1
|
Mahmoudi SK, Abdolahi S, Saniee P, Zali MR, Hatami B, Baghaei K. Limosilactobacillus fermentum role in combination with human mesenchymal stem cell-derived secretome: A novel approach to alleviate inflammation in NASH pathogenesis. Int Immunopharmacol 2025; 156:114686. [PMID: 40250073 DOI: 10.1016/j.intimp.2025.114686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 04/01/2025] [Accepted: 04/14/2025] [Indexed: 04/20/2025]
Abstract
BACKGROUND Nonalcoholic steatohepatitis (NASH) is caused by the accumulation of excess fat in the liver, chronic inflammation, and cell death. The role of the secretome derived from Wharton's jelly and mesenchymal stem cells (WJ-MSC) in reducing inflammation and apoptosis has been investigated in several studies. Also, the strain Limosilactobacillus fermentum (L.fermentum) was identified as an antimicrobial and antioxidant probiotic. This study looked into the role of a combination of secretome and L.fermentum on cellular stress, apoptosis, and inflammation-related pathways in an NASH in-vitro model. METHODS Oil Red O staining confirmed the NASH model was induced using oleic acid and palmitic acid. Then, the 3 different groups were treated with two concentrations of WJ-MSCs-derived secretome, cell-free extract (CFE) of L.fermentum, and their combination. Oxidative stress was evaluated, and western blotting was used to identify the protein. Gene expression and protein quantity were assessed using real-time PCR and ELISA. RESULT The analysis revealed the secretome, L.fermentum, and their combination decreased oxidative stress. Additionally, the low levels of Caspase 3 and 9 led to a reduction in apoptosis. The combined treatment significantly impacted inflammation by increasing IL-10 and decreasing IL-6. The expression of STAT3 was also confirmed to be reduced using western blotting. Despite the significant modulation of TNF-alpha and STAT3 by L. fermentum at a high dose, the combined approach led to enhanced performance and restored the cell proliferation. CONCLUSION This enhancement has the potential to substantially influence the treatment of NASH disease by impacting inflammation, apoptosis, and oxidative stress, thereby revealing therapeutic potential for NASH disease.
Collapse
Affiliation(s)
- Seyedeh Kosar Mahmoudi
- Department of Microbiology and Microbial Biotechnology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Shahrokh Abdolahi
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Parastoo Saniee
- Department of Microbiology and Microbial Biotechnology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Mohammad Reza Zali
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Behzad Hatami
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kaveh Baghaei
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
2
|
Zhu S, Tang S, Ye Z, Jia W. HNF-1α promotes urethral fibrosis by up-regulating STAT3 transcriptional activity in mice hypospadias. Biochim Biophys Acta Mol Basis Dis 2025:167893. [PMID: 40348066 DOI: 10.1016/j.bbadis.2025.167893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 03/29/2025] [Accepted: 05/06/2025] [Indexed: 05/14/2025]
Abstract
One of the most prevalent male urogenital anomalies is hypospadias. The general etiology of hypospadias is still not completely clear, despite the fact that the number of patients with the hypospadias is rising. HNF-1α was predicted as the most activated upstream regulator of severe hypospadias in our previous research. Our study aims to investigate the mechanism by which HNF-1α regulates hypospadias. We found that HNF-1α was highly expressed in hypospadias tissues, and was positively associated with fibrinogen related genes FGA and FGB, respectively. In animal experiments, silencing of HNF-1α resulted in a more narrow urethral groove, and a decreased periurethral pathologic fibrosis. Meanwhile, the inflammation cytokines and oxidative stress related factors were significantly reduced after inhibition of HNF-1α expression. In addition, the protein levels of STAT3, FGA and FGB were also depressed after HNF-1α knockdown. Mechanistically, we showed that the HNF-1α transcriptionally activated STAT3, and subsequently increased the expressions of FGA and FGB. Additionally, suppression of HNF-1α improved the murine hypospadias via inactivating the STAT3. Therefore, our findings confirmed that HNF-1α promoted hypospadias and periurethral fibrosis by activating STAT3. Targeting HNF-1α might the future direction for hypospadias treatment.
Collapse
Affiliation(s)
- Shibo Zhu
- Department of Pediatric Urology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No.9 Jinsui Road, Tianhe District, Guangzhou 510623, China
| | - Sunwei Tang
- Department of Pediatric Urology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No.9 Jinsui Road, Tianhe District, Guangzhou 510623, China
| | - Zijie Ye
- Department of Pediatric Urology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No.9 Jinsui Road, Tianhe District, Guangzhou 510623, China
| | - Wei Jia
- Department of Pediatric Urology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No.9 Jinsui Road, Tianhe District, Guangzhou 510623, China.
| |
Collapse
|
3
|
He Y, Liang Y, Fan M, Zhang J, Miao Q. Jieyu Guben decoction alleviates combined allergic rhinitis and asthma syndrome by balancing Th17/Treg expression and restoring PPARD. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 139:156508. [PMID: 40031093 DOI: 10.1016/j.phymed.2025.156508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 01/12/2025] [Accepted: 02/11/2025] [Indexed: 03/05/2025]
Abstract
BACKGROUND This study was designed to assess the efficacy of the Jieyu Guben Decoction (JYGBD), a novel formula that has not been reported, in treating rats with combined allergic rhinitis and asthma syndrome (CARAS) and the mechanism. METHODS CARAS rats were induced by ovalbumin (OVA) sensitization and treated with JYGBD to analyze the allergic symptoms and the production of OVA-specific antibodies. Hematoxylin-eosin staining, periodic acid-Schiff staining, Toluidine blue staining, Giemsa staining, and MASSON staining were applied to examine the impact of JYGBD treatment on the histopathological damage of nasal mucosa and lungs. Targets of JYGBD were predicted, and the impact of JYGBD on T helper 17 (Th17) inflammation was analyzed. Peroxisome proliferator-activated receptor delta (PPARD) was artificially silenced to assess the effects of PPARD deficiency on Th17 inflammation. The regulation of PPARD on methyl-CpG-binding protein 2 (MECP2) was analyzed as well. RESULTS JYGBD alleviated allergic conditions in rats and inhibited inflammatory cell infiltration and damage in nasal mucosa and lung tissues. The molecular targets of JYGBD were related to Th17 differentiation, and JYGBD alleviated Th17 inflammation in CARAS rats and inhibited Th17 differentiation in vitro. PPARD-mediated transcriptional inhibition of MECP2 blocked signal transducer and activator of transcription 3 (STAT3) activation to alleviate Th17/regulatory T cells (Treg) imbalance. MECP2 deletion and inhibition of STAT3 signaling alleviated PPARD knockdown-induced Th17/Treg imbalance and attenuated CARAS in rats. CONCLUSION JYGBD induces PPARD-mediated transcriptional inhibition of MECP2 to block STAT3 signaling pathway activation, which restores Th17/Treg homeostasis to alleviate CARAS.
Collapse
Affiliation(s)
- Yi He
- Department of Respiratory, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing 100091, PR China
| | - Yanxia Liang
- Department of Respiratory, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing 100091, PR China
| | - Maorong Fan
- Department of Respiratory, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing 100091, PR China.
| | - Jinzhi Zhang
- Department of Respiratory, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing 100091, PR China
| | - Qing Miao
- Department of Respiratory, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing 100091, PR China
| |
Collapse
|
4
|
Gonçalves A, Machado R, Gomes AC. Self-assembled nanoparticles of hybrid elastin-like and Oncostatin M polymers for improved wound healing. BIOMATERIALS ADVANCES 2025; 169:214150. [PMID: 39693870 DOI: 10.1016/j.bioadv.2024.214150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 12/10/2024] [Accepted: 12/12/2024] [Indexed: 12/20/2024]
Abstract
Oncostatin M (OSM) is a pleiotropic cytokine that can significantly enhance wound healing. Here, we report on the use of nanoparticles (NPs) formulated from a genetically engineered A200_hOSM protein polymer, which combines an elastin-like recombinamer (A200) with human OSM (hOSM) in the same molecule, aiming at enhancing wound healing processes. A200_hOSM NPs were obtained by self-assembly and evaluated for their bioactivity in human keratinocytes and fibroblasts. The NPs demonstrated superior efficacy in promoting cell proliferation in a dose-dependent manner, exhibiting nearly threefold greater proliferation at 48 and 72 h, compared to cells treated with commercial hOSM. Moreover, the NPs stimulated cell migration and collagen production through activation of JAK/STAT3 signaling. They also promoted the production of IL-6 and IL-8, pro-inflammatory cytokines with a critical role for wound healing. Promotion of keratinocyte proliferation and differentiation were further validated in non-commercial 3D skin equivalents. The A200_hOSM NPs revealed potential in accelerating wound healing, evidenced by reduced wound size and a thicker epidermal layer. This system represents a significant advancement in the field of bioinspired biomaterials by improving cytokine bioavailability, allowing for localized therapy and offering a cost-effective strategy for employing hOSM in wound healing management.
Collapse
Affiliation(s)
- Anabela Gonçalves
- CBMA (Centre of Molecular and Environmental Biology)/ Aquatic Research Network (ARNET) Associate Laboratory, Department of Biology, University of Minho, Campus of Gualtar, 4710-057 Braga, Portugal; IB-S Institute of Science and Innovation for Sustainability, University of Minho, Campus of Gualtar, 4710-057 Braga, Portugal
| | - Raul Machado
- CBMA (Centre of Molecular and Environmental Biology)/ Aquatic Research Network (ARNET) Associate Laboratory, Department of Biology, University of Minho, Campus of Gualtar, 4710-057 Braga, Portugal; IB-S Institute of Science and Innovation for Sustainability, University of Minho, Campus of Gualtar, 4710-057 Braga, Portugal.
| | - Andreia C Gomes
- CBMA (Centre of Molecular and Environmental Biology)/ Aquatic Research Network (ARNET) Associate Laboratory, Department of Biology, University of Minho, Campus of Gualtar, 4710-057 Braga, Portugal; IB-S Institute of Science and Innovation for Sustainability, University of Minho, Campus of Gualtar, 4710-057 Braga, Portugal.
| |
Collapse
|
5
|
Veliz AL, Hughes L, Carrillo D, Pecaut MJ, Kearns-Jonker M. Immunization induces inflammation in the mouse heart during spaceflight. BMC Genomics 2025; 26:229. [PMID: 40065216 PMCID: PMC11892206 DOI: 10.1186/s12864-025-11426-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 02/28/2025] [Indexed: 03/14/2025] Open
Abstract
Space travel is a growing area of interest and includes initiatives such as NASA's Moon-to-Mars Mission. Reports on the cardiovascular effects of space travel reveal changes in morphology, metabolism, and function of the cardiovascular system. In this study, the cardiovascular response to immunization in space was studied in mice which were housed and immunized while on the International Space Station (ISS). Mice were immunized with tetanus toxoid combined with the adjuvant CpG (TT + CpG) and the effects of vaccination in space were studied using transcriptomics. Analysis of the mouse heart transcriptome was performed on flight control and flight-immunized mice. The results show that immunization aboard the ISS stimulates heightened inflammation in the heart via induction of the nuclear factor kappa B (NF-κB) signaling pathway to promote the release of the pro-inflammatory cytokines IFNγ, IL-17 and IL-6. Additional transcriptomic changes included alterations in the cytoskeleton and in the expression of transcripts associated with protection from oxidative stress. In summary, inflammation in the heart can occur following immunization in space. This investigation explores the impact of immune challenges on the heart and lays the groundwork for future research into additional cardiac alterations which can occur during spaceflight.
