
- Faculty of Science, Zagazig University, Zagazig, Egypt
| | - Cara Clouse
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, North Carolina
| | - Michaela Gaffley
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, North Carolina
- General Surgery, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Emily Whitaker
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, North Carolina
| | - Cole B Peeler
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, North Carolina
- School of Biomedical Engineering and Sciences, Virginia Tech-Wake Forest University, Blacksburg, Virginia
| | - Jared A Weis
- School of Biomedical Engineering and Sciences, Virginia Tech-Wake Forest University, Blacksburg, Virginia
- Department of Biomedical Engineering, Wake Forest School of Medicine, Winston-Salem, North Carolina
- Comprehensive Cancer Center, Wake Forest Baptist Medical Center, Winston-Salem, North Carolina
| | - Marshall Z Schwartz
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, North Carolina
| | - Anthony Atala
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, North Carolina
| |
Collapse
|
17
|
Li HH, Bao LS, Deng SM, Liu L, Cheng J, Chen X, Pan YX, Zhang JS, Chu WY. Investigation of Proteus vulgaris and Elizabethkingia meningoseptica invasion on muscle oxidative stress and autophagy in Chinese soft-shelled turtle (Pelodiscus sinensis). Sci Rep 2021; 11:3657. [PMID: 33574492 PMCID: PMC7878920 DOI: 10.1038/s41598-021-83388-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 02/02/2021] [Indexed: 01/30/2023] Open
Abstract
Muscle is an important structural tissue in aquatic animals and it is susceptible to bacterial and fungal infection, which could affect flesh quality and health. In this study, Chinese soft-shelled turtles were artificially infected with two pathogens, Proteus vulgaris and Elizabethkingia meningoseptica and the effects on muscle nutritional characteristics, oxidative stress and autophagy were assayed. Upon infection, the muscle nutritional composition and muscle fiber structure were notably influenced. Meanwhile, the mRNA expression of Nrf2 was down-regulated and Keap1 up-regulated, thus resulting in a decrease in antioxidant capacity and oxidative stress. However, with N-acetylcysteine treatment, the level of oxidative stress was decreased, accompanied by significant increases in antioxidant enzyme activities and the mRNA levels of SOD, CAT, GSTCD, and GSTO1. Interestingly, there was a significant increase in autophagy in the muscle tissue after the pathogen infection, but this increase could be reduced by N-acetylcysteine treatment. Our findings suggest that muscle nutritional characteristics were dramatically changed after pathogen infection, and oxidative stress and autophagy were induced by pathogen infection. However, N-acetylcysteine treatment could compromise the process perhaps by decreasing the ROS level and regulating Nrf2-antioxidant signaling pathways.
Collapse
Affiliation(s)
- Hong-Hui Li
- Hunan Provincial Key Laboratory of Nutrition and Quality Control of Aquatic Animals, College of Biological and Environmental Engineering, Changsha University, Changsha, China
- College of Chemistry and Food Engineering, Changsha University of Science & Technology, Changsha, China
| | - Ling-Sheng Bao
- Hunan Provincial Key Laboratory of Nutrition and Quality Control of Aquatic Animals, College of Biological and Environmental Engineering, Changsha University, Changsha, China
| | | | - Li Liu
- Hunan Fisheries Science Institute, Changsha, China
| | - Jia Cheng
- Hunan Provincial Key Laboratory of Nutrition and Quality Control of Aquatic Animals, College of Biological and Environmental Engineering, Changsha University, Changsha, China
| | - Xiao Chen
- Hunan Provincial Key Laboratory of Nutrition and Quality Control of Aquatic Animals, College of Biological and Environmental Engineering, Changsha University, Changsha, China
| | - Ya-Xiong Pan
- Hunan Provincial Key Laboratory of Nutrition and Quality Control of Aquatic Animals, College of Biological and Environmental Engineering, Changsha University, Changsha, China
| | - Jian-She Zhang
- Hunan Provincial Key Laboratory of Nutrition and Quality Control of Aquatic Animals, College of Biological and Environmental Engineering, Changsha University, Changsha, China.
| | - Wu-Ying Chu
- Hunan Provincial Key Laboratory of Nutrition and Quality Control of Aquatic Animals, College of Biological and Environmental Engineering, Changsha University, Changsha, China.
| |
Collapse
|
18
|
Sialylated human milk oligosaccharides prevent intestinal inflammation by inhibiting toll like receptor 4/NLRP3 inflammasome pathway in necrotizing enterocolitis rats. Nutr Metab (Lond) 2021; 18:5. [PMID: 33407596 PMCID: PMC7789326 DOI: 10.1186/s12986-020-00534-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 12/11/2020] [Indexed: 12/21/2022] Open
Abstract
Background Necrotizing enterocolitis (NEC) remains a fatal gastrointestinal disorder in neonates and has very limited therapeutic options. Sialylated human milk oligosaccharides (SHMOs) improve pathological changes in experimental NEC models. The objectives of this study were to investigate the involvement of NLRP3 inflammasome in NEC pathology and to explore the effects of SHMOs on toll-like receptor 4 (TLR4)/nuclear factor κB (NF-κB)/NLRP3 inflammatory pathway in experimental NEC. Methods The intestinal-tissue segments were collected from NEC infants, NLRP3 and caspase-1 positive cell were examined by immunohistochemistry. Newborn rats were hand-fed with formula containing or non-containing SHMOs (1500 mg/L) and exposed to hypoxia/cold stress to induce experimental NEC. The NEC pathological scores were evaluated; ileum protein expression of membrane TLR4 (mTLR4), inhibitor κB-α (IκB-α), NF-κB p65 subunit and phospho-NF-κB p65, as well as NLRP3 and caspase-1 were analyzed; ileum concentrations of interleukin-1β, interleukin-6, tumor necrosis factor-α (TNF-α) were also measured. Human colon epithelial Caco-2 cells were pre-treated with or without SHMOs and stimulated with TLR4 activator, lipopolysaccharide. Cell viabilities, mitochondrial membrane potential and supernatant matrix metalloprotease 2 (MMP-2) activities were analyzed. Results Increased frequencies of NLRP3 and caspase-1 positive cells were found in the lamina propria of damaged intestinal area of NEC neonates. SHMOs supplementation reduced NEC incidence and pathological damage scores of rats challenged with hypoxia/cold stress. Accumulation of interleukin-1β, interleukin-6 and TNF-α in NEC group were attenuated in SHMOs + NEC group. Protein expression of mTLR4, NLRP3 and caspase-1 were elevated, cytoplasmic IκB-α were reduced, nuclear phospho-NF-κB p65 were increased in the ileum of NEC rats. SHMOs supplementation ameliorated the elevation of mTLR4, NLRP3 and caspase-1, restored IκB-α in the cytoplasmic fraction and reduced phospho-NF-κB p65 in the nuclear fraction in the ileum of NEC rats. SHMOs pre-treatment improved Caco-2 cell viability, mitigated loss of mitochondrial membrane potential and modulated MMP-2 activities in the presence of lipopolysaccharide in-vitro. Conclusions This study provided clinical evidence of involvement of NLRP3 inflammasome in NEC pathology, and demonstrated the protective actions of SHMOs might be owing to the suppression of TLR4/NF-κB/NLRP3-mediated inflammation in NEC.
Collapse
|
19
|
Hou S, Yang X, Tong Y, Yang Y, Chen Q, Wan B, Wei R, Wang Y, Zhang Y, Kong B, Huang J, Chen Y, Lu T, Hu Q, Du D. Structure-based discovery of 1H-indole-2-carboxamide derivatives as potent ASK1 inhibitors for potential treatment of ulcerative colitis. Eur J Med Chem 2020; 211:113114. [PMID: 33360793 DOI: 10.1016/j.ejmech.2020.113114] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 12/16/2020] [Accepted: 12/16/2020] [Indexed: 01/09/2023]
Abstract
Apoptosis signal-regulating kinase 1 (ASK1), a member of the mitogen-activated protein kinase (MAPK) family, is implicated in many human diseases. Here, we describe the structural optimization of hit compound 7 and conduct further structure-activity relationship (SAR) studies that result in the development of compound 19 with a novel indole-2-carboxamide hinge scaffold. Compound 19 displays potent anti-ASK1 kinase activity and stronger inhibitory effect on ASK1 in AP1-HEK293 cells than previously described ASK1 inhibitor GS-4997. Besides improved in vitro activity, compound 19 also exhibits an appropriate in vivo PK profile. In a dextran sulfate sodium (DSS)-induced mouse model of ulcerative colitis (UC), compound 19 shows significant anti-UC efficacy and markedly attenuates DSS-induced body weight loss, colonic shortening, elevation in disease activity index (DAI) and inflammatory cell infiltration in colon tissues. Mechanistically, compound 19 represses the phosphorylation of ASK1-p38/JNK signaling pathways and suppresses the overexpression of inflammatory cytokines. Together, these findings suggest that ASK1 inhibitors can potentially be used as a therapeutic strategy for UC.
Collapse
Affiliation(s)
- Shaohua Hou
- School of Sciences, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, PR China
| | - Xiping Yang
- School of Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, PR China
| | - Yu Tong
- School of Sciences, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, PR China
| | - Yuejing Yang
- School of Sciences, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, PR China
| | - Quanwei Chen
- School of Sciences, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, PR China
| | - Boheng Wan
- School of Sciences, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, PR China
| | - Ran Wei
- School of Sciences, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, PR China
| | - Yuchen Wang
- School of Sciences, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, PR China
| | - Yanmin Zhang
- School of Sciences, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, PR China
| | - Bo Kong
- School of Sciences, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, PR China
| | - Jianhang Huang
- School of Sciences, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, PR China
| | - Yadong Chen
- School of Sciences, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, PR China; State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, PR China
| | - Tao Lu
- School of Sciences, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, PR China; State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, PR China.
| | - Qinghua Hu
- School of Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, PR China.
| | - Ding Du
- School of Sciences, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, PR China.
| |
Collapse
|
20
|
Abstract
PURPOSE Intestinal absorption in premature infants occurs via direct epithelial cellular endocytosis and degradation by intracellular lysosomes. Autophagy is a mechanism by which cytoplasmic organelles contribute to lysosomal degradation. However, excessive autophagy can lead to cell death. The purpose of this study was to investigate whether autophagy and endocytosis are present in the small intestinal mucosa during experimental necrotizing enterocolitis (NEC). METHODS NEC was induced by gavage feeding of hyperosmolar formula, lipopolysaccharide and hypoxia between P5 and P9 (ethical approval 44032). Breastfed mice were used as control. Distal ileum was harvested on P5, P7 and P9 and analyzed for intestinal epithelial cellular morphology as well as autophagy/lysosomal activity, and cell death. Groups were compared using Student's t test. RESULTS During NEC, giant lysosomes were present in the intestinal villi, with some exceeding their degradation ability leading to their rupture. The NEC group had significantly increased inflammation and autophagy activity, decreased lysosome activity, and increased apoptosis compared to control. CONCLUSIONS NEC induction causes excessive autophagy and endocytosis leading to lysosomal overloading, lysosomal membrane permeabilization and rupture which results in cell death. These novel findings may help to clarify the pathogenesis of NEC. Reduction of lysosome overload and assisting in their degradation capability may reduce the burden of NEC.
Collapse
|
21
|
Chadha S, Behl T, Bungau S, Kumar A, Kaur R, Venkatachalam T, Gupta A, Kandhwal M, Chandel D. Focus on the Multimodal Role of Autophagy in Rheumatoid Arthritis. Inflammation 2020; 44:1-12. [PMID: 32954452 DOI: 10.1007/s10753-020-01324-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 08/10/2020] [Accepted: 08/17/2020] [Indexed: 12/24/2022]
Abstract
Autophagy exerts its dual role in eukaryotic cells and exerts its cytoprotective action through degradation mechanism and by regulating catabolic processes which results in elimination of pathogens. Under suitable conditions, autophagy is associated with recycling of cytoplasmic components which causes regeneration of energy whereas deregulated autophagy exerts its implicated role in development and pathogenesis of auto-immune diseases such as rheumatoid arthritis. The immune, innate, and adaptive responses are regulated through the development, proliferation, and growth of lymphocytes. Such innate and adaptive responses can act as mediator of arthritis; along with this, stimulation of osteoclast-mediated bone resorption takes place via transferring citrullinated peptides towards MHC (major histocompatibility complex) compartments, thereby resulting in degradation of bone. Processes such as apoptosis resistance are also regulated through autophagy. In this review, the current knowledge based on role of autophagy in pathogenesis of rheumatoid arthritis is summarized along with proteins associated.
Collapse
Affiliation(s)
- Swati Chadha
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India.
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, Oradea, Romania.
| | - Arun Kumar
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Rajwinder Kaur
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | | | - Amit Gupta
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Mimansa Kandhwal
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Deepak Chandel
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| |
Collapse
|
22
|
Stepien KM, Roncaroli F, Turton N, Hendriksz CJ, Roberts M, Heaton RA, Hargreaves I. Mechanisms of Mitochondrial Dysfunction in Lysosomal Storage Disorders: A Review. J Clin Med 2020; 9:jcm9082596. [PMID: 32796538 PMCID: PMC7463786 DOI: 10.3390/jcm9082596] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 08/04/2020] [Accepted: 08/06/2020] [Indexed: 12/12/2022] Open
Abstract
Mitochondrial dysfunction is emerging as an important contributory factor to the pathophysiology of lysosomal storage disorders (LSDs). The cause of mitochondrial dysfunction in LSDs appears to be multifactorial, although impaired mitophagy and oxidative stress appear to be common inhibitory mechanisms shared amongst these heterogeneous disorders. Once impaired, dysfunctional mitochondria may impact upon the function of the lysosome by the generation of reactive oxygen species as well as depriving the lysosome of ATP which is required by the V-ATPase proton pump to maintain the acidity of the lumen. Given the reported evidence of mitochondrial dysfunction in LSDs together with the important symbiotic relationship between these two organelles, therapeutic strategies targeting both lysosome and mitochondrial dysfunction may be an important consideration in the treatment of LSDs. In this review we examine the putative mechanisms that may be responsible for mitochondrial dysfunction in reported LSDs which will be supplemented with morphological and clinical information.
Collapse
Affiliation(s)
- Karolina M. Stepien
- Adult Inherited Metabolic Diseases, Salford Royal NHS Foundation Trust, Salford M6 8HD, UK
- Correspondence:
| | - Federico Roncaroli
- Division of Neuroscience and Experimental Psychology, School of Biology, Medicine and Health, University of Manchester and Manchester Centre for Clinical Neuroscience, Salford Royal NHS Foundation Trust, Salford M6 8HD, UK;
| | - Nadia Turton
- School of Pharmacy, Liverpool John Moore University, Byrom Street, Liverpool L3 3AF, UK; (N.T.); (R.A.H.); (I.H.)
| | - Christian J. Hendriksz
- Paediatrics and Child Health, Steve Biko Academic Unit, University of Pretoria, 0002 Pretoria, South Africa;
| | - Mark Roberts
- Neurology Department, Salford Royal NHS Foundation Trust, Salford M6 8HD, UK;
| | - Robert A. Heaton
- School of Pharmacy, Liverpool John Moore University, Byrom Street, Liverpool L3 3AF, UK; (N.T.); (R.A.H.); (I.H.)
| | - Iain Hargreaves
- School of Pharmacy, Liverpool John Moore University, Byrom Street, Liverpool L3 3AF, UK; (N.T.); (R.A.H.); (I.H.)
| |
Collapse
|
23
|
He-Yang J, Zhang W, Liu J, Xue P, Zhou X. Human breast milk oligosaccharides attenuate necrotizing enterocolitis in rats by suppressing mast cell accumulation, DPPI activity and TLR4 expression in ileum tissue, and regulating mitochondrial damage of Caco-2 cells. Int Immunopharmacol 2020; 88:106881. [PMID: 32795899 DOI: 10.1016/j.intimp.2020.106881] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 08/05/2020] [Accepted: 08/05/2020] [Indexed: 12/09/2022]
Abstract
Necrotizing enterocolitis (NEC), a devastating infant disease characterized by severe intestinal necrosis, its pathogenesis is poorly understood, but appears to be multifactorial and highly associated with immaturity of gastrointestinal tract and immature innate-immune system. Breast-milk is effective strategy to protect infants against NEC. This study is using a NEC rat model to investigate the pathological mechanism of NEC involved intestinal-damages, and the therapeutic mechanism of sialylated human milk oligosaccharides (SHMOs) on NEC rats; also using cell model to investigate the effects of SHMOs on colon-epithelial cells (Caco-2) in-vitro. Extraction and characterization of SHMOs from breast milk, establishment of a NEC rat model, histopathological analysis and mast cell accounting of the terminal ileum were taken; The levels of DPPI, TLR4, IL-6, TNF-α, MMP-2/9 and glutathione were measured using various methods. Caco-2 cells were pre-treated with SHMOs and cultured with LPS, histamine, chymase or DPPI, cell viabilities and mitochondrial membrane potential were examined; flow cytometry was used to detect cell cycle. The accumulation of mast cells was found in the ileum of NEC rats, but prohibited by SHMOs treatment; the increased levels of TLR4, DPPI, IL-6, TNF-α, MMP-2/9 in NEC ileum were suppressed by SHMOs in-vivo. SHMOs prevented Caco-2 cells from LPS, histamine, chymase induced damages by surviving cell viability, regulating G0/G1 and S phase in cell cycles, and increasing mitochondrial membrane potential. These findings provide a new insight into the pharmacological mechanism of SHMOs treatment for NEC and suggest that SHMOs needs well attention for therapeutic aims.
Collapse
Affiliation(s)
- Jingqiu He-Yang
- The School of Pharmacy, Changzhou University, Jiangsu 213164, China
| | - Wenting Zhang
- The School of Pharmacy, Changzhou University, Jiangsu 213164, China; Department of Pharmacy, Affiliated Changzhou Children's Hospital of Nantong University, Yanling Road 468, Changzhou 213003, China
| | - Jie Liu
- The School of Pharmacy, Changzhou University, Jiangsu 213164, China
| | - Peng Xue
- Department of Pharmacy, Affiliated Changzhou Children's Hospital of Nantong University, Yanling Road 468, Changzhou 213003, China
| | - Xiaoying Zhou
- The School of Pharmacy, Changzhou University, Jiangsu 213164, China; The School of Medicine, the University of Southampton, Southampton SO16 6YD, UK.
| |
Collapse
|
24
|
Panov J, Simchi L, Feuermann Y, Kaphzan H. Bioinformatics Analyses of the Transcriptome Reveal Ube3a-Dependent Effects on Mitochondrial-Related Pathways. Int J Mol Sci 2020; 21:ijms21114156. [PMID: 32532103 PMCID: PMC7312912 DOI: 10.3390/ijms21114156] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 06/07/2020] [Accepted: 06/08/2020] [Indexed: 12/14/2022] Open
Abstract
The UBE3A gene encodes the ubiquitin E3-ligase protein, UBE3A, which is implicated in severe neurodevelopmental disorders. Lack of UBE3A expression results in Angelman syndrome, while UBE3A overexpression, due to genomic 15q duplication, results in autism. The cellular roles of UBE3A are not fully understood, yet a growing body of evidence indicates that these disorders involve mitochondrial dysfunction and increased oxidative stress. We utilized bioinformatics approaches to delineate the effects of murine Ube3a deletion on the expression of mitochondrial-related genes and pathways. For this, we generated an mRNA sequencing dataset from mouse embryonic fibroblasts (MEFs) in which both alleles of Ube3a gene were deleted and their wild-type controls. Since oxidative stress and mitochondrial dysregulation might not be exhibited in the resting baseline state, we also activated mitochondrial functioning in the cells of these two genotypes using TNFα application. Transcriptomes of the four groups of MEFs, Ube3a+/+ and Ube3a-/-, with or without the application of TNFα, were analyzed using various bioinformatics tools and machine learning approaches. Our results indicate that Ube3a deletion affects the gene expression profiles of mitochondrial-associated pathways. We further confirmed these results by analyzing other publicly available human transcriptome datasets of Angelman syndrome and 15q duplication syndrome.