Collapse
Affiliation(s)
- Alicia L Veliz
- Department of Pathology and Human Anatomy, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| | - Lorelei Hughes
- Department of Pathology and Human Anatomy, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| | - Delia Carrillo
- Department of Pathology and Human Anatomy, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| | - Michael J Pecaut
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| | - Mary Kearns-Jonker
- Department of Pathology and Human Anatomy, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA.
| |
Collapse
|
6
|
Zhu JW, Wang HM, Aisikaer M, Zhou WJ, Yang TT, Aximujiang K. Application of Chinese Medicine in Treatment of Ulcerative Colitis and Elucidation of Relevant Mechanisms. Chin J Integr Med 2025:10.1007/s11655-025-3824-y. [PMID: 39821880 DOI: 10.1007/s11655-025-3824-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/19/2024] [Indexed: 01/19/2025]
Abstract
Ulcerative colitis (UC) is a chronic, non-specific intestinal disease of unknown etiology, with high incidence rates worldwide. At present, Western medicine treatments have been associated with more adverse effects and poor efficacy. Chinese medicine (CM) is commonly used as an adjuvant treatment for the unique advantages in regulating immune function, repairing intestinal mucosa, and alleviating intestinal inflammation. At the same time, network pharmacology is also providing new ideas and innovations about CM and development of new drugs. This review systematically discusses the progress of research regarding UC treatment using CM, with a main focus on intestinal flora balance, intestinal mucosal barrier, CM enema, acupuncture therapy, and acupoint embedding. This study provides new ideas that clarify the therapeutic targets of UC.
Collapse
Affiliation(s)
- Ji-Wei Zhu
- Xinjiang Medical University, Urumqi, 830017, China
| | | | | | - Wen-Jun Zhou
- Xinjiang Medical University, Urumqi, 830017, China
| | | | - Kasimujiang Aximujiang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi, 830017, China.
- Xinjiang Key Laboratory of Molecular Biology for Endemic Disease, Urumqi, 830017, China.
| |
Collapse
|
7
|
Guo J, Zhang Y, Du Y, Chen Y, Zhao X, Yu B, Cui T, Mao H, Lv B, Wang X, Gao X. Perilla frutescens leaf extracts alleviate acute lung injury in mice by inhibiting KAT2A. JOURNAL OF ETHNOPHARMACOLOGY 2025; 336:118730. [PMID: 39181280 DOI: 10.1016/j.jep.2024.118730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 08/11/2024] [Accepted: 08/21/2024] [Indexed: 08/27/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Acute lung injury (ALI) can lead to respiratory failure and even death. KAT2A is a key target to suppress the development of inflammation. A herb, perilla frutescens, is an effective treatment for pulmonary inflammatory diseases with anti-inflammatory effects; however, its mechanism of action remains unclear. AIM OF THE STUDY The purpose of this study was to investigate the therapeutic effect and underlying mechanism of perilla frutescens leaf extracts (PLE), in the treatment of ALI by focusing on its ability to treat inflammation. MATERIALS AND METHODS In vivo and in vitro models of ALI induced by LPS. Respiratory function, histopathological changes of lung, and BEAS-2B cells damage were assessed upon PLE. This effect is also tested under conditions of KAT2A over expression and KAT2A silencing. RESULTS PLE significantly attenuated LPS-induced histopathological changes in the lungs, improved respiratory function, and increased survival rate from LPS stimuation background in mice. PLE remarkably suppressed the phosphorylation of STAT3, AKT, ERK (1/2) and the release of cytokines (IL-6, TNF-α, and IL-1β) induced by LPS via inhibiting the expression of KAT2A. CONCLUSIONS PLE has a dose-dependent anti-inflammatory effect by inhibiting KAT2A expression to suppress LPS-induced ALI n mice. Our study expands the clinical indications of the traditional medicine PLE and provide a theoretical basis for clinical use of acute lung injury.
Collapse
Affiliation(s)
- Jinhe Guo
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; School of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| | - Yuqi Zhang
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Yaodong Du
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Yang Chen
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Xin Zhao
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Bin Yu
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Tianyi Cui
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Haoping Mao
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Bin Lv
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| | - Xiaoying Wang
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China.
| | - Xiumei Gao
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| |
Collapse
|
8
|
Liu G, Dong BB, Ding ZH, Lan C, Zhu CJ, Liu Q. Unphysiological lung strain promotes ventilation-induced lung injury via activation of the PECAM-1/Src/STAT3 signaling pathway. Front Pharmacol 2025; 15:1469783. [PMID: 39845800 PMCID: PMC11751019 DOI: 10.3389/fphar.2024.1469783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 12/20/2024] [Indexed: 01/24/2025] Open
Abstract
Introduction In patients with acute respiratory distress syndrome, mechanical ventilation often leads to ventilation-induced lung injury (VILI), which is attributed to unphysiological lung strain (UPLS) in respiratory dynamics. Platelet endothelial cell adhesion molecule-1 (PECAM-1), a transmembrane receptor, senses mechanical signals. The Src/STAT3 pathway plays a crucial role in the mechanotransduction network, concurrently triggering pyroptosis related inflammatory responses. We hypothesized that the mechanical stretch caused by UPLS can be sensed by PECAM-1 in the lungs, leading to VILI via the Src/STAT3 and pyroptosis pathway. Methods A VILI model was established in rats through UPLS. The link between lung strain and VILI as well as the change in the activation of PECAM-1, Src/STAT3, and pyroptosis was firstly being explored. Then, the inhibitors of PECAM-1, Src, STAT3 were adopted respectively, the effect on VILI, inflammation, the Src/STAT3 pathway, and pyroptosis was evaluated. In vitro, human umbilical vein endothelial cells (HUVECs) were used to validate the findings in vivo. Results UPLS activated PECAM-1, Src/STAT3 signaling pathway, inflammation, and pyroptosis in the VILI model with rats, whereas inhibition of PECAM-1 or the Src/STAT3 signaling pathway decreased lung injury, inflammatory responses, and pyroptosis. Inhibition of PECAM-1 also reduced activation of the Src/STAT3 signaling pathway. The mechanism was validated with HUVECs exposed to overload mechanical cyclic stretch. Conclusions This study suggests that UPLS contributes to VILI by activating the PECAM-1/Src/STAT3 pathway and inducing inflammatory responses as well aspyroptosis.
Collapse
Affiliation(s)
- Gang Liu
- Department of Emergency Intensive Care Unit, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Bin-Bin Dong
- Department of Emergency Intensive Care Unit, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Zi-Heng Ding
- Department of Emergency Intensive Care Unit, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Chao Lan
- Department of Emergency Intensive Care Unit, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Chang-Ju Zhu
- Department of Emergency, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Qi Liu
- Department of Emergency Intensive Care Unit, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
9
|
Ren M, Ma J, Qu M. Network pharmacology integrated with molecular docking and molecular dynamics simulations to explore the mechanism of Shaoyao Gancao Tang in the treatment of asthma and irritable bowel syndrome. Medicine (Baltimore) 2024; 103:e40929. [PMID: 39686413 PMCID: PMC11651441 DOI: 10.1097/md.0000000000040929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 11/24/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND Numerous studies have demonstrated a correlation between asthma and irritable bowel syndrome (IBS). The Chinese herbal compound Shaoyao Gancao Tang (SYGCT) has been found to have therapeutic effects on both asthma and IBS, but the underlying mechanisms are not yet fully understood. This study aims to explore the key components, key targets, and potential mechanisms of SYGCT in treating asthma with IBS by using network pharmacology, molecular docking techniques and molecular dynamics simulation. METHODS The major chemical components and potential target genes of SYGCT were screened by bioinformatics. The key targets of Asthma-IBS comorbidity were identified based on network modules. The intersection of the drug targets and disease targets was identified as the potential targets of SYGCT in treating asthma-IBS. Gene Ontology functional annotation and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis were performed to identify the biological processes and signaling pathways involved in these potential targets. A protein-protein interaction network was constructed to identify hub targets, while a drug-compound-target topological network was built to screen key compounds. Molecular docking was used to verify the affinity between the hub targets and key compounds. Molecular dynamics analysis was utilized to assess the binding stability of these interactions. RESULTS Network pharmacology analysis revealed that the therapeutic effect of SYGCT on asthma-IBS involved multiple biological processes and signaling pathways. It may exert therapeutic effects primarily through signaling pathways such as IL-17, TNF, and Th17 cell differentiation. The possible targets of SYGCT in the treatment of asthma-IBS could be IL6, TNF, JUN, PTGS2, STAT3, IL1B, CASP3, NFKBIA, IL10, and PPARG. Molecular docking verification showed that the predicted targets had good binding affinity with the compounds, among which PTGS2, CASP3, and PPARG had higher binding energy. Molecular dynamics simulation revealed that PTGS2, CASP3, and PPARG proteins had good stability and high binding strength with the compounds 2-[(3R)-8,8-dimethyl-3,4-dihydro-2H-pyrano[6,5-f]chromen-3-yl]-5-methoxyphenol and shinpterocarpin. CONCLUSION SYGCT plays a therapeutic role in asthma and IBS through multiple targets and pathways, providing a theoretical basis for explaining the mechanism and clinical application of SYGCT in treating different diseases with the same treatment in asthma and IBS.
Collapse
Affiliation(s)
- Mengjiao Ren
- Department of Warm Disease, School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jian Ma
- Department of Warm Disease, School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Minye Qu
- Department of Warm Disease, School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
10
|
DeJesus J, Wang X, Gu Y, Mao RM, Zhou J, Radhakrishnan R. Novel Compound HJC0416 Attenuates Hepatic Fibrosis via HSP90/NF-κB-Associated Mechanism. J Surg Res 2024; 304:305-314. [PMID: 39579470 DOI: 10.1016/j.jss.2024.09.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 08/19/2024] [Accepted: 09/14/2024] [Indexed: 11/25/2024]
Abstract
INTRODUCTION Chronic liver disease is driven by a prolonged wound healing response leading to fibrogenesis, potentially progressing to cirrhosis. Hepatic stellate cells (HSCs) are the primary cells driving hepatic fibrosis because they are major producers of extracellular matrix. The nuclear factor kappa-light-chain-enhancer of activated B cells (NF-ΚB) pathway is a key regulator of inflammatory signaling, and survival of activated HSCs has been found to be NF-KB dependent. Our team previously synthesized HJC0416-a signal transducer and activator of transcription three inhibitor with potent anti-inflammatory effects. In HSCs, HJC0416 reduced cell viability, extracellular matrix production, and-notably-NF-KB activation. However, HJC0416's antifibrogenetic mechanism remains unknown. This study examined the effects of HJC0416 on NF-KB and its associate factor HSP-90 in HSCs. METHODS The activated human HSC line LX-2 was treated with either HJC0416 or 17-AAG, then exposed to TNFα as indicated. Nuclear and cytosolic proteins were isolated for Western blot or immunofluorescence assay. RESULTS HJC0416 significantly attenuated TNFα-induced IκBα phosphorylation, NF-KBp65 nuclear translocation, and DNA binding activity. Endogenous and TNFα-induced p65 phosphorylation of S536 was suppressed by HJC0416. Notably, HJC0416 dose-dependently attenuated the expression of FAK, IKKα, and signal transducer and activator of transcription three which are Heat Shock Protein 90 (HSP90) interacting proteins. The expression of other HSP90 interacting proteins-RIP1, AKT, FAK, and cyclin-dependent kinase nine-were decreased. HSP90-specific inhibitor 17-AAG significantly attenuated TNFα-induced IκBα phosphorylation and degradation, p65 nuclear translocation, DNA binding, and production of collagen type I and fibronectin. CONCLUSIONS The HSP90 chaperone protein may be a key intermediary linking HJC0416's ability to inhibit NF-κB activity. HJC0416 may be a promising drug candidate for liver fibrosis.