Collapse
|
25
|
Progressive Metabolic Dysfunction and Nutritional Variability Precedes Necrotizing Enterocolitis. Nutrients 2020; 12:nu12051275. [PMID: 32365850 PMCID: PMC7281969 DOI: 10.3390/nu12051275] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 04/27/2020] [Accepted: 04/27/2020] [Indexed: 12/16/2022] Open
Abstract
Necrotizing Enterocolitis (NEC) is associated with prematurity, enteral feedings, and enteral dysbiosis. Accordingly, we hypothesized that along with nutritional variability, metabolic dysfunction would be associated with NEC onset. Methods: We queried a multicenter longitudinal database that included 995 preterm infants (<32 weeks gestation) and included 73 cases of NEC. Dried blood spot samples were obtained on day of life 1, 7, 28, and 42. Metabolite data from each time point included 72 amino acid (AA) and acylcarnitine (AC) measures. Nutrition data were averaged at each of the same time points. Odds ratios and 95% confidence intervals were calculated using samples obtained prior to NEC diagnosis and adjusted for potential confounding variables. Nutritional and metabolic data were plotted longitudinally to determine relationship to NEC onset. Results: Day 1 analyte levels of alanine, phenylalanine, free carnitine, C16, arginine, C14:1/C16, and citrulline/phenylalanine were associated with the subsequent development of NEC. Over time, differences in individual analyte levels associated with NEC onset shifted from predominantly AAs at birth to predominantly ACs by day 42. Subjects who developed NEC received significantly lower weight-adjusted total calories (p < 0.001) overall, a trend that emerged by day of life 7 (p = 0.020), and persisted until day of life 28 (p < 0.001) and 42 (p < 0.001). Conclusion: Premature infants demonstrate metabolic differences at birth. Metabolite abnormalities progress in parallel to significant differences in nutritional delivery signifying metabolic dysfunction in premature newborns prior to NEC onset. These observations provide new insights to potential contributing pathophysiology of NEC and opportunity for clinical care-based prevention.
Collapse
|
26
|
Nabil M, El Demellawy MA, Mahmoud MF, Mahmoud AAA. Prolonged overnutrition with fructose or fat induces metabolic derangements in rats by disrupting the crosstalk between the hypothalamus and periphery: Possible amelioration with fenofibrate. Eur J Pharmacol 2020; 879:173136. [PMID: 32360834 DOI: 10.1016/j.ejphar.2020.173136] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 04/14/2020] [Accepted: 04/20/2020] [Indexed: 11/15/2022]
Abstract
Metabolic Syndrome (MetS) increases the risk of developing type 2 diabetes mellitus and cardiovascular complications. The crosstalk between the hypothalamus and periphery is vital for regulating food intake and energy homeostasis. However, it is impaired during MetS. The present study aimed to compare the distinct central and peripheral metabolic derangements induced by a high-fructose drink or high-fat diet, as well as the possible intervention by fenofibrate. Rats were divided into five groups: standard chow diet (SCD) group, high-fructose group (FR), high-fat group (HF), FR plus fenofibrate group (FR-F), and HF plus fenofibrate group (HF-F). FR and HF groups showed hyperglycemia, hyperinsulinemia, hypertriglyceridemia, hyperleptinemia, steatosis, and adipocyte hypertrophy. This was associated with elevated circulating levels of proinflammatory cytokines and free fatty acids (FFAs). The latter mediators are involved in the hypothalamic inflammation and dysregulation of signaling cascades that control food intake and glucose homeostasis. The effects were more pronounced in the HF group than FR group, which were matched with the observed higher levels of plasma FFAs and cytokines. Fenofibrate administration improved not only the peripheral metabolic disturbances, but also the central disturbances associated with insulin resistance induced by FR or HF diet. This study sheds light on the pivotal role of the hypothalamus in diet-induced MetS. Furthermore, the study suggests the utmost importance of developing a standardized model of metabolic syndrome in place of the great diversity between available models, which can induce different effects and negatively impact the validity of prospective studies.
Collapse
Affiliation(s)
- Mohamed Nabil
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt; Pharmaceutical and Fermentation Industries Development Center (PFIDC), City of Scientific Research and Technological Applications (SRTA-City), New Borg El-Arab, Alexandria, 21934, Egypt
| | - Maha A El Demellawy
- Department of Medical Biotechnology, Genetic Engineering & Biotechnology Research Institute (GEBRI), City of Scientific Research and Technological Applications (SRTA-City), New Borg El-Arab, Alexandria, 21934, Egypt
| | - Mona F Mahmoud
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt
| | - Amr A A Mahmoud
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt.
| |
Collapse
|
27
|
Zhu X, Cui N, Yu L, Cheng P, Cui M, Zhu X, Wang J. Potential role of endoplasmic reticulum stress is involved in the protection of fish oil on neonatal rats with necrotizing enterocolitis. Sci Rep 2020; 10:6448. [PMID: 32296092 PMCID: PMC7160196 DOI: 10.1038/s41598-020-63309-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 03/09/2020] [Indexed: 11/09/2022] Open
Abstract
Neonatal necrotizing enterocolitis (NEC) is a serious gastrointestinal disease with high death rate in premature infants. Fish oil (FO) and its constituents have been shown to ameliorate intestinal inflammation and mucosal damage. However, the underlying mechanism of action is not known. In the present study, we divided Sprague-Dawley rats into three groups: control group, NEC model group, and FO pre-feeding+NEC model group. Briefly, one week before NEC modeling, in addition to being fed with milk, the FO pre-feeding+NEC modeling group was fed with FO, the NEC group was fed with saline, and the control group was only inserted a gastric-tube for 7 days. Subsequently, histological assay, Western blot, and ELISA were performed. Pretreatment with FO attenuated the NEC symptoms, alleviated intestinal pathological injury, and decreased the expressions of pro-inflammatory cytokines such as interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α). Furthermore, pretreatment with FO reduced the expressions of endoplasmic reticulum stress (ERS) related proteins, caspase-12, and glucose-regulated protein 78 (GRP78). In addition, intestinal histopathological scores showed a significant positive correlation with intestinal expressions of IL-6, TNF-α, and caspase-12. Collectively, these results indicate that ERS pathway might be involved in the effect of FO in alleviating intestinal mucosal inflammation and injury in rats with NEC.
Collapse
Affiliation(s)
- Xiaoli Zhu
- Department of Intervention, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Ningxun Cui
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou, 215025, China
| | - Lingling Yu
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou, 215025, China
| | - Ping Cheng
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou, 215025, China
| | - Mingling Cui
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou, 215025, China
| | - Xueping Zhu
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou, 215025, China.
| | - Jian Wang
- Department of Neonatology Surgery, Children's Hospital of Soochow University, Suzhou, 215025, Jiangsu, China.
| |
Collapse
|
28
|
Zhang F, Zhu K, Liu L, Zhu J, Li J, Zhang P, Hu Z, Yuan Y. IRF2-INPP4B axis inhibits apoptosis of acute myeloid leukaemia cells via regulating T helper 1/2 cell differentiation. Cell Biochem Funct 2020; 38:582-590. [PMID: 32115737 DOI: 10.1002/cbf.3511] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 01/20/2020] [Accepted: 01/24/2020] [Indexed: 01/24/2023]
Abstract
The interferon-regulatory factor 2 (IRF2)-inositol polyphosphate-4-phosphatase type-II (INPP4B) axis has been shown to suppress cell apoptosis by inducing autophagy in acute myeloid leukaemia (AML) cells. T helper type 1 cell (Th1)/Th2 imbalance is involved in development of autophagy and AML. The aim of this study was to investigate whether IRF2-INPP4B axis regulates autophagy and apoptosis of AML cells via regulating Th1/Th2 cell differentiation. The frequencies of Th2 cells and Th1 cells in transfected CD4+ T cells were determined by flow cytometry. The levels of TNF-α, IFN-γ, IL-4, and IL-13 in peripheral blood and transfected CD4+ T cells from AML patients and healthy donors were examined by ELISA assay. The mRNA levels of IRF2 and INPP4B in peripheral blood mononuclear cells (PBMCs) from AML patients and healthy donors were detected by qRT-PCR. Autophagy was evaluated by green fluorescent protein (GFP)-LC3 immunofluorescence and western blot analysis of autophagy-associated proteins. AML cell apoptosis was detected by flow cytometry. In this study, elevated frequencies of Th2 cells and reduced frequencies of Th1 cells, as well as higher expression of IRF2 and INPP4B, were observed in PBMCs from AML patients relative to healthy donors. Furthermore, IRF2 inhibited Th1 cell differentiation and promoted Th2 cell differentiation through INPP4B. Moreover, we confirmed that IRF2-INPP4B-mediated regulation of Th1/Th2 differentiation promoted autophagy and inhibited apoptosis of AML cells. Collectively, IRF2-INPP4B axis regulates autophagy and apoptosis of AML cells via regulating Th1/Th2 cell differentiation. SIGNIFICANCE OF THE STUDY: In the present study, we confirmed that IRF2-INPP4B-mediated regulation of Th1/Th2 balance promoted autophagy and inhibited apoptosis of AML cells. These findings might provide clues in better understanding of the mechanisms of AML.
Collapse
Affiliation(s)
- Feng Zhang
- Department of Hematology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Kai Zhu
- Department of Hematology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Lin Liu
- Department of Hematology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Junfeng Zhu
- Department of Hematology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Jiajia Li
- Department of Hematology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Pingping Zhang
- Department of Hematology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Zhongli Hu
- Department of Hematology Laboratory, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Yuan Yuan
- Department of Hematology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| |
Collapse
|
29
|
Romani L, Del Chierico F, Chiriaco M, Foligno S, Reddel S, Salvatori G, Cifaldi C, Faraci S, Finocchi A, Rossi P, Bagolan P, D'Argenio P, Putignani L, Fusaro F. Gut Mucosal and Fecal Microbiota Profiling Combined to Intestinal Immune System in Neonates Affected by Intestinal Ischemic Injuries. Front Cell Infect Microbiol 2020; 10:59. [PMID: 32158700 PMCID: PMC7052114 DOI: 10.3389/fcimb.2020.00059] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 02/06/2020] [Indexed: 02/06/2023] Open
Abstract
Background and Purpose: Early life microbiota plays a crucial role in human health by acting as a barrier from pathogens' invasion and maintaining the intestinal immune homoeostasis. Altered fecal microbiota (FM) ecology was reported in newborns affected by intestinal ischemia. Our purpose was to describe, in these patients, the FM, the mucosal microbiota (MM) and the mucosal immunity. Methods: Fourteen newborns underwent intestinal resection because of intestinal ischemia. FM and MM were determined through targeted-metagenomics, diversity assignment and Kruskal-Wallis analyses of Operational taxonomic units (OTUs). The mucosal immune cells were analyzed through cytofluorimetry. Results and Conclusion: Based on the severity intestinal injueris we identified two groups: extensive (EII) and focal intestinal ischemia (FII). FM and MM varied in EII and FII groups, showing in the EII group the predominance of Proteobacteria and Enterobacteriaceae and the reduction of Bacteroidetes and Verrucomicrobia for both microbiota. The MM was characterized by a statistically significant reduction of Bacteroides, Lachnospiraceae and Ruminococcaceae and by a higher diversity in the EII compared to FII group. FM showed a prevalence of Proteobacteria, while the Shannon index was lower in the EII compared to FII group. An overall increment in B- and T-lymphocytes and Natural killer (NK) T-like cells was found for EII mucosal samples associated to an increment of TNF-α and INF-γ expressing cells, compared to FII group. FM and MM carry specific signatures of intestinal ischemic lesions. Further research may be crucial to address the role of specific taxa in EII, expecially with reference to inflammation grade and ischemia extension.