Collapse
Affiliation(s)
- Jana DeJesus
- Department of Surgery, University of Texas Medical Branch, Galveston, Texas
| | - Xiaofu Wang
- Department of Surgery, University of Texas Medical Branch, Galveston, Texas
| | - Yanping Gu
- Department of Neurobiology, University of Texas Medical Branch, Galveston, Texas
| | - Rui-Min Mao
- Department of Surgery, University of Texas Medical Branch, Galveston, Texas
| | - Jia Zhou
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas
| | - Ravi Radhakrishnan
- Department of Surgery, University of Texas Medical Branch, Galveston, Texas.
| |
Collapse
|
11
|
Younes OA, Elsherbiny DM, Hanna DMF, Gad AM, Azab SS. Tocilizumab unfolds colo-protective and immunomodulatory effect in experimentally induced ulcerative colitis via mitigating autophagy and ER stress signaling. Inflammopharmacology 2024; 32:3881-3898. [PMID: 39134818 PMCID: PMC11550239 DOI: 10.1007/s10787-024-01527-7] [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: 03/11/2024] [Accepted: 07/05/2024] [Indexed: 11/10/2024]
Abstract
Ulcerative colitis (UC) is an idiopathic, chronic, relapsing inflammatory bowel disease (IBD), characterized by chronic inflammation of the gastrointestinal tract. The pathophysiology of UC is complicated and involves several factors including immune, genetic, and environmental factors. Recently, a huge amount of research has concentrated on the role of interleukins including interleukin-6 (IL-6) in its pathophysiology. Thus, this study aims to examine the colo-protective and immunomodulatory effect of Tocilizumab (TCZ) in an experimental model of dextran sulfate sodium (DSS) induced UC. In the current study, we analyzed the inflammatory, immunomodulatory, apoptotic, autophagy, and endoplasmic reticulum (ER) stress markers and other clinical features including stool consistency, rectal bleeding, and edema markers in rats. Our results showed that induction of colitis caused bloody diarrhea and increased IL-6 levels. Treatment with TCZ significantly ameliorated DSS-induced injury via decreasing inflammatory markers of colon injury (IL-6), signal transducer and activator of transcription-3 (STAT-3), and C-reactive protein (CRP). Furthermore, TCZ attenuated the apoptotic marker (caspase-3), and down-regulated endoplasmic reticulum stress sensor proteins (inositol- requiring transmembrane kinase endonuclease-1 (IRE-1) and activated transcription factor-6 (ATF-6)) and autophagy proteins (autophagy-related 16-like protein 1 (ATG16L1) and nucleotide-binding oligomerization domain-containing protein-2 (NOD2)), as compared to DSS group. Altogether, the current data suggest TCZ to be a promising protective therapy against UC.
Collapse
Affiliation(s)
- Omnia A Younes
- Biologicals Unit at General Administration of Clinical Studies, Egyptian Drug Authority (EDA), Giza, Egypt
| | - Doaa M Elsherbiny
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Diana M F Hanna
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Amany M Gad
- Department of Pharmacology, Egyptian Drug Authority (EDA), Formerly NODCAR, Giza, Egypt
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Sinai University Kantara Branch, Ismailia, Egypt
| | - Samar S Azab
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt.
| |
Collapse
|
12
|
Zeng M, Zhu Z, Yuan W, Tang Z, Qing Z, Lu Q, Wu X, He J, Li Y, Li Z. Verapamil inhibits inflammation and promotes autophagy to alleviate ureteral scar by regulation of CaMK IIδ/STAT3 axis. Ren Fail 2024; 46:2387432. [PMID: 39177245 PMCID: PMC11346332 DOI: 10.1080/0886022x.2024.2387432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 07/08/2024] [Accepted: 07/29/2024] [Indexed: 08/24/2024] Open
Abstract
BACKGROUND Ureteral stricture (US) is a pathological stenosis in the urinary tract characterized by increased collagen synthesis and inflammation. Autophagy activation has been shown to ameliorate tissue fibrosis and protect against fibrotic diseases. Verapamil has beneficial therapeutic benefits on fibrotic disorders. The pharmacological effects of verapamil on fibroblast autophagy in US and the underlying mechanism need to be investigated further. METHODS US patients were recruited to isolate scar tissues, hematoxylin-eosin (HE) and Masson trichrome staining were performed to analyze histopathological changes. The US animal model was established and administered with verapamil (0.05 mg/kg) in the drinking water. Transforming growth factor (TGF)-β1 was adopted to facilitate collagen synthesis in fibroblasts. The mRNA and protein expressions were examined by qRT-PCR, western blot, immunofluorescence and immunohistochemistry. ELISA was adopted to measure interleukin (IL)-1β and IL-6 levels. Molecular interaction experiments like dual luciferase reporter and chromatin immunoprecipitation (ChIP) assays were performed to analyze the interaction between signal transducers and activators of transcription 3 (STAT3) and RNA polymerase II associated factor 1 (PAF1). RESULTS Herein, our results revealed that verapamil activated TGF-β1-treated fibroblast autophagy and inhibited inflammation and fibrosis by repressing Ca2+⁄calmodulin-dependent protein kinase II (CaMK II) δ-mediated STAT3 activation. Our following tests revealed that STAT3 activated PAF1 transcription. PAF1 upregulation abrogated the regulatory effect of verapamil on fibroblast autophagy and fibrosis during US progression. Finally, verapamil mitigated US in vivo by activating fibroblast autophagy. CONCLUSION Taken together, verapamil activated TGF-β1-treated fibroblast autophagy and inhibited fibrosis by repressing the CaMK IIδ/STAT3/PAF1 axis.
Collapse
Affiliation(s)
- Mingqiang Zeng
- Department of Urology, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan Province, P.R. China
- Hunan Provincial Institute of Geriatrics, Research Center for Lower Urinary Tract and Pelvic Floor Functional Diseases, Changsha, Hunan Province, P.R. China
| | - Zhiwei Zhu
- Department of Urology, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan Province, P.R. China
| | - Wuxiong Yuan
- Department of Urology, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan Province, P.R. China
| | - Zhengyan Tang
- Provincial Laboratory for Diagnosis and Treatment of Genitourinary System Disease, Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan Province, P.R. China
| | - Zhibiao Qing
- Department of Urology, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan Province, P.R. China
| | - Qiang Lu
- Department of Urology, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan Province, P.R. China
| | - Xuecheng Wu
- Department of Urology, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan Province, P.R. China
| | - Junhuan He
- Department of Urology, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan Province, P.R. China
| | - Yuanwei Li
- Department of Urology, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan Province, P.R. China
| | - Zhuo Li
- Department of Urology, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan Province, P.R. China
| |
Collapse
|
13
|
Jarmakiewicz-Czaja S, Sokal-Dembowska A, Ferenc K, Filip R. Mechanisms of Insulin Signaling as a Potential Therapeutic Method in Intestinal Diseases. Cells 2024; 13:1879. [PMID: 39594627 PMCID: PMC11593555 DOI: 10.3390/cells13221879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 11/04/2024] [Accepted: 11/12/2024] [Indexed: 11/28/2024] Open
Abstract
Gastrointestinal diseases are becoming a growing public health problem. One of them is inflammatory bowel disease (IBD), which includes ulcerative colitis (UC) and Crohn's disease (CD). The incidence of IBD is increasing in developing countries and declining in developed countries, affecting people of all ages. Researchers have been exploring new treatment options including insulin signaling pathways in the inflammation of the gastrointestinal tract. It seems that a better understanding of the mechanism of IGF-1, GLP-1 and TL1A on the gut microbiota and inflammation may provide new advances in future therapeutic strategies for patients with IBD, but also other intestinal diseases. This review aims to synthesize insights into the effects of GLP, IGF and anti-TL1A on inflammation and the gut microbiota, which may enable their future use in therapy for people with intestinal diseases.
Collapse
Affiliation(s)
- Sara Jarmakiewicz-Czaja
- Institute of Health Sciences, Medical College of Rzeszow University, 35-959 Rzeszow, Poland; (S.J.-C.); (A.S.-D.)
| | - Aneta Sokal-Dembowska
- Institute of Health Sciences, Medical College of Rzeszow University, 35-959 Rzeszow, Poland; (S.J.-C.); (A.S.-D.)
| | - Katarzyna Ferenc
- Institute of Medicine, Medical College of Rzeszow University, 35-959 Rzeszow, Poland;
| | - Rafał Filip
- Institute of Medicine, Medical College of Rzeszow University, 35-959 Rzeszow, Poland;
- Department of Gastroenterology with IBD Unit, Clinical Hospital No. 2, 35-301 Rzeszow, Poland
| |
Collapse
|
14
|
Sander ML, Eulenburg V, Maeyashiki T, Jang JH, Müller SD, Stehr SN, Jungraithmayr W, Piegeler T. Remote Kidney and Liver Injury After Transplantation of Lung Allografts in an Allogeneic Mouse Model. Transplant Proc 2024; 56:2046-2053. [PMID: 39448275 DOI: 10.1016/j.transproceed.2024.10.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 09/16/2024] [Accepted: 10/03/2024] [Indexed: 10/26/2024]
Abstract
BACKGROUND Remote organ dysfunction is common after lung transplantation and might negatively affect the outcome. The local anesthetic ropivacaine was previously demonstrated to attenuate acute rejection after allogeneic lung transplantation in mice. We hypothesized that lung transplantation might result in detectable molecular signs of injury in kidneys and liver and that ropivacaine might attenuate this damage. METHODS Organs from C57BL/6 mice undergoing allogeneic orthotopic single-lung transplantation were procured at postoperative day 5 and analyzed using Western blot and real-time quantitative polymerase chain reaction probing for Src protein tyrosine kinase, STAT3, and bax/bcl-2. During cold ischemia, the allograft had either been flushed with normal saline only or in combination with ropivacaine (1 µM). A nontransplanted group of animals served as the baseline controls. RESULTS The allogeneic stimulus induced by transplantation led to an increase in Src-phosphorylation and STAT3-expression in the kidneys and livers of lung-transplanted mice compared to nontransplanted animals. Bax/bcl-2 as a marker of cellular apoptosis was not affected by the transplantation. In contrast to the findings in the transplanted lungs, the addition of ropivacaine did not have an effect on the examined markers of inflammation in the remote organs. CONCLUSIONS The observed increase in the inflammatory signaling provides first insight into a possible mechanism, by which remote organ dysfunction after lung transplantation might occur.
Collapse
Affiliation(s)
- Marcin L Sander
- Department of Anesthesiology and Intensive Care, University of Leipzig Medical Center, Leipzig, Germany.
| | - Volker Eulenburg
- Department of Anesthesiology and Intensive Care, University of Leipzig Medical Center, Leipzig, Germany; Current address: Translational Anesthesiology and Intensive Care, Medical Faculty, University of Augsburg, Augsburg, Germany
| | - Tatsuo Maeyashiki
- Department of Thoracic Surgery, Center Hospital of the National Center for Global Health and Medicine, Tokyo, Japan; Department of Thoracic Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Jae-Hwi Jang
- Department of Thoracic Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Sarah D Müller
- Department of Anesthesiology and Intensive Care, University of Leipzig Medical Center, Leipzig, Germany
| | - Sebastian N Stehr
- Department of Anesthesiology and Intensive Care, University of Leipzig Medical Center, Leipzig, Germany
| | - Wolfgang Jungraithmayr
- Department of Thoracic Surgery, University Hospital Zurich, Zurich, Switzerland; Department of Thoracic Surgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Tobias Piegeler
- Department of Anesthesiology and Intensive Care, University of Leipzig Medical Center, Leipzig, Germany.
| |
Collapse
|
15
|
Yu E, Kim H, Park H, Hong JH, Jin J, Song Y, Woo JM, Min JK, Yun J. Targeting the VEGFR2 signaling pathway for angiogenesis and fibrosis regulation in neovascular age-related macular degeneration. Sci Rep 2024; 14:25682. [PMID: 39465270 PMCID: PMC11514265 DOI: 10.1038/s41598-024-76258-4] [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: 05/03/2024] [Accepted: 10/11/2024] [Indexed: 10/29/2024] Open
Abstract
Neovascular age-related macular degeneration (nAMD) is characterized by abnormal blood vessel growth from the choroid, leading to complications and eventual blindness. Despite anti-VEGF therapy, subretinal fibrosis remains a major concern, as VEGF/VEGF receptor-2 (VEGFR2) signaling can contribute to both angiogenesis and fibrosis. For the identification of the aqueous humor proteome, we performed liquid chromatography with tandem mass spectrometry analysis. To investigate the potential therapeutic effects of targeting the VEGF signaling pathway using apatinib, a highly selective VEGFR2 tyrosine kinase inhibitor, this study employed in vitro (THP-1 conditioned media-treated ARPE-19 cells) and in vivo (laser-induced choroidal neovascularization mouse) models of nAMD. This study revealed elevated VEGFR2 protein levels in the aqueous humor of nAMD patients, suggesting a potential target to mitigate neovascularization and fibrosis in nAMD. Apatinib effectively reduced VEGFA and αSMA levels in both in vitro and in vivo models. Moreover, apatinib showed improvement in laser-induced subretinal hyper-reflective lesions. The action mechanism was linked to the inhibition of VEGFR2 activation, leading to the suppression of both angiogenesis and fibrosis through the downregulation of STAT3 phosphorylation. Therefore, the VEGFR2 signaling pathway appears to play a central role in the development of nAMD by regulating both angiogenesis and fibrosis.
Collapse
Affiliation(s)
- Eunhye Yu
- College of Pharmacy, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Osong-eup, Heungduk-gu, Cheongju-si, Chungcheongbuk-do, 28160, Republic of Korea
| | - Haechan Kim
- College of Pharmacy, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Osong-eup, Heungduk-gu, Cheongju-si, Chungcheongbuk-do, 28160, Republic of Korea
| | - Hyeonji Park
- New Drug Development Center, Osong Medical Innovation Foundation, 123, Osongsaengmyeong-ro, Osong-eup, Heungduk-gu, Cheongju-si, Chungcheongbuk-do, 28160, Republic of Korea
- College of Pharmacy, Chungbuk National University, 194-21, Osongsaengmyeong 1-ro, Osong-eup, Heungduk-gu, Cheongju-si, Chungcheongbuk-do, 28160, Republic of Korea
| | - Ji Hye Hong
- New Drug Development Center, Osong Medical Innovation Foundation, 123, Osongsaengmyeong-ro, Osong-eup, Heungduk-gu, Cheongju-si, Chungcheongbuk-do, 28160, Republic of Korea
| | - Jonghwa Jin
- New Drug Development Center, Osong Medical Innovation Foundation, 123, Osongsaengmyeong-ro, Osong-eup, Heungduk-gu, Cheongju-si, Chungcheongbuk-do, 28160, Republic of Korea
| | - Yunjeong Song
- Ministry of Food and Drug Safety, 187, Osongsaengmyeong 2-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungcheongbuk-do, 28159, Republic of Korea
| | - Je Moon Woo
- Department of Ophthalmology, College of Medicine, Ulsan University Hospital, University of Ulsan, 25, Daehakbyeongwon-ro, Dong-gu, Ulsan, 44033, Republic of Korea
| | - Jung Kee Min
- Department of Ophthalmology, College of Medicine, Ulsan University Hospital, University of Ulsan, 25, Daehakbyeongwon-ro, Dong-gu, Ulsan, 44033, Republic of Korea.
| | - Jaesuk Yun
- College of Pharmacy, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Osong-eup, Heungduk-gu, Cheongju-si, Chungcheongbuk-do, 28160, Republic of Korea.
| |
Collapse
|
16
|
Chen Z, Jiang P, Su D, Zhao Y, Zhang M. Therapeutic inhibition of the JAK-STAT pathway in the treatment of inflammatory bowel disease. Cytokine Growth Factor Rev 2024; 79:1-15. [PMID: 39179485 DOI: 10.1016/j.cytogfr.2024.07.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 07/19/2024] [Accepted: 07/20/2024] [Indexed: 08/26/2024]
Abstract
Inflammatory bowel disease (IBD) encompasses a group of non-specific chronic intestinal inflammatory conditions of unclear etiology. The current treatment and long-term management primarily involve biologics. Nevertheless, some patients experience treatment failure or intolerance to biologics [1], making these patients a primary focus of IBD research. The Janus kinase (JAK)-Signal Transducers and Activator of Transcription (STAT) signal transduction pathway is crucial to the regulation of immune and inflammatory responses [2], and plays an important role in the pathogenesis of IBD. JAK inhibitors alleviate IBD by suppressing the transmission of JAK-STAT signaling pathway. As the first small-molecule oral inhibitor for IBD, JAK inhibitors greatly improved the treatment of IBD and have demonstrated significant efficacy, with tofacitinib and upadacitinib being approved for the treatment of ulcerative colitis (UC) [3]. JAK inhibitors can effectively alleviate intestinal inflammation in IBD patients who have failed to receive biologics, which may bring new treatment opportunities for refractory IBD patients. This review aims to elucidate the crucial roles of JAK-STAT signal transduction pathway in IBD pathogenesis, examine its role in various cell types within IBD, and explore the research progress of JAK inhibitors as therapeutic agents, paving the road for new IBD treatment strategies.
Collapse
Affiliation(s)
- Zihan Chen
- School of Medicine, Shanghai Jiao Tong University, Shanghai 200001, China
| | - Ping Jiang
- Department of Gastroenterology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing University, Nanjing 210093, China
| | - Dan Su
- FUJIFILM Diosynth Biotechnologies, Watertown, 02472, MA, United States
| | - Yu Zhao
- University of Chicago, Pritzker School of Molecular Engineering, Chicago, IL, 60637
| | - Mingming Zhang
- School of Medicine, Shanghai Jiao Tong University, Shanghai 200001, China; Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200001, China.
| |
Collapse
|
17
|
Maier JA, Castiglioni S, Petrelli A, Cannatelli R, Ferretti F, Pellegrino G, Sarzi Puttini P, Fiorina P, Ardizzone S. Immune-Mediated Inflammatory Diseases and Cancer - a dangerous liaison. Front Immunol 2024; 15:1436581. [PMID: 39359726 PMCID: PMC11445042 DOI: 10.3389/fimmu.2024.1436581] [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: 05/22/2024] [Accepted: 08/29/2024] [Indexed: 10/04/2024] Open
Abstract
Patients with Immune-Mediated Inflammatory Diseases (IMIDs) are known to have an elevated risk of developing cancer, but the exact causative factors remain subject to ongoing debate. This narrative review aims to present the available evidence concerning the intricate relationship between these two conditions. Environmental influences and genetic predisposition lead to a dysregulated immune response resulting in chronic inflammation, which is crucial in the pathogenesis of IMIDs and oncogenic processes. Mechanisms such as the inflammatory microenvironment, aberrant intercellular communication due to abnormal cytokine levels, excessive reparative responses, and pathological angiogenesis are involved. The chronic immunosuppression resulting from IMIDs treatments further adds to the complexity of the pathogenic scenario. In conclusion, this review highlights critical gaps in the current literature, suggesting potential avenues for future research. The intricate interplay between IMIDs and cancer necessitates more investigation to deepen our understanding and improve patient management.
Collapse
Affiliation(s)
- Jeanette A Maier
- Department of Biomedical and Clinical Sciences, Università di Milano, Milano, Italy
| | - Sara Castiglioni
- Department of Biomedical and Clinical Sciences, Università di Milano, Milano, Italy
| | - Alessandra Petrelli
- Department of Clinical Sciences and Community Health, University of Milan, Milano, Italy
| | | | | | | | - Piercarlo Sarzi Puttini
- Department of Biomedical and Clinical Sciences, Università di Milano, Milano, Italy
- IRCCS Ospedale Galeazzi-Sant'Ambrogio, Milano, Italy
| | - Paolo Fiorina
- Department of Biomedical and Clinical Sciences, Università di Milano, Milano, Italy
| | - Sandro Ardizzone
- Gastroenterology Unit, ASST Fatebenefratelli-Sacco, Milano, Italy
| |
Collapse
|
18
|
Xue Y, Chen Y, Sun S, Tong X, Chen Y, Tang S, Wang X, Bi S, Qiu Y, Zhao Q, Qin Z, Xu Q, Ai Y, Chen L, Zhang B, Liu Z, Ji M, Lang M, Chen L, Xu G, Hu L, Ye D, Ji H. TET2-STAT3-CXCL5 nexus promotes neutrophil lipid transfer to fuel lung adeno-to-squamous transition. J Exp Med 2024; 221:e20240111. [PMID: 38805014 PMCID: PMC11129275 DOI: 10.1084/jem.20240111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 03/21/2024] [Accepted: 04/05/2024] [Indexed: 05/29/2024] Open
Abstract
Phenotypic plasticity is a rising cancer hallmark, and lung adeno-to-squamous transition (AST) triggered by LKB1 inactivation is significantly associated with drug resistance. Mechanistic insights into AST are urgently needed to identify therapeutic vulnerability in LKB1-deficient lung cancer. Here, we find that ten-eleven translocation (TET)-mediated DNA demethylation is elevated during AST in KrasLSL-G12D/+; Lkb1L/L (KL) mice, and knockout of individual Tet genes reveals that Tet2 is required for squamous transition. TET2 promotes neutrophil infiltration through STAT3-mediated CXCL5 expression. Targeting the STAT3-CXCL5 nexus effectively inhibits squamous transition through reducing neutrophil infiltration. Interestingly, tumor-infiltrating neutrophils are laden with triglycerides and can transfer the lipid to tumor cells to promote cell proliferation and squamous transition. Pharmacological inhibition of macropinocytosis dramatically inhibits neutrophil-to-cancer cell lipid transfer and blocks squamous transition. These data uncover an epigenetic mechanism orchestrating phenotypic plasticity through regulating immune microenvironment and metabolic communication, and identify therapeutic strategies to inhibit AST.
Collapse
Affiliation(s)
- Yun Xue
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yuting Chen
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
- School of Life Science and Technology, Shanghai Tech University, Shanghai, China
| | - Sijia Sun
- Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), and Molecular and Cell Biology Laboratory, Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| | - Xinyuan Tong
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Yujia Chen
- Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), and Molecular and Cell Biology Laboratory, Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| | - Shijie Tang
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Xue Wang
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Simin Bi
- Department of Physics, State Key Laboratory of Surface Physics, Academy for Engineering and Technology, Key Laboratory of Micro and Nano Photonic Structures (Ministry of Education), Shanghai, China
| | - Yuqin Qiu
- Key Laboratory of Advanced Polymeric Materials, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, China
| | - Qiqi Zhao
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
- School of Life Science and Technology, Shanghai Tech University, Shanghai, China
| | - Zhen Qin
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Qin Xu
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Yingjie Ai
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Leilei Chen
- Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), and Molecular and Cell Biology Laboratory, Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| | - Beizhen Zhang
- University of Chinese Academy of Sciences, Beijing, China
| | - Zhijie Liu
- Department of Physics, State Key Laboratory of Surface Physics, Academy for Engineering and Technology, Key Laboratory of Micro and Nano Photonic Structures (Ministry of Education), Shanghai, China
| | - Minbiao Ji
- Department of Physics, State Key Laboratory of Surface Physics, Academy for Engineering and Technology, Key Laboratory of Micro and Nano Photonic Structures (Ministry of Education), Shanghai, China
| | - Meidong Lang
- Key Laboratory of Advanced Polymeric Materials, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, China
- Department of General Surgery and Laboratory of General Surgery, Xinhua Hospital, Affiliated to School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Luonan Chen
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
- School of Life Science and Technology, Shanghai Tech University, Shanghai, China
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, China
| | - Guoliang Xu
- State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
- Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Chinese Academy of Medical Sciences (RU069), Shanghai, China
| | - Liang Hu
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Dan Ye
- Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), and Molecular and Cell Biology Laboratory, Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| | - Hongbin Ji
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
- School of Life Science and Technology, Shanghai Tech University, Shanghai, China
| |
Collapse
|
19
|
Sharma S, Gali S, Kundu A, Park JH, Kim JS, Kim HS. Tenovin-1, a Selective SIRT1/2 Inhibitor, Attenuates High-fat Diet-induced Hepatic Fibrosis via Inhibition of HSC Activation in ZDF Rats. Int J Biol Sci 2024; 20:3334-3352. [PMID: 38993557 PMCID: PMC11234213 DOI: 10.7150/ijbs.97304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Accepted: 05/21/2024] [Indexed: 07/13/2024] Open
Abstract
Type 2 diabetes mellitus (T2DM) increases the risk of non-alcoholic fatty liver disease (NAFLD) progression to advanced stages, especially upon high-fat diet (HFD). HFD-induced hepatic fibrosis can be marked by oxidative stress, inflammation, and activation of hepatic stellate cells. Sirtuin 1/2 (SIRT1/2), NAD-dependent class III histone deacetylases, are involved in attenuation of fibrosis. In our conducted research, TGF-β1-activated LX-2 cells, free fatty acid (FFA)-treated simultaneous co-culture (SCC) cells, and HFD-induced hepatic fibrosis in Zucker diabetic fatty (ZDF) rats, a widely used animal model in the study of metabolic syndromes, were used to evaluate the protective effect of Tenovin-1, a SIRT1/2 inhibitor. ZDF rats were divided into chow diet, HFD, and HFD + Tenovin-1 groups. Tenovin-1 reduced hepatic damage, inhibited inflammatory cell infiltration, micro/ macro-vesicular steatosis and prevented collagen deposition HFD-fed rats. Tenovin-1 reduced serum biochemical parameters, triglyceride (TG) and malondialdehyde (MDA) levels but increased glutathione, catalase, and superoxide dismutase levels. Tenovin-1 mitigated proinflammatory cytokines IL-6, IL-1β, TNFα and fibrosis biomarkers in HFD rats, TGF-β1-activated LX-2 and FFA treated SCC cells. Additionally, Tenovin-1 suppressed SIRT1/2 expression and inhibited JNK-1 and STAT3 phosphorylation in HFD rats and FFA-treated SCC cells. In conclusion, Tenovin-1 attenuates hepatic fibrosis by stimulating antioxidants and inhibiting inflammatory cytokines under HFD conditions in diabetic rats.