Collapse
Affiliation(s)
- Lorenza Romani
- Division of Immunology and Infectious Diseases, University-Hospital Pediatric Department (DPUO), Bambino Gesù Children's Hospital, IRCSS, Rome, Italy
| | | | - Maria Chiriaco
- Division of Immunology and Infectious Diseases, University-Hospital Pediatric Department (DPUO), Bambino Gesù Children's Hospital, IRCSS, Rome, Italy
| | - Silvia Foligno
- Neonatal Intensive Care Unit, Department of Medical and Surgical Neonatology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Sofia Reddel
- Unit of Human Microbiome, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Guglielmo Salvatori
- Neonatal Intensive Care Unit, Department of Medical and Surgical Neonatology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Cristina Cifaldi
- Division of Immunology and Infectious Diseases, University-Hospital Pediatric Department (DPUO), Bambino Gesù Children's Hospital, IRCSS, Rome, Italy
| | - Simona Faraci
- Digestive Endoscopy and Surgery Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Andrea Finocchi
- Division of Immunology and Infectious Diseases, University-Hospital Pediatric Department (DPUO), Bambino Gesù Children's Hospital, IRCSS, Rome, Italy
| | - Paolo Rossi
- Division of Immunology and Infectious Diseases, University-Hospital Pediatric Department (DPUO), Bambino Gesù Children's Hospital, IRCSS, Rome, Italy
| | - Pietro Bagolan
- Department of Medical and Surgical Neonatology, Neonatal Surgery Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Patrizia D'Argenio
- Division of Immunology and Infectious Diseases, University-Hospital Pediatric Department (DPUO), Bambino Gesù Children's Hospital, IRCSS, Rome, Italy
| | - Lorenza Putignani
- Unit of Parasitology and Unit of Human Microbiome, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Fabio Fusaro
- Department of Medical and Surgical Neonatology, Neonatal Surgery Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| |
Collapse
|
30
|
Long Y, Lu M, Cheng T, Zhan X, Zhan X. Multiomics-Based Signaling Pathway Network Alterations in Human Non-functional Pituitary Adenomas. Front Endocrinol (Lausanne) 2019; 10:835. [PMID: 31920959 PMCID: PMC6928143 DOI: 10.3389/fendo.2019.00835] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 11/15/2019] [Indexed: 12/18/2022] Open
Abstract
Non-functional pituitary adenoma (NFPA) seriously affects hypothanamus-pituitary-target organ axis system, with a series of molecule alterations in the multiple levels of genome, transcriptome, proteome, and post-translational modifications, and those molecules mutually interact in a molecular-network system. Meta analysis coupled with IPA pathway-network program was used to comprehensively analyze nine sets of documented NFPA omics data, including NFPA quantitative transcriptomics data [280 differentially expressed genes (DEGs)], NFPA quantitative proteomics data [50 differentially expressed proteins (DEPs)], NFPA mapping protein data (218 proteins), NFPA mapping protein nitration data (9 nitroproteins and 3 non-nitrated proteins), invasive NFPA quantitative transriptomics data (346 DEGs), invasive NFPA quantitative proteomics data (57 DEPs), control mapping protein data (1469 proteins), control mapping protein nitration data (8 nitroproteins), and control mapping phosphorylation data (28 phosphoproteins). A total of 62 molecular-networks with 861 hub-molecules and 519 canonical-pathways including 54 cancer-related canonical pathways were revealed. A total of 42 hub-molecule panels and 9 canonical-pathway panels were identified to significantly associate with tumorigenesis. Four important molecular-network systems, including PI3K/AKT, mTOR, Wnt, and ERK/MAPK pathway-systems, were confirmed in NFPAs by PTMScan experiments with altered expression-patterns and phosphorylations. Nineteen high-frequency hub-molecules were also validated in NFPAs with PTMScan experiment with at least 2.5-fold changes in expression or phosphorylation, including ERK, ERK1/2, Jnk, MAPK, Mek, p38 MAPK, AKT, PI3K complex, p85, PKC, FAK, Rac, Shc, HSP90, NFκB Complex, histone H3, AP1, calmodulin, and PLC. Furthermore, mTOR and Wnt pathway-systems were confirmed in NFPAs by immunoaffinity Western blot analysis, with significantly decreased expression of PRAS40 and increased phosphorylation levels of p-PRAS40 (Thr246) in mTOR pathway in NFPAs compared to controls, and with the decreased protein expressions of GSK-3β and GSK-3β, significantly increased phosphorylation levels of p-GSK3α (Ser21) and p-GSK3β (Ser9), and increased expression level of β-catenin in Wnt pathway in NFPAs compared to controls. Those findings provided a comphrensive and large-scale pathway network data for NFPAs, and offer the scientific evidence for insights into the accurate molecular mechanisms of NFPA and discovery of the effective biomarkers for diagnosis, prognosis, and determination of therapeutic targets.
Collapse
Affiliation(s)
- Ying Long
- Key Laboratory of Cancer Proteomics of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, China
- Hunan Engineering Laboratory for Structural Biology and Drug Design, Xiangya Hospital, Central South University, Changsha, China
- State Local Joint Engineering Laboratory for Anticancer Drugs, Xiangya Hospital, Central South University, Changsha, China
| | - Miaolong Lu
- Key Laboratory of Cancer Proteomics of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, China
- Hunan Engineering Laboratory for Structural Biology and Drug Design, Xiangya Hospital, Central South University, Changsha, China
- State Local Joint Engineering Laboratory for Anticancer Drugs, Xiangya Hospital, Central South University, Changsha, China
| | - Tingting Cheng
- Key Laboratory of Cancer Proteomics of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, China
- Hunan Engineering Laboratory for Structural Biology and Drug Design, Xiangya Hospital, Central South University, Changsha, China
- State Local Joint Engineering Laboratory for Anticancer Drugs, Xiangya Hospital, Central South University, Changsha, China
| | - Xiaohan Zhan
- Key Laboratory of Cancer Proteomics of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, China
- Hunan Engineering Laboratory for Structural Biology and Drug Design, Xiangya Hospital, Central South University, Changsha, China
- State Local Joint Engineering Laboratory for Anticancer Drugs, Xiangya Hospital, Central South University, Changsha, China
| | - Xianquan Zhan
- Key Laboratory of Cancer Proteomics of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, China
- Hunan Engineering Laboratory for Structural Biology and Drug Design, Xiangya Hospital, Central South University, Changsha, China
- State Local Joint Engineering Laboratory for Anticancer Drugs, Xiangya Hospital, Central South University, Changsha, China
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
- National Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
31
|
Yang G, Jin T, Yin S, Guo D, Zhang C, Xia X, Shi C. trans-Cinnamaldehyde mitigated intestinal inflammation induced by Cronobacter sakazakii in newborn mice. Food Funct 2019; 10:2986-2996. [PMID: 31074758 DOI: 10.1039/c9fo00410f] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Necrotizing enterocolitis (NEC) is a serious intestinal disease associated with a high mortality (40-60%) in newborn infants. Cronobacter sakazakii is an important factor for NEC. However, studies regarding NEC pathogenesis and therapeutic treatments are still limited. Here, a C. sakazakii-induced mouse neonatal intestinal inflammation model was employed to determine the effects of trans-cinnamaldehyde (TC) on infections. TC treatment reduced the number of C. sakazakii colony-forming units in the ileal tissues and mitigated the morphological damage in intestinal tissues. Additionally, it reduced the mRNA transcription of inflammatory genes and production of interleukin 6 and tumor necrosis factor-α in mice infected with C. sakazakii. Moreover, TC treatment suppressed caspase-3 activity, modulated enterocyte apoptosis, and inhibited the nuclear factor-kappa B signaling pathway activation induced by C. sakazakii. These findings suggest that TC has protective effects on C. sakazakii-induced murine intestinal inflammation and that it may be a potential agent for preventing NEC.
Collapse
Affiliation(s)
- Gaoji Yang
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi 712100, P.R. China.
| | | | | | | | | | | | | |
Collapse
|
32
|
TUDCA attenuates intestinal injury and inhibits endoplasmic reticulum stress-mediated intestinal cell apoptosis in necrotizing enterocolitis. Int Immunopharmacol 2019; 74:105665. [DOI: 10.1016/j.intimp.2019.05.050] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 05/20/2019] [Accepted: 05/26/2019] [Indexed: 01/06/2023]
|
33
|
Teng JF, Qin DL, Mei QB, Qiu WQ, Pan R, Xiong R, Zhao Y, Law BYK, Wong VKW, Tang Y, Yu CL, Zhang F, Wu JM, Wu AG. Polyphyllin VI, a saponin from Trillium tschonoskii Maxim. induces apoptotic and autophagic cell death via the ROS triggered mTOR signaling pathway in non-small cell lung cancer. Pharmacol Res 2019; 147:104396. [PMID: 31404628 DOI: 10.1016/j.phrs.2019.104396] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Revised: 08/06/2019] [Accepted: 08/08/2019] [Indexed: 01/04/2023]
Abstract
Non-small cell lung cancer (NSCLC) accounts for approximately 85% of all lung cancers. Our previous studies have proven that Trillium tschonoskii Maxim. (TTM), a traditional Chinese medicine, possesses potent anti-tumor effect. However, the detailed components and molecular mechanism of TTM in anti-NSCLC are still unknown. In the present experiment, polyphyllin VI (PPVI) was successfully isolated from TTM with guidance of the anti-proliferative effect in A549 cells, and the cell death of PPVI treated A549 and H1299 cells was closely linked with the increased intracellular ROS levels. In addition, PPVI induced apoptosis by promoting the protein expression of Bax/Bcl2, caspase-3 and caspase-9, and activated autophagy by improving LC3 II conversion and GFP-LC3 puncta formation in A549 and H1299 cells. The mechanism study found that the activity of mTOR which regulates cell growth, proliferation and autophagy was significantly suppressed by PPVI. Accordingly, the PI3K/AKT and MEK/ERK pathways positively regulating mTOR were inhibited, and AMPK negatively regulating mTOR was activated. In addition, the downstream of mTOR, ULK1 at Ser 757 which downregulates autophagy was inhibited by PPVI. The apoptotic cell death induced by PPVI was confirmed, and it was significantly suppressed by the overexpression of AKT, ERK and mTOR, and the induced autophagic cell death which was depended on the Atg7 was decreased by the inhibitors, such as LY294002 (LY), Bafilomycin A1 (Baf), Compound C (CC) and SBI-0206965 (SBI). Furthermore, the mTOR signaling pathway was regulated by the increased ROS as the initial signal in A549 and H1299 cells. Finally, the anti-tumor growth activity of PPVI in vivo was validated in A549 bearing athymic nude mice. Taken together, our data have firstly demonstrated that PPVI is the main component in TTM that exerts the anti-proliferative effect by inducing apoptotic and autophagic cell death in NSCLC via the ROS-triggered mTOR signaling pathway, and PPVI may be a promising candidate for the treatment of NSCLC in future.