Collapse
Affiliation(s)
- Swati Sharma
- School of Pharmacy, Sungkyunkwan University, 2066, Seobu-ro, Jangan-gu, Suwon 26419, Republic of Korea
| | - Sreevarsha Gali
- School of Pharmacy, Sungkyunkwan University, 2066, Seobu-ro, Jangan-gu, Suwon 26419, Republic of Korea
| | - Amit Kundu
- School of Pharmacy, Sungkyunkwan University, 2066, Seobu-ro, Jangan-gu, Suwon 26419, Republic of Korea
- Department of Pharmacology, GITAM School of Pharmacy, GITAM (Deemed to be University), Rushikonda, Visakhapatnam-530045, Andhra Pradesh, India
| | - Jae Hyeon Park
- School of Pharmacy, Sungkyunkwan University, 2066, Seobu-ro, Jangan-gu, Suwon 26419, Republic of Korea
| | - Jae-Sung Kim
- Department of Surgery, Washington University in St. Louis, St. Louis, MO, 63110, USA
| | - Hyung Sik Kim
- School of Pharmacy, Sungkyunkwan University, 2066, Seobu-ro, Jangan-gu, Suwon 26419, Republic of Korea
| |
Collapse
|
20
|
Wang C, Oishi K, Kobayashi T, Fujii K, Horii M, Fushida N, Kitano T, Maeda S, Ikawa Y, Komuro A, Hamaguchi Y, Matsushita T. The Role of TLR7 and TLR9 in the Pathogenesis of Systemic Sclerosis. Int J Mol Sci 2024; 25:6133. [PMID: 38892317 PMCID: PMC11172923 DOI: 10.3390/ijms25116133] [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: 04/14/2024] [Revised: 05/29/2024] [Accepted: 05/29/2024] [Indexed: 06/21/2024] Open
Abstract
The bleomycin-induced scleroderma model is a well-established and dependable method for creating a mouse model of SSc (systemic sclerosis). In the field of skin connective tissue diseases, increasing evidence from clinical and animal experiments suggests that TLRs (Toll-like receptors) play an important role in several diseases. This study aimed to determine the role of TLR7 (Toll-like receptor 7) and TLR9 (Toll-like receptor 9) in the mechanisms of immune abnormalities and fibrosis in SSc. This study used TLR7-KO mice (TLR7-knockout mice with a balb/c background) and TLR9-KO mice (TLR9-knockout mice with a balb/c background) as well as WT mice (wild-type balb/c mice). All three kinds of mice were induced by BLM (bleomycin) in a scleroderma model as the experimental group; meanwhile, WT mice treated with PBS (phosphate-buffered saline) were used as the control group. We analyzed the fibrotic phenotype and the immunological abnormality phenotype of TLR7-deficient and TLR9-deficient mice in the SSc disease model using flow cytometry, RT-PCR (reverse transcription-polymerase chain reaction), a histological examination, and IHC (immunohistochemical staining). In a mouse model of SSc disease, the deletion of TLR7 attenuated skin and lung fibrosis, while the deletion of TLR9 exacerbated skin and lung fibrosis. The deletion of TLR7 resulted in a relative decrease in the infiltration and expression of various pro-inflammatory and fibrotic cells and cytokines in the skin. On the other hand, the deletion of TLR9 resulted in a relative increase in the infiltration and expression of various pro-inflammatory and cytokine-inhibiting cells and cytokines in the skin. Under the influence of pDCs (plasmacytoid dendritic cells), the balances of Beff/Breg (IL-6 + CD19 + B cell/IL-10 + CD19 + B cell), Th17/Treg (IL-17A + CD4 + T cell/Foxp3 + CD25 + CD4 + T cell), M1/M2 (CD86 + macrophage/CD206 + macrophage), and Th1/Th2 (TNFα + CD3 + CD4 + T cell/IL-4 + CD3 + CD4 + T cell) were biased towards the suppression of inflammation and fibrosis as a result of the TLR7 deletion. Comparatively, the balance was biased towards promoting inflammation and fibrosis due to the TLR9 deletion. In the SSc model, TLR7 promoted inflammation and fibrosis progression, while TLR9 played a protective role. These results suggest that TLR7 and TLR9 play opposite roles in triggering SSc to produce immune system abnormalities and skin fibrosis.
Collapse
Affiliation(s)
- Chenyang Wang
- Department of Dermatology, Kanazawa University Graduate School of Medical Sciences, Kanazawa 920-8641, Japan; (C.W.)
| | - Kyosuke Oishi
- Department of Dermatology, Kanazawa University Graduate School of Medical Sciences, Kanazawa 920-8641, Japan; (C.W.)
| | - Tadahiro Kobayashi
- Department of Dermatology, Kanazawa University Graduate School of Medical Sciences, Kanazawa 920-8641, Japan; (C.W.)
| | - Ko Fujii
- Department of Dermatology, Kanazawa University Graduate School of Medical Sciences, Kanazawa 920-8641, Japan; (C.W.)
| | - Motoki Horii
- Department of Dermatology, Kanazawa University Graduate School of Medical Sciences, Kanazawa 920-8641, Japan; (C.W.)
| | - Natsumi Fushida
- Department of Dermatology, Kanazawa University Graduate School of Medical Sciences, Kanazawa 920-8641, Japan; (C.W.)
| | - Tasuku Kitano
- Department of Dermatology, Kanazawa University Graduate School of Medical Sciences, Kanazawa 920-8641, Japan; (C.W.)
| | - Shintaro Maeda
- Department of Dermatology, Kanazawa University Graduate School of Medical Sciences, Kanazawa 920-8641, Japan; (C.W.)
| | - Yuichi Ikawa
- Department of Dermatology, Kanazawa University Graduate School of Medical Sciences, Kanazawa 920-8641, Japan; (C.W.)
- Department of Plastic Surgery, Kanazawa University Hospital, Kanazawa 920-8641, Japan
| | - Akito Komuro
- Department of Dermatology, Kanazawa University Graduate School of Medical Sciences, Kanazawa 920-8641, Japan; (C.W.)
- Department of Plastic Surgery, Kanazawa University Hospital, Kanazawa 920-8641, Japan
| | - Yasuhito Hamaguchi
- Department of Dermatology, Kanazawa University Graduate School of Medical Sciences, Kanazawa 920-8641, Japan; (C.W.)
| | - Takashi Matsushita
- Department of Dermatology, Kanazawa University Graduate School of Medical Sciences, Kanazawa 920-8641, Japan; (C.W.)
| |
Collapse
|
21
|
Jeong HJ, Koo S, Kang YH, Kim TW, Kim HK, Park YJ. Hepatoprotective effects of paeonol by suppressing hepatic stellate cell activation via inhibition of SMAD2/3 and STAT3 pathways. Food Sci Biotechnol 2024; 33:1939-1946. [PMID: 38752108 PMCID: PMC11091017 DOI: 10.1007/s10068-023-01440-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 09/07/2023] [Accepted: 09/19/2023] [Indexed: 05/18/2024] Open
Abstract
Hepatic stellate cell (HSC) activation is a key event in extracellular matrix accumulation, causing hepatic fibrosis. Therefore, identifying chemicals that inhibit HSC activation is an important therapeutic strategy for hepatic fibrosis. The aim of this study was to investigate the therapeutic effects of paeonol on HSC activation. In LX-2 cells, paeonol inhibited the expression of collagen and decreased the expression of HSC activation markers. In mice with thioacetamide-induced liver fibrosis, paeonol treatment decreased the serum levels of aspartate aminotransferase and alanine transaminase and mRNA expression of α-smooth muscle actin, platelet-derived growth factor-β, and connective-tissue growth factor. Investigation of the underlying molecular mechanism of paeonol showed that paeonol inhibits the SMAD2/3 and STAT3 signaling pathways that are important for HSC activation. On the basis of these results, paeonol should be investigated and developed further for hepatic fibrosis treatment. Supplementary Information The online version contains supplementary material available at 10.1007/s10068-023-01440-9.
Collapse
Affiliation(s)
- Hye-Jin Jeong
- College of Pharmacy, Kyungsung University, Busan, 48434 Republic of Korea
| | - Sooyeon Koo
- College of Pharmacy, Kyungsung University, Busan, 48434 Republic of Korea
| | - Yeon-Ho Kang
- College of Pharmacy, Kyungsung University, Busan, 48434 Republic of Korea
| | - Tae Won Kim
- College of Pharmacy, Kyungsung University, Busan, 48434 Republic of Korea
- Brain Busan 21 plus Research Project Group, Kyungsung University, Busan, Republic of Korea
| | - Hye Kyung Kim
- College of Pharmacy, Kyungsung University, Busan, 48434 Republic of Korea
- Brain Busan 21 plus Research Project Group, Kyungsung University, Busan, Republic of Korea
| | - Yong Joo Park
- College of Pharmacy, Kyungsung University, Busan, 48434 Republic of Korea
| |
Collapse
|
22
|
Wang K, Zhan HQ, Hu Y, Yuan ZY, Yang JF, Yang DS, Tao LS, Xu T. The role of interleukin-20 in liver disease: Functions, mechanisms and clinical applications. Heliyon 2024; 10:e29853. [PMID: 38699038 PMCID: PMC11064155 DOI: 10.1016/j.heliyon.2024.e29853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 04/16/2024] [Accepted: 04/16/2024] [Indexed: 05/05/2024] Open
Abstract
Liver disease is a severe public health concern worldwide. There is a close relationship between the liver and cytokines, and liver inflammation from a variety of causes leads to the release and activation of cytokines. The functions of cytokines are complex and variable, and are closely related to their cellular origin, target molecules and mode of action. Interleukin (IL)-20 has been studied as a pro-inflammatory cytokine that is expressed and regulated in some diseases. Furthermore, accumulating evidences has shown that IL-20 is highly expressed in clinical samples from patients with liver disease, promoting the production of pro-inflammatory molecules involved in liver disease progression, and antagonists of IL-20 can effectively inhibit liver injury and produce protective effects. This review highlights the potential of targeting IL-20 in liver diseases, elucidates the potential mechanisms of IL-20 inducing liver injury, and suggests multiple viable strategies to mitigate the pro-inflammatory response to IL-20. Genomic CRISPR/Cas9-based screens may be a feasible way to further explore the signaling pathways and regulation of IL-20 in liver diseases. Nanovector systems targeting IL-20 offer new possibilities for the treatment and prevention of liver diseases.
Collapse
Affiliation(s)
- Kun Wang
- School of Clinical Medicine, Anhui Medical University, Hefei, 230032, China
| | - He-Qin Zhan
- Department of Pathology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Ying Hu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
| | - Zhan-Yuan Yuan
- Department of Plastic Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230032, China
| | - Jun-Fa Yang
- Department of orthopedics, Anhui Children's Hospital, Hefei, Anhui, 230032, China
| | - Da-Shuai Yang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
| | - Liang-Song Tao
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
| | - Tao Xu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- Institute for Liver Diseases of Anhui Medical University, Anhui Medical University, Hefei, 230032, China
| |
Collapse
|
23
|
Moulari B, Pallandre JR, Béduneau A, Borg C, Pellequer Y, Pudlo M. Comparison of a selective STAT3 inhibitor with a dual STAT3/STAT1 inhibitor using a dextran sulfate sodium murine colitis model: new insight into the debate on selectivity. Ann Gastroenterol 2024; 37:333-340. [PMID: 38779644 PMCID: PMC11107407 DOI: 10.20524/aog.2024.0880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 03/26/2024] [Indexed: 05/25/2024] Open
Abstract
Background Recent advances in the treatment of inflammatory bowel disease include antitumor necrosis factor antibodies and the Janus kinase inhibitor tofacitinib, approved for ulcerative colitis. Janus kinase recruits signal transducers and activators of transcriptions (STAT), which are promising targets in inflammatory bowel diseases. However few inhibitors have been evaluated, and their selectivity with respect to STAT1 and STAT3 remains controversial. Here, we investigated the therapeutic potential of a selective inhibitor vs. a non-selective, closely related compound, in a dextran sulfate sodium (DSS) murine colitis model. Methods Thirty Swiss/CD-1 male mice were used in this study. They were divided into a healthy control group, a colitis-DSS control group, a compound (cpd) 23-treated group, a cpd 46-treated group and an icariin-treated group. For the coadministration experiment with rutin, the cpd 46-treated group and the icariin-treated group were replaced by the oral rutin-treated group and the coadministration rutin/cpd 23-treated group. The effect of the tested inhibitors was also assessed by quantification of proinflammatory markers. Results The selective inhibitor had a significantly greater effect than the dual inhibitor on the disease activity index. We also noticed in curative treatment a significant decrease in the most abundant proinflammatory biomarker present in neutrophilic granulocytes, myeloperoxidase and on proinflammatory cytokines, including tumor necrosis factor-α, interferon-γ, interleukins -6 and -23, with a mild synergy with rutin, the glycoside of quercetin. Conclusion The current study shows how STAT3 selective inhibitors can exert a significant therapeutic effect in the treatment of experimental DSS-colitis.
Collapse
Affiliation(s)
- Brice Moulari
- Université de Franche-Comté, EFS, INSERM, UMR RIGHT (Brice Moulari, Jean-Réné Pallandre, Arnaud Béduneau, Christophe Borg, Yann Pellequer, Marc Pudlo)
| | - Jean-Réné Pallandre
- Université de Franche-Comté, EFS, INSERM, UMR RIGHT (Brice Moulari, Jean-Réné Pallandre, Arnaud Béduneau, Christophe Borg, Yann Pellequer, Marc Pudlo)
| | - Arnaud Béduneau
- Université de Franche-Comté, EFS, INSERM, UMR RIGHT (Brice Moulari, Jean-Réné Pallandre, Arnaud Béduneau, Christophe Borg, Yann Pellequer, Marc Pudlo)
| | - Christophe Borg
- Université de Franche-Comté, EFS, INSERM, UMR RIGHT (Brice Moulari, Jean-Réné Pallandre, Arnaud Béduneau, Christophe Borg, Yann Pellequer, Marc Pudlo)
- Department of Medical Oncology, University Hospital Center (Christophe Borg), Besançon, France
| | - Yann Pellequer
- Université de Franche-Comté, EFS, INSERM, UMR RIGHT (Brice Moulari, Jean-Réné Pallandre, Arnaud Béduneau, Christophe Borg, Yann Pellequer, Marc Pudlo)
| | - Marc Pudlo
- Université de Franche-Comté, EFS, INSERM, UMR RIGHT (Brice Moulari, Jean-Réné Pallandre, Arnaud Béduneau, Christophe Borg, Yann Pellequer, Marc Pudlo)
| |
Collapse
|
24
|
Ma Y, Ye S, Sun K, Gu Y. Effect of curcumin nanoparticles on proliferation and migration of mouse airway smooth muscle cells and airway inflammatory infiltration. Front Pharmacol 2024; 15:1344333. [PMID: 38708080 PMCID: PMC11066239 DOI: 10.3389/fphar.2024.1344333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 04/02/2024] [Indexed: 05/07/2024] Open
Abstract
Curcumin (CUR) possesses the capability to inhibit various inflammatory factors, exert anti-inflammatory effects, and alleviate asthma attacks; however, its hydrophobicity and instability significantly impede its clinical application. In this study, we synthesized CUR-loaded nanoparticles (CUR-NPs) and evaluated their impact on the proliferation, migration, and inflammatory infiltration of mouse airway smooth muscle cells (ASMCs), while investigating their underlying mechanisms. To achieve this objective, ASMCs were isolated from BALB/c mice and subjected to TGF-β1-induced cell proliferation and migration. Our findings demonstrate that CUR-NPs effectively regulate the release of CUR within cells with superior intracellular uptake compared to free CUR. The CCK-8 assay results indicate that the blank carrier does not exhibit any cytotoxic effects on cells, thus rendering the impact of the carrier itself negligible. The TGF-β1 group exhibited a significant increase in cell proliferation, whereas treatment with CUR-NPs significantly suppressed TGF-β1-induced cell proliferation. The findings from both the cell scratch assay and transwell assay demonstrated that TGF-β1 substantially enhanced cell migration, while CUR-NPs treatment effectively attenuated TGF-β1-induced cell migration. The Western blot analysis demonstrated a substantial increase in the expression levels of TGF-β1, p-STAT3, and CTGF in ASMCs following treatment with TGF-β1 when compared to the control group. Nevertheless, this effect was effectively counteracted upon administration of CUR-NPs. Furthermore, an asthma mouse model was successfully established and CUR-NPs were administered through tail vein injection. The serum levels of TGF-β1 and the expression levels of TGF-β1, p-STAT3, and CTGF proteins in the lung tissue of mice in the model group exhibited significant increases compared to those in the control group. However, CUR-NPs treatment effectively attenuated this change. Our research findings suggest that CUR-NPs possess inhibitory effects on ASMC proliferation, migration, and inflammatory infiltration by suppressing activation of the TGF-β1/p-STAT3/CTGF signaling pathway, thereby facilitating inhibition of airway remodeling.
Collapse
Affiliation(s)
- Yucong Ma
- Department of Pediatric Respiration, Children’s Medical Center, The First Affiliated Hospital of Jilin University, Changchun, Jilin, China
| | - Suping Ye
- Department of Reparatory and Critical Care Medicine, The First Affiliated Hospital of Jilin University, Changchun, Jilin, China
| | - Kunpeng Sun
- Department of Reparatory and Critical Care Medicine, The First Affiliated Hospital of Jilin University, Changchun, Jilin, China
| | - Yue Gu
- Department of Reparatory and Critical Care Medicine, The First Affiliated Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
25
|
Pignolo RJ, Kaplan FS, Wang H. Cell Senescence in Heterotopic Ossification. Biomolecules 2024; 14:485. [PMID: 38672501 PMCID: PMC11047966 DOI: 10.3390/biom14040485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 04/08/2024] [Accepted: 04/15/2024] [Indexed: 04/28/2024] Open
Abstract
The formation of bone outside the normal skeleton, or heterotopic ossification (HO), occurs through genetic and acquired mechanisms. Fibrodysplasia ossificans progressiva (FOP), the most devastating genetic condition of HO, is due to mutations in the ACVR1/ALK2 gene and is relentlessly progressive. Acquired HO is mostly precipitated by injury or orthopedic surgical procedures but can also be associated with certain conditions related to aging. Cellular senescence is a hallmark of aging and thought to be a tumor-suppressive mechanism with characteristic features such as irreversible growth arrest, apoptosis resistance, and an inflammatory senescence-associated secretory phenotype (SASP). Here, we review possible roles for cellular senescence in HO and how targeting senescent cells may provide new therapeutic approaches to both FOP and acquired forms of HO.
Collapse
Affiliation(s)
- Robert J. Pignolo
- Department of Medicine, Section of Geriatric Medicine & Gerontology, Mayo Clinic, Rochester, MN 55905, USA
- Divisions of Endocrinology and Hospital Internal Medicine, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, USA;
| | - Frederick S. Kaplan
- Department of Orthopaedic Surgery, The Perelman School of Medicine of the University of Pennsylvania, Philadelphia, PA 19104, USA;
- Department of Medicine, The Perelman School of Medicine of the University of Pennsylvania, Philadelphia, PA 19104, USA
- The Center for Research in FOP and Related Disorders, The Perelman School of Medicine of the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Haitao Wang
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, USA;
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
26
|
Luo Q, Wang J, Ge W, Li Z, Mao Y, Wang C, Zhang L. Exploration of the potential causative genes for inflammatory bowel disease: Transcriptome-wide association analysis, Mendelian randomization analysis and Bayesian colocalisation. Heliyon 2024; 10:e28944. [PMID: 38617957 PMCID: PMC11015108 DOI: 10.1016/j.heliyon.2024.e28944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 03/27/2024] [Accepted: 03/27/2024] [Indexed: 04/16/2024] Open
Abstract
Background Inflammatory bowel disease (IBD) poses a complex challenge due to its intricate underlying mechanisms, and curative treatments remain elusive. Consequently, there is an urgent need to identify genes causally associated with IBD. Methods We extracted blood eQTL data from the GTExv8.ALL.Whole_Blood database, genome-wide association studies (GWAS) summary statistics of IBD from the IEU GWAS database, and performed a three-fold analysis protocol, including transcriptome-wide association analysis, Mendelian randomisation analysis, Bayesian colocalisation, and subsequent potential therapeutic agents identification. Results We identified four pathogenic genes, namely CARD9, RTEL1, STMN3 and ARFRP1, that promote the development of IBD, encompassing both ulcerative colitis (UC) and Crohn's disease (CD). Notably, ARFRP1 exhibited the ability to suppress IBD (encompassing UC and CD) development. Regarding drug prediction, cyclophosphamide emerged as a promising novel therapeutic option for IBD, encompassing UC and CD. Conclusion We identified several potential genes related to IBD (UC and CD), including CARD9, RTEL1, STMN3 and ARFRP1, warranting further investigation in functional studies to elucidate underlying disease mechanisms. Additionally, clinical studies exploring the potential of cyclophosphamide as a treatment avenue for IBD are warranted.
Collapse
Affiliation(s)
- Qinghua Luo
- Clinical Medical College, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Jiawen Wang
- Department of Proctology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wei Ge
- Department of Proctology, Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang, China
| | - Zihao Li
- Office of the President, Jiangmen Wuyi Hospital of Traditional Chinese Medicine, Jiangmen, China
| | - Yuanting Mao
- Clinical Medical College, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Chen Wang
- Department of Proctology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Leichang Zhang
- Formula-Pattern Research Center, Jiangxi University of Chinese Medicine, Jiangxi, China
| |
Collapse
|
27
|
Yan Q, Liu Z, Chen Y, Zhang X, Zheng W, Liu X, Huang H, Liu Q, Jiang Y, Zhan S, Huang X. ITGAM-macrophage modulation as a potential strategy for treating neutrophilic Asthma: insights from bioinformatics analysis and in vivo experiments. Apoptosis 2024; 29:393-411. [PMID: 37950848 DOI: 10.1007/s10495-023-01914-5] [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] [Accepted: 10/27/2023] [Indexed: 11/13/2023]
Abstract
Identification of molecular biomarkers associated with neutrophilic asthma (NA) phenotype may inform the discovery of novel pathobiological mechanisms and the development of diagnostic markers. Three mRNA transcriptome datasets extracted from induced sputum of asthma patients with various inflammatory types were used to screen for macrophage-related molecular mechanisms and targets in NA. Furthermore, the predicted targets were also validated on an independent dataset (N = 3) and animal model (N = 5). A significant increase in total cells, neutrophils and macrophages was observed in bronchoalveolar lavage (BAL) fluid of NA mice induced by ovalbumin/freund's adjuvant, complete (OVA/CFA). And we also found elevated levels of neutrophil and macrophage infiltration in NA subtype in external datasets. NA mice had increased secretion of IgE, IL-1β, TNF-α and IL-6 in serum and BAL fluid. MPO, an enzyme present in neutrophils, was also highly expressed in NA mice. Then, weighted gene co-expression network analysis (WGCNA) identified 684 targets with the strongest correlation with NA, and we obtained 609 macrophage-related specific differentially expressed genes (DEGs) in NA by integrating macrophage-related genes. The top 10 genes with high degree values were obtained and their mRNA levels and diagnostic performance were then determined by RT-qPCR and receiver operator characteristic (ROC) analysis. Statistically significant correlations were found between macrophages and all key targets, with the strongest correlation between ITGAM and macrophages in NA. Double-Immunofluorescence staining further confirmed the co-localization of ITGAM and F4/80 in NA. ITGAM was identified as a critical target to distinguish NA from healthy/non-NA individuals, which may provide a novel avenue to further uncover the mechanisms and therapy of NA.