Collapse
Affiliation(s)
- Jin-Feng Teng
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Da-Lian Qin
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China; Education Ministry Key Laboratory of Medical Electrophysiology, Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Southwest Medical University, Luzhou, 646000, China
| | - Qi-Bing Mei
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Wen-Qiao Qiu
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Rong Pan
- Department of Human Anatomy, School of Preclinical Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Rui Xiong
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Ya Zhao
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Betty Yuen-Kwan Law
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Vincent Kam-Wai Wong
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Yong Tang
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Chong-Lin Yu
- Department of Human Anatomy, School of Preclinical Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Feng Zhang
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China.
| | - Jian-Ming Wu
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China; Education Ministry Key Laboratory of Medical Electrophysiology, Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Southwest Medical University, Luzhou, 646000, China.
| | - An-Guo Wu
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China; Education Ministry Key Laboratory of Medical Electrophysiology, Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Southwest Medical University, Luzhou, 646000, China.
| |
Collapse
|
34
|
TLR4 Agonist Monophosphoryl Lipid A Alleviated Radiation-Induced Intestinal Injury. J Immunol Res 2019; 2019:2121095. [PMID: 31275998 PMCID: PMC6589195 DOI: 10.1155/2019/2121095] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 02/03/2019] [Accepted: 04/23/2019] [Indexed: 12/20/2022] Open
Abstract
The small intestine is one of the most sensitive organs to irradiation injury, and the development of high effective radioprotectants especially with low toxicity for intestinal radiation sickness is urgently needed. Monophosphoryl lipid A (MPLA) was found to be radioprotective in our previous study, while its effect against the intestinal radiation injury remained unknown. In the present study, we firstly determined the intestinal apoptosis after irradiation injury according to the TUNEL assay. Subsequently, we adopted the immunofluorescence technique to assess the expression levels of different biomarkers including Ki67, γ-H2AX, and defensin 1 in vivo. Additionally, the inflammatory cytokines were detected by RT-PCR. Our data indicated that MPLA could protect the intestine from ionizing radiation (IR) damage through activating TLR4 signal pathway and regulating the inflammatory cytokines. This research shed new light on the protective effect of the novel TLR4 agonist MPLA against intestine detriment induced by IR.
Collapse
|
35
|
Wang Y, Zhang L, Zhang S, Liu Z, Chen L. High Spatiotemporal Resolution Observation of Glutathione Hydropersulfides in Living Cells and Tissue via a Two-Photon Ratiometric Fluorescent Probe. Anal Chem 2019; 91:7812-7818. [DOI: 10.1021/acs.analchem.9b01511] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Yue Wang
- CAS Key Laboratory of Coastal Environmental Processes and Ecological Remediation, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Liangwei Zhang
- CAS Key Laboratory of Coastal Environmental Processes and Ecological Remediation, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China
| | - Songzi Zhang
- CAS Key Laboratory of Coastal Environmental Processes and Ecological Remediation, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China
| | - Zhe Liu
- Department of Chemistry and Chemical Engineering, Qufu Normal University, Qufu, 273165, China
| | - Lingxin Chen
- CAS Key Laboratory of Coastal Environmental Processes and Ecological Remediation, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China
- Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology, Qingdao 266237, China
- Department of Chemistry and Chemical Engineering, Qufu Normal University, Qufu, 273165, China
- Center for Ocean Mega-Science, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao 266071, China
| |
Collapse
|
36
|
Burge K, Gunasekaran A, Eckert J, Chaaban H. Curcumin and Intestinal Inflammatory Diseases: Molecular Mechanisms of Protection. Int J Mol Sci 2019; 20:ijms20081912. [PMID: 31003422 PMCID: PMC6514688 DOI: 10.3390/ijms20081912] [Citation(s) in RCA: 108] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 04/15/2019] [Accepted: 04/17/2019] [Indexed: 02/07/2023] Open
Abstract
Intestinal inflammatory diseases, such as Crohn’s disease, ulcerative colitis, and necrotizing enterocolitis, are becoming increasingly prevalent. While knowledge of the pathogenesis of these related diseases is currently incomplete, each of these conditions is thought to involve a dysfunctional, or overstated, host immunological response to both bacteria and dietary antigens, resulting in unchecked intestinal inflammation and, often, alterations in the intestinal microbiome. This inflammation can result in an impaired intestinal barrier allowing for bacterial translocation, potentially resulting in systemic inflammation and, in severe cases, sepsis. Chronic inflammation of this nature, in the case of inflammatory bowel disease, can even spur cancer growth in the longer-term. Recent research has indicated certain natural products with anti-inflammatory properties, such as curcumin, can help tame the inflammation involved in intestinal inflammatory diseases, thus improving intestinal barrier function, and potentially, clinical outcomes. In this review, we explore the potential therapeutic properties of curcumin on intestinal inflammatory diseases, including its antimicrobial and immunomodulatory properties, as well as its potential to alter the intestinal microbiome. Curcumin may play a significant role in intestinal inflammatory disease treatment in the future, particularly as an adjuvant therapy.
Collapse
Affiliation(s)
- Kathryn Burge
- Department of Pediatrics, Division of Neonatology, University of Oklahoma Health Sciences Center, 1200 North Everett Drive, ETNP7504, Oklahoma City, OK 73104, USA.
| | - Aarthi Gunasekaran
- Department of Pediatrics, Division of Neonatology, University of Oklahoma Health Sciences Center, 1200 North Everett Drive, ETNP7504, Oklahoma City, OK 73104, USA.
| | - Jeffrey Eckert
- Department of Pediatrics, Division of Neonatology, University of Oklahoma Health Sciences Center, 1200 North Everett Drive, ETNP7504, Oklahoma City, OK 73104, USA.
| | - Hala Chaaban
- Department of Pediatrics, Division of Neonatology, University of Oklahoma Health Sciences Center, 1200 North Everett Drive, ETNP7504, Oklahoma City, OK 73104, USA.
| |
Collapse
|
37
|
Giampieri F, Afrin S, Forbes-Hernandez TY, Gasparrini M, Cianciosi D, Reboredo-Rodriguez P, Varela-Lopez A, Quiles JL, Battino M. Autophagy in Human Health and Disease: Novel Therapeutic Opportunities. Antioxid Redox Signal 2019; 30:577-634. [PMID: 29943652 DOI: 10.1089/ars.2017.7234] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
SIGNIFICANCE In eukaryotes, autophagy represents a highly evolutionary conserved process, through which macromolecules and cytoplasmic material are degraded into lysosomes and recycled for biosynthetic or energetic purposes. Dysfunction of the autophagic process has been associated with the onset and development of many human chronic pathologies, such as cardiovascular, metabolic, and neurodegenerative diseases as well as cancer. Recent Advances: Currently, comprehensive research is being carried out to discover new therapeutic agents that are able to modulate the autophagic process in vivo. Recent evidence has shown that a large number of natural bioactive compounds are involved in the regulation of autophagy by modulating several transcriptional factors and signaling pathways. CRITICAL ISSUES Critical issues that deserve particular attention are the inadequate understanding of the complex role of autophagy in disease pathogenesis, the limited availability of therapeutic drugs, and the lack of clinical trials. In this context, the effects that natural bioactive compounds exert on autophagic modulation should be clearly highlighted, since they depend on the type and stage of the pathological conditions of diseases. FUTURE DIRECTIONS Research efforts should now focus on understanding the survival-supporting and death-promoting roles of autophagy, how natural compounds interact exactly with the autophagic targets so as to induce or inhibit autophagy and on the evaluation of their pharmacological effects in a more in-depth and mechanistic way. In addition, clinical studies on autophagy-inducing natural products are strongly encouraged, also to highlight some fundamental aspects, such as the dose, the duration, and the possible synergistic action of these compounds with conventional therapy.
Collapse
Affiliation(s)
- Francesca Giampieri
- 1 Dipartimento di Scienze Cliniche Specialistiche ed Odontostomatologiche-Sez. Biochimica , Facoltà di Medicina, Università Politecnica delle Marche , Ancona, Italy
| | - Sadia Afrin
- 1 Dipartimento di Scienze Cliniche Specialistiche ed Odontostomatologiche-Sez. Biochimica , Facoltà di Medicina, Università Politecnica delle Marche , Ancona, Italy
| | - Tamara Y Forbes-Hernandez
- 1 Dipartimento di Scienze Cliniche Specialistiche ed Odontostomatologiche-Sez. Biochimica , Facoltà di Medicina, Università Politecnica delle Marche , Ancona, Italy .,2 Area de Nutricion y Salud, Universidad Internacional Iberoamericana , Campeche, Mexico
| | - Massimiliano Gasparrini
- 1 Dipartimento di Scienze Cliniche Specialistiche ed Odontostomatologiche-Sez. Biochimica , Facoltà di Medicina, Università Politecnica delle Marche , Ancona, Italy
| | - Danila Cianciosi
- 1 Dipartimento di Scienze Cliniche Specialistiche ed Odontostomatologiche-Sez. Biochimica , Facoltà di Medicina, Università Politecnica delle Marche , Ancona, Italy
| | - Patricia Reboredo-Rodriguez
- 1 Dipartimento di Scienze Cliniche Specialistiche ed Odontostomatologiche-Sez. Biochimica , Facoltà di Medicina, Università Politecnica delle Marche , Ancona, Italy .,3 Departamento de Quimica Analıtica y Alimentaria, Grupo de Nutricion y Bromatologıa, Universidade Vigo , Ourense, Spain
| | - Alfonso Varela-Lopez
- 1 Dipartimento di Scienze Cliniche Specialistiche ed Odontostomatologiche-Sez. Biochimica , Facoltà di Medicina, Università Politecnica delle Marche , Ancona, Italy
| | - Jose L Quiles
- 4 Department of Physiology, Institute of Nutrition and Food Technology "Jose Mataix," Biomedical Research Centre, University of Granada , Granada, Spain
| | - Maurizio Battino
- 1 Dipartimento di Scienze Cliniche Specialistiche ed Odontostomatologiche-Sez. Biochimica , Facoltà di Medicina, Università Politecnica delle Marche , Ancona, Italy .,5 Centre for Nutrition and Health, Universidad Europea del Atlantico (UEA) , Santander, Spain
| |
Collapse
|
38
|
Bazacliu C, Neu J. Pathophysiology of Necrotizing Enterocolitis: An Update. Curr Pediatr Rev 2019; 15:68-87. [PMID: 30387398 DOI: 10.2174/1573396314666181102123030] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 09/12/2018] [Accepted: 09/15/2018] [Indexed: 12/12/2022]
Abstract
NEC is a devastating disease that, once present, is very difficult to treat. In the absence of an etiologic treatment, preventive measures are required. Advances in decoding the pathophysiology of NEC are being made but a more comprehensive understanding is needed for the targeting of preventative strategies. A better definition of the disease as well as diagnostic criteria are needed to be able to specifically label a disease as NEC. Multiple environmental factors combined with host susceptibility appear to contribute to enhanced risks for developing this disease. Several different proximal pathways are involved, all leading to a common undesired outcome: Intestinal necrosis. The most common form of this disease appears to involve inflammatory pathways that are closely meshed with the intestinal microbiota, where a dysbiosis may result in dysregulated inflammation. The organisms present in the intestinal tract prior to the onset of NEC along with their diversity and functional capabilities are just beginning to be understood. Fulfillment of postulates that support causality for particular microorganisms is needed if bacteriotherapies are to be intelligently applied for the prevention of NEC. Identification of molecular effector pathways that propagate inflammation, understanding of, even incipient role of genetic predisposition and of miRNAs may help solve the puzzle of this disease and may bring the researchers closer to finding a treatment. Despite recent progress, multiple limitations of the current animal models, difficulties related to studies in humans, along with the lack of a "clear" definition will continue to make it a very challenging disease to decipher.
Collapse
Affiliation(s)
- Catalina Bazacliu
- Department of Pediatrics, Division of Neonatology, University of Florida, FL, United States
| | - Josef Neu
- Department of Pediatrics, Division of Neonatology, University of Florida, FL, United States
| |
Collapse
|
39
|
Demina EP, Pierre WC, Nguyen ALA, Londono I, Reiz B, Zou C, Chakraberty R, Cairo CW, Pshezhetsky AV, Lodygensky GA. Persistent reduction in sialylation of cerebral glycoproteins following postnatal inflammatory exposure. J Neuroinflammation 2018; 15:336. [PMID: 30518374 PMCID: PMC6282350 DOI: 10.1186/s12974-018-1367-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 11/14/2018] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND The extension of sepsis encompassing the preterm newborn's brain is often overlooked due to technical challenges in this highly vulnerable population, yet it leads to substantial long-term neurodevelopmental disabilities. In this study, we demonstrate how neonatal neuroinflammation following postnatal E. coli lipopolysaccharide (LPS) exposure in rat pups results in persistent reduction in sialylation of cerebral glycoproteins. METHODS Male Sprague-Dawley rat pups at postnatal day 3 (P3) were injected in the corpus callosum with saline or LPS. Twenty-four hours (P4) or 21 days (P24) following injection, brains were extracted and analyzed for neuraminidase activity and expression as well as for sialylation of cerebral glycoproteins and glycolipids. RESULTS At both P4 and P24, we detected a significant increase of the acidic neuraminidase activity in LPS-exposed rats. It correlated with significantly increased neuraminidase 1 (Neu1) mRNA in LPS-treated brains at P4 and with neuraminidases 1 and 4 at P24 suggesting that these enzymes were responsible for the rise of neuraminidase activity. At both P4 and P24, sialylation of N-glycans on brain glycoproteins decreased according to both mass-spectrometry analysis and lectin blotting, but the ganglioside composition remained intact. Finally, at P24, analysis of brain tissues by immunohistochemistry showed that neurons in the upper layers (II-III) of somatosensory cortex had a reduced surface content of polysialic acid. CONCLUSIONS Together, our data demonstrate that neonatal LPS exposure results in specific and sustained induction of Neu1 and Neu4, causing long-lasting negative changes in sialylation of glycoproteins on brain cells. Considering the important roles played by sialoglycoproteins in CNS function, we speculate that observed re-programming of the brain sialome constitutes an important part of pathophysiological consequences in perinatal infectious exposure.
Collapse
Affiliation(s)
- Ekaterina P Demina
- Department of Paediatrics, Sainte-Justine Hospital Research Center, Université de Montréal, Montreal, H3T 1C5, QC, Canada
| | - Wyston C Pierre
- Department of Paediatrics, Sainte-Justine Hospital Research Center, Université de Montréal, Montreal, H3T 1C5, QC, Canada
| | - Annie L A Nguyen
- Department of Paediatrics, Sainte-Justine Hospital Research Center, Université de Montréal, Montreal, H3T 1C5, QC, Canada
| | - Irene Londono
- Department of Paediatrics, Sainte-Justine Hospital Research Center, Université de Montréal, Montreal, H3T 1C5, QC, Canada
| | - Bela Reiz
- Alberta Glycomics Centre and Department of Chemistry, University of Alberta, Edmonton, T6G 2G2, AB, Canada
| | - Chunxia Zou
- Alberta Glycomics Centre and Department of Chemistry, University of Alberta, Edmonton, T6G 2G2, AB, Canada
| | - Radhika Chakraberty
- Alberta Glycomics Centre and Department of Chemistry, University of Alberta, Edmonton, T6G 2G2, AB, Canada
| | - Christopher W Cairo
- Alberta Glycomics Centre and Department of Chemistry, University of Alberta, Edmonton, T6G 2G2, AB, Canada
| | - Alexey V Pshezhetsky
- Department of Paediatrics, Sainte-Justine Hospital Research Center, Université de Montréal, Montreal, H3T 1C5, QC, Canada. .,Department of Anatomy and Cell Biology, McGill University, Montreal, H3A0C7, QC, Canada. .,Centre de recherche, CHU Sainte-Justine, 3175 Côte-Sainte-Catherine, Montreal, QC, H3T 1C5, Canada.
| | - Gregory A Lodygensky
- Department of Paediatrics, Sainte-Justine Hospital Research Center, Université de Montréal, Montreal, H3T 1C5, QC, Canada. .,Department of Pharmacology and Physiology, Université de Montréal, Montreal, H3T 1J4, QC, Canada. .,Montreal Heart Institute, Montreal, H1T 1C8, QC, Canada. .,Centre de recherche, CHU Sainte-Justine, 3175 Côte-Sainte-Catherine, Montreal, QC, H3T 1C5, Canada.
| |
Collapse
|
40
|
Bigger BW, Begley DJ, Virgintino D, Pshezhetsky AV. Anatomical changes and pathophysiology of the brain in mucopolysaccharidosis disorders. Mol Genet Metab 2018; 125:322-331. [PMID: 30145178 DOI: 10.1016/j.ymgme.2018.08.003] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 08/09/2018] [Accepted: 08/09/2018] [Indexed: 11/28/2022]
Abstract
Mucopolysaccharidosis (MPS) disorders are caused by deficiencies in lysosomal enzymes, leading to impaired glycosaminoglycan (GAG) degradation. The resulting GAG accumulation in cells and connective tissues ultimately results in widespread tissue and organ dysfunction. The seven MPS types currently described are heterogeneous and progressive disorders, with somatic and neurological manifestations depending on the type of accumulating GAG. Heparan sulfate (HS) is one of the GAGs stored in patients with MPS I, II, and VII and the main GAG stored in patients with MPS III. These disorders are associated with significant central nervous system (CNS) abnormalities that can manifest as impaired cognition, hyperactive and/or aggressive behavior, epilepsy, hydrocephalus, and sleeping problems. This review discusses the anatomical and pathophysiological CNS changes accompanying HS accumulation as well as the mechanisms believed to cause CNS abnormalities in MPS patients. The content of this review is based on presentations and discussions on these topics during a meeting on the brain in MPS attended by an international group of MPS experts.
Collapse
Affiliation(s)
- Brian W Bigger
- Stem Cell & Neurotherapies Laboratory, Faculty of Medical and Human Sciences, University of Manchester, Manchester, UK.
| | - David J Begley
- Drug Delivery Group, Institute of Pharmaceutical Science, King's College London, London, UK
| | - Daniela Virgintino
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, Human Anatomy and Histology Unit, Bari University School of Medicine, Bari, Italy
| | - Alexey V Pshezhetsky
- Departments of Pediatrics and Biochemistry, CHU Sainte-Justine, Research Center, University of Montreal, Montreal, QC, Canada
| |
Collapse
|
41
|
Therapeutic and preventative effects of ankaferd blood stopper in an experimental necrotizing enterocolitis model. Biomed Pharmacother 2018; 110:105-110. [PMID: 30465999 DOI: 10.1016/j.biopha.2018.11.023] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 11/06/2018] [Accepted: 11/06/2018] [Indexed: 02/02/2023] Open
Abstract
Necrotizing enterocolitis (NEC) is a major neonatal health problem that especially affects preterm infants and causes severe morbidity and mortality. Although its pathogenesis is not fully understood, important risk factors include prematurity, oxidative stress, inflammation, and apoptosis. Ankaferd Blood Stopper® (ABS) has antioxidant, antiinflammatory, antimicrobial, antiapoptotic, and wound healing accelerant properties. In this study, we aimed to investigate whether treatment with ABS reduced the severity of NEC in rat pups in an experimental NEC model. Thirty-six newborn Wistar albino rat pups were randomly assigned to the control, NEC + saline, or NEC + ABS groups. NEC was induced by intraperitoneal injection of lipopolysaccharide, feeding with hyperosmolar enteral formula, and exposure to hypoxia/hyperoxia and cold stress. ABS was administered intraperitoneally to the pups in the NEC + ABS group daily starting on day 1 of the study at a dose of 2 ml/kg by diluting 2 ml with saline at a ratio of 1:3. All pups were sacrificed on day 4. The terminal ileum including the proximal colon was removed for histopathological and immunohistochemical examination and biochemical analysis. Macroscopic assessment and intestinal injury scores were lower in NEC + ABS group compared to the NEC + saline group (p < 0.05). Immunohistochemical evaluations of caspase-3, -8, and -9 revealed significantly reduced apoptosis in the NEC + ABS group compared to the NEC + saline group (p = 0.001). Total oxidant status, oxidative stress index, tumor necrosis factor α and interleukin-1β levels, and lipid, protein, and deoxyribonucleic acid oxidation products were significantly lower in the NEC + ABS group compared to NEC + saline group (p < 0.001 for all), while total antioxidant status, glutathione, and superoxide dismutase levels were higher in the NEC + ABS group (p < 0.001, p < 0.001, p = 0.01, respectively). ABS treatment has the potential to effectively reduce the severity of intestinal damage in NEC due to its antioxidant, antiinflammatory, and antiapoptotic properties. Therefore, NEC may be an alternative option for treatment.