Collapse
Affiliation(s)
- Qian Yan
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center of Guangzhou, University of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Clinical Research Academy of Chinese Medicine, Guangzhou, China
| | - Zixing Liu
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yujing Chen
- Guangzhou University of Chinese Medicine, Guangzhou, China
- Haikou hospital of Chinese traditional medicine, Haikou, China
| | - Xinxin Zhang
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center of Guangzhou, University of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Clinical Research Academy of Chinese Medicine, Guangzhou, China
| | - Wenjiang Zheng
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaohong Liu
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Huiting Huang
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qiong Liu
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yong Jiang
- Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Shenzhen, China.
| | - Shaofeng Zhan
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.
| | - Xiufang Huang
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.
- Guangzhou University of Chinese Medicine, Guangzhou, China.
- Lingnan Medical Research Center of Guangzhou, University of Chinese Medicine, Guangzhou, China.
- Guangdong Provincial Clinical Research Academy of Chinese Medicine, Guangzhou, China.
- Lingnan Medical Research Center of Guangzhou, University of Chinese Medicine, the First Affiliated Hospital of Guangzhou University of Chinese Medicine, 12 Airport Road, Guangzhou, 510405, People's Republic of China.
| |
Collapse
|
28
|
Wang M, Chen S, He X, Yuan Y, Wei X. Targeting inflammation as cancer therapy. J Hematol Oncol 2024; 17:13. [PMID: 38520006 PMCID: PMC10960486 DOI: 10.1186/s13045-024-01528-7] [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: 08/23/2023] [Accepted: 02/07/2024] [Indexed: 03/25/2024] Open
Abstract
Inflammation has accompanied human beings since the emergence of wounds and infections. In the past decades, numerous efforts have been undertaken to explore the potential role of inflammation in cancer, from tumor development, invasion, and metastasis to the resistance of tumors to treatment. Inflammation-targeted agents not only demonstrate the potential to suppress cancer development, but also to improve the efficacy of other therapeutic modalities. In this review, we describe the highly dynamic and complex inflammatory tumor microenvironment, with discussion on key inflammation mediators in cancer including inflammatory cells, inflammatory cytokines, and their downstream intracellular pathways. In addition, we especially address the role of inflammation in cancer development and highlight the action mechanisms of inflammation-targeted therapies in antitumor response. Finally, we summarize the results from both preclinical and clinical studies up to date to illustrate the translation potential of inflammation-targeted therapies.
Collapse
Affiliation(s)
- Manni Wang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No.17, Block3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Siyuan Chen
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No.17, Block3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Xuemei He
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No.17, Block3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Yong Yuan
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, People's Republic of China.
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No.17, Block3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China.
| |
Collapse
|
29
|
Guo J, Yang T, Zhang W, Yu K, Xu X, Li W, Song L, Gu X, Cao R, Cui S. Inhibition of CD44 suppresses the formation of fibrotic scar after spinal cord injury via the JAK2/STAT3 signaling pathway. iScience 2024; 27:108935. [PMID: 38323002 PMCID: PMC10846335 DOI: 10.1016/j.isci.2024.108935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 11/17/2023] [Accepted: 01/12/2024] [Indexed: 02/08/2024] Open
Abstract
Fibrotic scar is one of the main impediments to axon regeneration following spinal cord injury (SCI). In this study, we found that CD44 was upregulated during the formation of fibrotic scar, and blocking CD44 by IM7 caused downregulation of fibrosis-related extracellular matrix proteins at both 2 and 12 weeks post-spinal cord injury. More Biotinylated dextran amine (BDA)-traced corticospinal tract axons crossed the scar area and extended into the distal region after IM7 administration. A recovery of motor and sensory function was observed based on Basso Mouse Scale (BMS) scores and tail-flick test. In vitro experiments revealed that inhibiting CD44 and JAK2/STAT3 signaling pathway decreased the proliferation, differentiation, and migration of fibroblasts induced by the inflammatory supernatant. Collectively, these findings highlight the critical role of CD44 and its downstream JAK2/STAT3 signaling pathway in fibrotic scar formation, suggesting a potential therapeutic target for SCI.
Collapse
Affiliation(s)
- Jin Guo
- Department of Hand and Foot Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province 130033, China
- Key Laboratory of Peripheral Nerve Injury and Regeneration of Jilin Province, Changchun, Jilin Province 130033, China
| | - Tuo Yang
- Department of Hand and Foot Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province 130033, China
- Key Laboratory of Peripheral Nerve Injury and Regeneration of Jilin Province, Changchun, Jilin Province 130033, China
| | - Weizhong Zhang
- Department of Hand and Foot Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province 130033, China
- Key Laboratory of Peripheral Nerve Injury and Regeneration of Jilin Province, Changchun, Jilin Province 130033, China
| | - Kaiming Yu
- Department of Hand and Foot Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province 130033, China
- Key Laboratory of Peripheral Nerve Injury and Regeneration of Jilin Province, Changchun, Jilin Province 130033, China
| | - Xiong Xu
- Department of Hand and Foot Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province 130033, China
- Key Laboratory of Peripheral Nerve Injury and Regeneration of Jilin Province, Changchun, Jilin Province 130033, China
| | - Weizhen Li
- Department of Hand and Foot Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province 130033, China
- Key Laboratory of Peripheral Nerve Injury and Regeneration of Jilin Province, Changchun, Jilin Province 130033, China
| | - Lili Song
- Department of Hand & Microsurgery, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Xiaosong Gu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu 226001, China
| | - Rangjuan Cao
- Department of Hand and Foot Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province 130033, China
- Key Laboratory of Peripheral Nerve Injury and Regeneration of Jilin Province, Changchun, Jilin Province 130033, China
| | - Shusen Cui
- Department of Hand and Foot Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province 130033, China
- Key Laboratory of Peripheral Nerve Injury and Regeneration of Jilin Province, Changchun, Jilin Province 130033, China
| |
Collapse
|
30
|
Zheng CY, Yu YX, Bai X. Polycystic ovary syndrome and related inflammation in radiomics; relationship with patient outcome. Semin Cell Dev Biol 2024; 154:328-333. [PMID: 36933953 DOI: 10.1016/j.semcdb.2023.02.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 02/26/2023] [Accepted: 02/26/2023] [Indexed: 03/19/2023]
Abstract
Polycystic ovary syndrome (PCOS) refers to a condition that often has 'poly' liquid containing sacks around ovaries. It affects reproductive-aged females giving rise to menstrual and related reproductive issues. PCOS is marked by hormonal imbalance often resulting in hyperandrogenism. Inflammation is now considered a central manifestation of this disease with several inflammatory biomarkers such as TNF-α, C-reactive protein and Interleukins-6/18 found to be particularly elevated in PCOS patients. Diagnosis is often late, and MRI-based diagnosis, along with blood-based analyses, are still the best bet for a definitive diagnosis. Radiomics also offers several advantages and should be exploited to the maximum. The mechanisms of PCOS onset and progression are not very well known but pituitary dysfunction and elevated gonadotrophin releasing hormone resulting in high levels of luteinizing hormone are indicative of an activated hypothalamic-pituitary-ovarian axis in PCOS. A number of studies have also identified signaling pathways such as PI3K/Akt, NF-κB and STAT in PCOS etiology. The links of these signaling pathways to inflammation further underline the importance of inflammation in PCOS, which needs to be resolved for improving patient outcomes.
Collapse
Affiliation(s)
- Chun-Yang Zheng
- Embryo Laboratory, Jinghua Hospital of Shenyang, No. 83, Zhongshan Road, Heping District, Shenyang 110000, Liaoning Province, China
| | - Yue-Xin Yu
- Department of Reproductive Medicine, General Hospital of Northern Theater Command, No. 5, Guangrong Street, Heping District, Shenyang 110000, Liaoning Province, China
| | - Xue Bai
- Department of Reproductive Medicine, General Hospital of Northern Theater Command, No. 5, Guangrong Street, Heping District, Shenyang 110000, Liaoning Province, China.
| |
Collapse
|
31
|
Mohanty D, Padhee S, Sahoo C, Jena S, Sahoo A, Chandra Panda P, Nayak S, Ray A. Integrating network pharmacology and experimental verification to decipher the multitarget pharmacological mechanism of Cinnamomum zeylanicum essential oil in treating inflammation. Heliyon 2024; 10:e24120. [PMID: 38298712 PMCID: PMC10828654 DOI: 10.1016/j.heliyon.2024.e24120] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 01/03/2024] [Accepted: 01/03/2024] [Indexed: 02/02/2024] Open
Abstract
Inflammatory diseases contribute to more than 50 % of global deaths. Research suggests that network pharmacology can reveal the biological mechanisms underlying inflammatory diseases and drug effects at the molecular level. The aim of the study was to clarify the biological mechanism of Cinnamomum zeylanicum essential oil (CZEO) and predict molecular targets of CZEO against inflammation by employing network pharmacology and in vitro assays. First, the genes related to inflammation were identified from the Genecards and Online Mendelian Inheritance in Man (OMIM) databases. The CZEO targets were obtained from the SwissTargetPrediction and Similarity Ensemble Approach (SEA) database. A total of 1057 CZEO and 526 anti-inflammation targets were obtained. The core hub target of CZEO anti-inflammatory was obtained using the protein-protein interaction network. KEGG pathway analysis suggested CZEO to exert anti-inflammatory effect mainly through Tumor necrosis factor, Toll-like receptor and IL-17 signalling pathway. Molecular docking of active ingredients-core targets interactions was modelled using Pyrx software. Docking and simulation studies revealed benzyl benzoate to exhibit good binding affinity towards IL8 protein. MTT assay revealed CZEO to have non-cytotoxic effect on RAW 264.7 cells. CZEO also inhibited the production of NO, PGE2, IL-6, IL-1β and TNF-α and promoted the activity of endogenous antioxidant enzymes in LPS-stimulated RAW 264.7 cells. Additionally, CZEO inhibited intracellular ROS generation, NF-kB nuclear translocation and modulated the expression of downstream genes involved in Toll-like receptor signalling pathway. The results deciphered the mechanism of CZEO in treating inflammation and provided a theoretical basis for its clinical application.
Collapse
Affiliation(s)
- Debajani Mohanty
- Centre for Biotechnology, Siksha O Anusandhan (Deemed to be University), Kalinganagar, Ghatikia, Bhubaneswar-751003, Odisha, India
| | - Sucheesmita Padhee
- Centre for Biotechnology, Siksha O Anusandhan (Deemed to be University), Kalinganagar, Ghatikia, Bhubaneswar-751003, Odisha, India
| | - Chiranjibi Sahoo
- Centre for Biotechnology, Siksha O Anusandhan (Deemed to be University), Kalinganagar, Ghatikia, Bhubaneswar-751003, Odisha, India
| | - Sudipta Jena
- Centre for Biotechnology, Siksha O Anusandhan (Deemed to be University), Kalinganagar, Ghatikia, Bhubaneswar-751003, Odisha, India
| | - Ambika Sahoo
- Centre for Biotechnology, Siksha O Anusandhan (Deemed to be University), Kalinganagar, Ghatikia, Bhubaneswar-751003, Odisha, India
| | - Pratap Chandra Panda
- Centre for Biotechnology, Siksha O Anusandhan (Deemed to be University), Kalinganagar, Ghatikia, Bhubaneswar-751003, Odisha, India
| | - Sanghamitra Nayak
- Centre for Biotechnology, Siksha O Anusandhan (Deemed to be University), Kalinganagar, Ghatikia, Bhubaneswar-751003, Odisha, India
| | - Asit Ray
- Centre for Biotechnology, Siksha O Anusandhan (Deemed to be University), Kalinganagar, Ghatikia, Bhubaneswar-751003, Odisha, India
| |
Collapse
|
32
|
Almeaqli MT, Alaidaa Y, Alnajjar FM, Al Shararh AS, Alharbi DS, Almslmani YI, Alotibi YA, Alrashidi HS, Alshehri WA, Hassan HM, Al-Gayyar MMH. Therapeutic Effects of Arctiin on Alzheimer's Disease-like Model in Rats by Reducing Oxidative Stress, Inflammasomes and Fibrosis. Curr Alzheimer Res 2024; 21:276-288. [PMID: 39136502 DOI: 10.2174/0115672050333388240801043509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 06/28/2024] [Accepted: 07/19/2024] [Indexed: 10/25/2024]
Abstract
BACKGROUND Alzheimer's disease (AD) affects approximately 50 million people globally and is expected to triple by 2050. Arctiin is a lignan found in the Arctium lappa L. plant. Arctiin possesses anti-proliferative, antioxidative and anti-adipogenic. OBJECTIVES We aimed to explore the potential therapeutic effects of Arctiin on rats with AD by evaluating the expression of TLR4, NLRP3, STAT3, TGF-β, cyclin D1, and CDK2. METHODS AD was induced in rats by administering 70 mg/kg of aluminum chloride through intraperitoneal injection daily for six weeks. After inducing AD, some rats were treated with 25 mg/kg of Arctiin daily for three weeks through oral gavage. Furthermore, to examine the brain tissue structure, hippocampal sections were stained with hematoxylin/eosin and anti-TLR4 antibodies. The collected samples were analyzed for gene expression and protein levels of TLR4, NLRP3, STAT3, TGF-β, cyclin D1, and CDK2. RESULTS In behavioral tests, rats showed a significant improvement in their behavior when treated with Arctiin. Microimages stained with hematoxylin/eosin showed that Arctiin helped to improve the structure and cohesion of the hippocampus, which was previously impaired by AD. Furthermore, Arctiin reduced the expression of TLR4, NLRP3, STAT3, TGF-β, cyclin D1, and CDK2. CONCLUSION Arctiin can enhance rats' behavior and structure of the hippocampus in AD rats. This is achieved through its ability to reduce the expression of both TLR4 and NLRP3, hence inhibiting the inflammasome pathway. Furthermore, Arctiin can improve tissue fibrosis by regulating STAT3 and TGF-β. Lastly, it can block the cell cycle proteins cyclin D1 and CDK2.