Collapse
|
42
|
Pshezhetsky AV, Martins C, Ashmarina M. Sanfilippo type C disease: pathogenic mechanism and potential therapeutic applications. Expert Opin Orphan Drugs 2018. [DOI: 10.1080/21678707.2018.1534585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Affiliation(s)
- Alexey V. Pshezhetsky
- Sainte-Justine Hospital Research Center, Department of Paediatrics, University of Montreal, Montreal, Canada
- Department of Anatomy and Cell Biology, McGill University, Montreal, Canada
| | - Carla Martins
- Sainte-Justine Hospital Research Center, Department of Paediatrics, University of Montreal, Montreal, Canada
| | - Mila Ashmarina
- Sainte-Justine Hospital Research Center, Department of Paediatrics, University of Montreal, Montreal, Canada
| |
Collapse
|
43
|
Méndez-García LA, Trejo-Millán F, Martínez-Reyes CP, Manjarrez-Reyna AN, Esquivel-Velázquez M, Melendez-Mier G, Islas-Andrade S, Rojas-Bernabé A, Kzhyshkowska J, Escobedo G. Infliximab ameliorates tumor necrosis factor-alpha-induced insulin resistance by attenuating PTP1B activation in 3T3L1 adipocytes in vitro. Scand J Immunol 2018; 88:e12716. [PMID: 30260514 DOI: 10.1111/sji.12716] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 08/27/2018] [Accepted: 09/02/2018] [Indexed: 02/06/2023]
Abstract
Insulin resistance is the inability to respond to insulin and is considered a key pathophysiological factor in the development of type 2 diabetes. Tumor necrosis factor-alpha (TNF-alpha) can directly contribute to insulin resistance by disrupting the insulin signalling pathway via protein-tyrosine phosphatase 1B (PTP1B) activation, especially in adipocytes. Infliximab (Remicade® ) is a TNF-alpha-neutralizing antibody that has not been fully studied in insulin resistance. We investigated the effect of infliximab on TNF-alpha-induced insulin resistance in 3T3L1 adipocytes in vitro, and examined the possible molecular mechanisms involved. Once differentiated, adipocytes were cultured with 5 mmol L-1 2-deoxy-D-glucose-3 H and stimulated twice with 2 μmol L-1 insulin, in the presence or absence of 5 ng/mL TNF-alpha and/or 10 ng/mL infliximab. Glucose uptake was measured every 20 minutes for 2 hour, and phosphorylated forms of insulin receptor (IR), insulin receptor substrate-2 (IRS-2), protein kinase B (AKT) and PTP1B were determined by Western blotting. TNF-alpha-treated adipocytes showed a significant 64% decrease in insulin-stimulated glucose uptake as compared with control cells, whereas infliximab reversed TNF-alpha actions by significantly improving glucose incorporation. Although IR phosphorylation remained unaltered, TNF-alpha was able to increase PTP1B activation and decrease phosphorylation of IRS-2 and AKT. Notably, infliximab restored phosphorylation of IRS-2 and AKT by attenuating PTP1B activation. This work demonstrates for the first time that infliximab ameliorates TNF-alpha-induced insulin resistance in 3T3L1 adipocytes in vitro by restoring the insulin signalling pathway via PTP1B inhibition. Further clinical research is needed to determine the potential benefit of using infliximab for treating insulin resistance in patients.
Collapse
Affiliation(s)
- Lucia A Méndez-García
- Laboratory for Proteomics and Metabolomics, Research Division, General Hospital of Mexico "Dr. Eduardo Liceaga", Mexico City, Mexico
| | - Fernanda Trejo-Millán
- Laboratory for Proteomics and Metabolomics, Research Division, General Hospital of Mexico "Dr. Eduardo Liceaga", Mexico City, Mexico
| | - Camilo P Martínez-Reyes
- Laboratory for Proteomics and Metabolomics, Research Division, General Hospital of Mexico "Dr. Eduardo Liceaga", Mexico City, Mexico
| | - Aarón N Manjarrez-Reyna
- Laboratory for Proteomics and Metabolomics, Research Division, General Hospital of Mexico "Dr. Eduardo Liceaga", Mexico City, Mexico
| | - Marcela Esquivel-Velázquez
- Laboratory for Proteomics and Metabolomics, Research Division, General Hospital of Mexico "Dr. Eduardo Liceaga", Mexico City, Mexico
| | - Guillermo Melendez-Mier
- Laboratory for Proteomics and Metabolomics, Research Division, General Hospital of Mexico "Dr. Eduardo Liceaga", Mexico City, Mexico
| | - Sergio Islas-Andrade
- Laboratory for Proteomics and Metabolomics, Research Division, General Hospital of Mexico "Dr. Eduardo Liceaga", Mexico City, Mexico
| | - Araceli Rojas-Bernabé
- Research Unit for Experimental Medicine, School of Medicine, National Autonomous University of Mexico, Mexico City, Mexico
| | - Julia Kzhyshkowska
- Department of Innate Immunity and Tolerance, Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Galileo Escobedo
- Laboratory for Proteomics and Metabolomics, Research Division, General Hospital of Mexico "Dr. Eduardo Liceaga", Mexico City, Mexico
| |
Collapse
|
44
|
Giménez-Bastida JA, Laparra-Llopis JM, Baczek N, Zielinski H. Buckwheat and buckwheat enriched products exert an anti-inflammatory effect on the myofibroblasts of colon CCD-18Co. Food Funct 2018; 9:3387-3397. [PMID: 29870039 PMCID: PMC6597957 DOI: 10.1039/c8fo00193f] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Buckwheat (BW) constitutes a good source of bioactive components that show anti-inflammatory effects in vitro and in vivo. The use of functional foods in the prevention and treatment of inflammatory bowel diseases (IBDs) has aroused increasing interest. This study investigates the effect of in vitro digested BW and BW-enriched products (BW-enriched wheat breads, roasted BW groats -fermented and non-fermented-, and BW sprouts) on colon myofibroblasts, the cells involved in the regulation of inflammatory response in the intestine. The cells were treated with different digested-BW products, alone or together with TNF-α (20 ng mL-1), and the effects on the cell migration, mitochondrial membrane potential and cell cycle, processes altered during intestinal inflammation, were investigated. A significant reduction in TNF-α-induced migration (25.5%, p < 0.05) and attenuation of the TNF-α-altered cell cycle (p < 0.05) was observed in myofibroblasts treated with BW-enriched white wheat bread. These results contribute to extend the beneficial effects derived from BW bioactive compounds, and suggest that BW consumption can exert beneficial effects on IBDs.
Collapse
Affiliation(s)
- J A Giménez-Bastida
- Department of Pharmacology. Vanderbilt University School of Medicine, RRB 514, 23rd Ave. S. at Pierce, Nashville, TN 37232-6602, USA
| | | | | | | |
Collapse
|
45
|
Ge Y, Huang M, Yao YM. Autophagy and proinflammatory cytokines: Interactions and clinical implications. Cytokine Growth Factor Rev 2018; 43:38-46. [PMID: 30031632 DOI: 10.1016/j.cytogfr.2018.07.001] [Citation(s) in RCA: 123] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 07/10/2018] [Accepted: 07/11/2018] [Indexed: 02/07/2023]
Abstract
Autophagy is a ubiquitous cellular process that regulates cell growth, survival, development and death. Its process is closely associated with diverse conditions, such as liver diseases, neurodegenerative diseases, myopathy, heart diseases, cancer, immunization, and inflammatory diseases. Thus, understanding the modulation of autophagy may provide novel insight into potential therapeutic targets. Autophagy is closely intertwined with inflammatory and immune responses, and cytokines may help mediate this interaction. Autophagy has been shown to regulate, and be regulated by, a wide range of proinflammatory cytokines. This review aims to summarize recent progress in elucidating the interplay between autophagy and proinflammatory cytokines, including IFN-γ, TNF-α, IL-17, and cytokines of the IL-1 family (e.g., IL-1α, IL-1β, IL-33, and IL-36).
Collapse
Affiliation(s)
- Yun Ge
- Department of General Intensive Care Unit, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Man Huang
- Department of General Intensive Care Unit, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Yong-Ming Yao
- Trauma Research Center, First Hospital Affiliated to the Chinese PLA General Hospital, No.51 Fu-Cheng Road, Beijing 100048, China.
| |
Collapse
|
46
|
Digested Early Preterm Human Milk Suppresses Tumor Necrosis Factor-induced Inflammation and Cytotoxicity in Intestinal Epithelial Cells. J Pediatr Gastroenterol Nutr 2018; 66:e153-e157. [PMID: 29470288 DOI: 10.1097/mpg.0000000000001932] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVES The aim of this study was to determine the effect of digested whole human milk (HM; first sample available after birth from mothers of premature infants) on inflammation, oxidative stress, and cytotoxicity in Caco-2 human intestinal epithelial cells stimulated with lipopolysaccharides or tumor necrosis factor (TNF) to mimic the potential in vivo insults facing the premature infant's gastrointestinal tract. METHODS Fully differentiated Caco-2 cells were exposed to digested HM (n = 10; samples from 10 different individuals) before stimulation with lipopolysaccharides, TNF, or no stimulation overnight. Inflammation was determined by production of interleukin-8, oxidative stress by levels of F2-isoprostane, and cytotoxicity by released lactate dehydrogenase. RESULTS HM significantly suppressed interleukin-8 production and cytotoxicity in TNF-stimulated cells, while also suppressing cell death under baseline conditions. Individual HM samples differed widely in their ability to modulate cellular responses. CONCLUSIONS Results from this study provide evidence that digested HM can reduce both an exaggerated inflammatory response and intestinal damage that contribute to the pathogenesis of necrotizing enterocolitis.
Collapse
|
47
|
Suh KS, Chon S, Choi EM. Protective effects of piceatannol on methylglyoxal-induced cytotoxicity in MC3T3-E1 osteoblastic cells. Free Radic Res 2018; 52:712-723. [DOI: 10.1080/10715762.2018.1467010] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Kwang Sik Suh
- Department of Endocrinology and Metabolism, School of Medicine, Kyung Hee University, Dongdaemun-gu, Republic of Korea
| | - Suk Chon
- Department of Endocrinology and Metabolism, School of Medicine, Kyung Hee University, Dongdaemun-gu, Republic of Korea
| | - Eun Mi Choi
- Department of Endocrinology and Metabolism, School of Medicine, Kyung Hee University, Dongdaemun-gu, Republic of Korea
| |
Collapse
|
48
|
Gwangwa MV, Joubert AM, Visagie MH. Crosstalk between the Warburg effect, redox regulation and autophagy induction in tumourigenesis. Cell Mol Biol Lett 2018; 23:20. [PMID: 29760743 PMCID: PMC5935986 DOI: 10.1186/s11658-018-0088-y] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 04/27/2018] [Indexed: 02/07/2023] Open
Abstract
Tumourigenic tissue uses modified metabolic signalling pathways in order to support hyperproliferation and survival. Cancer-associated aerobic glycolysis resulting in lactic acid production was described nearly 100 years ago. Furthermore, increased reactive oxygen species (ROS) and lactate quantities increase metabolic, survival and proliferation signalling, resulting in increased tumourigenesis. In order to maintain redox balance, the cell possesses innate antioxidant defence systems such as superoxide dismutase, catalase and glutathione. Several stimuli including cells deprived of nutrients or failure of antioxidant systems result in oxidative stress and cell death induction. Among the cell death machinery is autophagy, a compensatory mechanism whereby energy is produced from damaged and/or redundant organelles and proteins, which prevents the accumulation of waste products, thereby maintaining homeostasis. Furthermore, autophagy is maintained by several pathways including phosphoinositol 3 kinases, the mitogen-activated protein kinase family, hypoxia-inducible factor, avian myelocytomatosis viral oncogene homolog and protein kinase receptor-like endoplasmic reticulum kinase. The persistent potential of cancer metabolism, redox regulation and the crosstalk with autophagy in scientific investigation pertains to its ability to uncover essential aspects of tumourigenic transformation. This may result in clinical translational possibilities to exploit tumourigenic oxidative status and autophagy to advance our capabilities to diagnose, monitor and treat cancer.
Collapse
Affiliation(s)
- Mokgadi Violet Gwangwa
- Department of Physiology, Faculty of Health Sciences, University of Pretoria, Private Bag X323, Arcadia, 0007 South Africa
| | - Anna Margaretha Joubert
- Department of Physiology, Faculty of Health Sciences, University of Pretoria, Private Bag X323, Arcadia, 0007 South Africa
| | - Michelle Helen Visagie
- Department of Physiology, Faculty of Health Sciences, University of Pretoria, Private Bag X323, Arcadia, 0007 South Africa
| |
Collapse
|
49
|
Shukla V, Fuchs P, Liu A, Cohan CH, Dong C, Wright CB, Perez-Pinzon MA, Dave KR. Recurrent Hypoglycemia Exacerbates Cerebral Ischemic Damage in Diabetic Rats via Enhanced Post-Ischemic Mitochondrial Dysfunction. Transl Stroke Res 2018; 10:78-90. [PMID: 29569040 DOI: 10.1007/s12975-018-0622-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 03/06/2018] [Accepted: 03/08/2018] [Indexed: 12/17/2022]
Abstract
Diabetes significantly increases the risk of stroke and post-stroke mortality. Recurrent hypoglycemia (RH) is common among diabetes patients owing to glucose-lowering therapies. Earlier, we showed that RH in a rat model of insulin-dependent diabetes exacerbates cerebral ischemic damage. Impaired mitochondrial function has been implicated as a central player in the development of cerebral ischemic damage. Hypoglycemia is also known to affect mitochondrial functioning. The present study tested the hypothesis that prior exposure of insulin-treated diabetic (ITD) rats to RH exacerbates brain damage via enhanced post-ischemic mitochondrial dysfunction. In a rat model of streptozotocin-induced diabetes, we evaluated post-ischemic mitochondrial function in RH-exposed ITD rats. Rats were exposed to five episodes of moderate hypoglycemia prior to the induction of cerebral ischemia. We also evaluated the impact of RH, both alone and in combination with cerebral ischemia, on cognitive function using the Barnes circular platform maze test. We observed that RH exposure to ITD rats leads to increased cerebral ischemic damage and decreased mitochondrial complex I activity. Exposure of ITD rats to RH impaired spatial learning and memory. Our results demonstrate that RH exposure to ITD rats potentially increases post-ischemic damage via enhanced post-ischemic mitochondrial dysfunction.
Collapse
Affiliation(s)
- Vibha Shukla
- Cerebral Vascular Disease Research Laboratories, University of Miami School of Medicine, 1420 NW 9th Ave, NRB/203E, Miami, FL, 33136, USA.,Department of Neurology, University of Miami School of Medicine, Miami, FL, 33136, USA
| | - Perry Fuchs
- Cerebral Vascular Disease Research Laboratories, University of Miami School of Medicine, 1420 NW 9th Ave, NRB/203E, Miami, FL, 33136, USA.,Department of Neurology, University of Miami School of Medicine, Miami, FL, 33136, USA
| | - Allen Liu
- Cerebral Vascular Disease Research Laboratories, University of Miami School of Medicine, 1420 NW 9th Ave, NRB/203E, Miami, FL, 33136, USA.,Department of Neurology, University of Miami School of Medicine, Miami, FL, 33136, USA
| | - Charles H Cohan
- Cerebral Vascular Disease Research Laboratories, University of Miami School of Medicine, 1420 NW 9th Ave, NRB/203E, Miami, FL, 33136, USA.,Department of Neurology, University of Miami School of Medicine, Miami, FL, 33136, USA.,Evelyn F. McKnight Brain Institute, University of Miami School of Medicine, Miami, FL, 33136, USA
| | - Chuanhui Dong
- Department of Neurology, University of Miami School of Medicine, Miami, FL, 33136, USA.,Evelyn F. McKnight Brain Institute, University of Miami School of Medicine, Miami, FL, 33136, USA
| | - Clinton B Wright
- Department of Neurology, University of Miami School of Medicine, Miami, FL, 33136, USA.,Evelyn F. McKnight Brain Institute, University of Miami School of Medicine, Miami, FL, 33136, USA.,Neuroscience Program, University of Miami School of Medicine, Miami, FL, 33136, USA
| | - Miguel A Perez-Pinzon
- Cerebral Vascular Disease Research Laboratories, University of Miami School of Medicine, 1420 NW 9th Ave, NRB/203E, Miami, FL, 33136, USA.,Department of Neurology, University of Miami School of Medicine, Miami, FL, 33136, USA.,Evelyn F. McKnight Brain Institute, University of Miami School of Medicine, Miami, FL, 33136, USA.,Neuroscience Program, University of Miami School of Medicine, Miami, FL, 33136, USA
| | - Kunjan R Dave
- Cerebral Vascular Disease Research Laboratories, University of Miami School of Medicine, 1420 NW 9th Ave, NRB/203E, Miami, FL, 33136, USA. .,Department of Neurology, University of Miami School of Medicine, Miami, FL, 33136, USA. .,Evelyn F. McKnight Brain Institute, University of Miami School of Medicine, Miami, FL, 33136, USA. .,Neuroscience Program, University of Miami School of Medicine, Miami, FL, 33136, USA.
| |
Collapse
|
50
|
Zhao M, Tang S, Xin J, Wei Y, Liu D. Reactive oxygen species induce injury of the intestinal epithelium during hyperoxia. Int J Mol Med 2017; 41:322-330. [PMID: 29138796 PMCID: PMC5746288 DOI: 10.3892/ijmm.2017.3247] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 10/27/2017] [Indexed: 12/31/2022] Open
Abstract
Long-term therapeutic hyperoxia may exert serious toxic effects on intestinal epithelial cells in vitro and in vivo. The aim of the present study was to investigate the cause of this intestinal injury under conditions of hyperoxia. Caco-2 cells were treated with different concentrations of hydrogen peroxide (H2O2) and 85% hyperoxia for 24 h. higher rates of injury of Caco-2 cells were observed in the hyperoxia and H2O2 groups compared with the control group. The reactive oxygen species (ROS) level of the hyperoxia group was significantly higher compared with that of the 400 µM H2O2 group. The protein and gene levels of RelA, RelB, hypoxia‑inducible factor-1α, tumor necrosis factor-α and apoptosis signal‑regulating kinase 1 were significantly higher in the hyperoxia and H2O2 groups compared with those in the control group. In conclusion, during hyperoxia, intestinal epithelial cells were destroyed and the levels of ROS were increased. Therefore, ROS may play an important role in intestinal injury in a hyperoxic environment.
Collapse
Affiliation(s)
- Min Zhao
- Medical Research Center, Shengjing Hospital, China Medical University, Shenyang, Liaoning 110000, P.R. China
| | - Shimiao Tang
- Medical Research Center, Shengjing Hospital, China Medical University, Shenyang, Liaoning 110000, P.R. China
| | - Junchi Xin
- Medical Research Center, Shengjing Hospital, China Medical University, Shenyang, Liaoning 110000, P.R. China
| | - Yingliang Wei
- Department of Orthopaedic Surgery, Shengjing Hospital, China Medical University, Shenyang, Liaoning 110000, P.R. China
| | - Dongyan Liu
- Medical Research Center, Shengjing Hospital, China Medical University, Shenyang, Liaoning 110000, P.R. China
| |
Collapse
|