Collapse
Affiliation(s)
- Mohamed T Almeaqli
- PharmD Program, Faculty of Pharmacy, University of Tabuk, Tabuk, 71491, Saudi Arabia
| | - Yazeed Alaidaa
- PharmD Program, Faculty of Pharmacy, University of Tabuk, Tabuk, 71491, Saudi Arabia
| | - Faisal M Alnajjar
- PharmD Program, Faculty of Pharmacy, University of Tabuk, Tabuk, 71491, Saudi Arabia
| | - Abdullah S Al Shararh
- PharmD Program, Faculty of Pharmacy, University of Tabuk, Tabuk, 71491, Saudi Arabia
| | - Danah S Alharbi
- PharmD Program, Faculty of Pharmacy, University of Tabuk, Tabuk, 71491, Saudi Arabia
| | - Yazeed I Almslmani
- PharmD Program, Faculty of Pharmacy, University of Tabuk, Tabuk, 71491, Saudi Arabia
| | - Yousef A Alotibi
- PharmD Program, Faculty of Pharmacy, University of Tabuk, Tabuk, 71491, Saudi Arabia
| | - Hani S Alrashidi
- PharmD Program, Faculty of Pharmacy, University of Tabuk, Tabuk, 71491, Saudi Arabia
| | - Wael A Alshehri
- PharmD Program, Faculty of Pharmacy, University of Tabuk, Tabuk, 71491, Saudi Arabia
| | - Hanan M Hassan
- Department of Pharmacology and Biochemistry, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa City, Egypt
| | - Mohammed M H Al-Gayyar
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Tabuk, Tabuk, 71491, Saudi Arabia
| |
Collapse
|
33
|
Gandhi GR, Mohana T, Athesh K, Hillary VE, Vasconcelos ABS, Farias de Franca MN, Montalvão MM, Ceasar SA, Jothi G, Sridharan G, Gurgel RQ, Xu B. Anti-inflammatory natural products modulate interleukins and their related signaling markers in inflammatory bowel disease: A systematic review. J Pharm Anal 2023; 13:1408-1428. [PMID: 38223446 PMCID: PMC10785269 DOI: 10.1016/j.jpha.2023.09.012] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 09/17/2023] [Accepted: 09/18/2023] [Indexed: 01/16/2024] Open
Abstract
This review aims to identify in vivo studies investigating the potential of plant substances and their natural molecules in managing inflammatory bowel disease (IBD). Specifically, the objective is to examine the impact of these substances on interleukins and other key inflammatory signaling markers. Relevant articles published up to December 2022 were identified through a search of the PubMed, Scopus, Web of Science, and Embase databases. The search used keywords including "inflammatory bowel disease", "medicinal plants", "natural molecules", "anti-inflammatory", and "ulcerative colitis", and identified 1,878 potentially relevant articles, of which 89 were included in this review after completion of the selection process. This study provides preclinical data on natural products (NPs) that can potentially treat IBD, including ulcerative colitis. The main actions of these NPs relate to their effects on nuclear factor kappa B (NF-κB), the Janus kinase (JAK)/signal transducer and activator of transcription (STAT) signaling pathway, the regulation of T helper 17/regulatory T cells balance, and oxidative stress. The ability of these NPs to inhibit intestinal inflammation appears to be dependent on lowering levels of the pro-inflammatory cytokines tumor necrosis factor-alpha (TNF-α), interleukin (IL)-1β, and IL-17, via the Jun N-terminal kinase (JNK)1, NF-κβ-p65, and STAT3 pathways. In addition, NPs were shown to reduce oxidative stress and the severity of ulcerative colitis, as well as increase the activity of antioxidant enzymes. These actions suggest that NPs represent a promising treatment for IBD, and potentially have greater efficacy and safety than current treatments.
Collapse
Affiliation(s)
- Gopalsamy Rajiv Gandhi
- Division of Phytochemistry and Drug Design, Department of Biosciences, Rajagiri College of Social Sciences (Autonomous), Kalamaserry, Kochi, 683104, Kerala, India
| | - Thiruchenduran Mohana
- Department of Biochemistry, Meenakshi Ammal Dental College and Hospital (MAHER), Maduravoyal, 600095, Chennai, Tamil Nadu, India
| | - Kumaraswamy Athesh
- Department of Biochemistry, Srimad Andavan Arts and Science College (Autonomous), Affiliated to Bharathidasan University, Tiruchirapalli, 620005, Tamil Nadu, India
| | - Varghese Edwin Hillary
- Division of Plant Molecular Biology and Biotechnology, Department of Biosciences, Rajagiri College of Social Sciences (Autonomous), Kalamaserry, Kochi, 683104, Kerala, India
| | - Alan Bruno Silva Vasconcelos
- Laboratory of Biology and Immunology of Cancer and Leishmania, Department of Morphology, Federal University of Sergipe, São Cristóvão, Sergipe, Brazil
| | - Mariana Nobre Farias de Franca
- Laboratory of Biology and Immunology of Cancer and Leishmania, Department of Morphology, Federal University of Sergipe, São Cristóvão, Sergipe, Brazil
- Postgraduate Program in Health Sciences (PPGCS), Federal University of Sergipe (UFS), Campus Prof. João Cardoso Nascimento, Aracaju, CEP 49060.108, Sergipe, Brazil
| | - Monalisa Martins Montalvão
- Laboratory of Biology and Immunology of Cancer and Leishmania, Department of Morphology, Federal University of Sergipe, São Cristóvão, Sergipe, Brazil
- Postgraduate Program in Health Sciences (PPGCS), Federal University of Sergipe (UFS), Campus Prof. João Cardoso Nascimento, Aracaju, CEP 49060.108, Sergipe, Brazil
| | - Stanislaus Antony Ceasar
- Division of Plant Molecular Biology and Biotechnology, Department of Biosciences, Rajagiri College of Social Sciences (Autonomous), Kalamaserry, Kochi, 683104, Kerala, India
| | - Gnanasekaran Jothi
- Department of Biochemistry, Srimad Andavan Arts and Science College (Autonomous), Affiliated to Bharathidasan University, Tiruchirapalli, 620005, Tamil Nadu, India
| | - Gurunagarajan Sridharan
- Department of Biochemistry, Srimad Andavan Arts and Science College (Autonomous), Affiliated to Bharathidasan University, Tiruchirapalli, 620005, Tamil Nadu, India
| | - Ricardo Queiroz Gurgel
- Postgraduate Program in Health Sciences (PPGCS), Federal University of Sergipe (UFS), Campus Prof. João Cardoso Nascimento, Aracaju, CEP 49060.108, Sergipe, Brazil
| | - Baojun Xu
- Programme of Food Science and Technology, Department of Life Sciences, BNU-HKBU United International College, Zhuhai, Guangdong, 519087, China
| |
Collapse
|
34
|
Liu J, Li T, Zhang S, Lu E, Qiao W, Chen H, Liu P, Tang X, Cheng T, Chen H. Proteomic and single-cell analysis shed new light on the anti-inflammatory role of interferonβ in chronic periodontitis. Front Pharmacol 2023; 14:1232539. [PMID: 37876725 PMCID: PMC10590904 DOI: 10.3389/fphar.2023.1232539] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 09/26/2023] [Indexed: 10/26/2023] Open
Abstract
Periodontitis, a condition that results in periodontal attachment loss and alveolar bone resorption, contributes to the global burden of oral disease. The underlying mechanism of periodontitis involves the dysbiosis and dyshomeostasis between host and oral microbes, among which the macrophage is one of the major innate immune cell players, producing interferon β (IFNβ) in response to bacterial infection. The objective of this research was to examine the interaction of macrophages with periodontitis and the role and mechanism of IFNβ on macrophages. IFNβ has been shown to have the potential to induce the differentiation of M1 to M2 macrophages, which are stimulated by low levels of lipopolysaccharide (LPS). Additionally, IFNβ has been demonstrated to promote the production of ISG15 by macrophages, which leads to the inhibition of the innate immune response. Moreover, our investigation revealed that IFNβ has the potential to augment the secretion of ISG15 and its downstream cytokine, IL10, in LPS-stimulated macrophages. Single-cell analysis was conducted on the gingival tissues of patients with periodontitis, which revealed a higher proportion of macrophages in the periodontitis-diseased tissue and increased expression of IFNβ, ISG15, and IL10. Gene Set Enrichment Analysis indicated that bacterial infection was associated with upregulation of IFNβ, ISG15, and IL10. Notably, only IL10 has been linked to immunosuppression, indicating that the IFNβ-ISG15-IL10 axis might promote an anti-inflammatory response in periodontitis through IL10 expression. It is also found that macrophage phenotype transitions in periodontitis involve the release of higher levels of IFNβ, ISG15, and IL10 by the anti-inflammatory M2 macrophage phenotype compared to the pro-inflammatory M1 phenotype and myeloid-derived suppressor cells (MDSCs). This implies that the IFNβ-induced production of IL10 might be linked to the M2 macrophage phenotype. Furthermore, cell communication analysis demonstrated that IL10 can promote fibroblast proliferation in periodontal tissues via STAT3 signaling.
Collapse
Affiliation(s)
- Jieying Liu
- Department of Medical Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Tianle Li
- Faculty of Dentistry, Prince Philip Dental Hospital, University of Hong Kong, Pokfulam, China
| | - Shunhao Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Eryi Lu
- Department of Stomatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei Qiao
- Faculty of Dentistry, Prince Philip Dental Hospital, University of Hong Kong, Pokfulam, China
| | - Huimin Chen
- Faculty of Dentistry, Prince Philip Dental Hospital, University of Hong Kong, Pokfulam, China
| | - Peng Liu
- Department of Medical Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xiaoyue Tang
- Department of Medical Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Tianfan Cheng
- Faculty of Dentistry, Prince Philip Dental Hospital, University of Hong Kong, Pokfulam, China
| | - Hui Chen
- Faculty of Dentistry, Prince Philip Dental Hospital, University of Hong Kong, Pokfulam, China
| |
Collapse
|
35
|
Molina-Jiménez F, Ugalde-Triviño L, Arias-González L, Relaño-Rupérez C, Casabona S, Pérez-Fernández MT, Martín-Domínguez V, Fernández-Pacheco J, Laserna-Mendieta EJ, Muñoz-Hernández P, Arias-Arias Á, Cano A, Muñoz J, Lucendo AJ, Santander C, Majano P. Proteomic analysis of the esophageal epithelium reveals key features of eosinophilic esophagitis pathophysiology. Allergy 2023; 78:2732-2744. [PMID: 37287363 DOI: 10.1111/all.15779] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 04/27/2023] [Accepted: 05/03/2023] [Indexed: 06/09/2023]
Abstract
BACKGROUND Eosinophilic esophagitis (EoE) is a chronic non-IgE-mediated allergic disease of the esophagus. An unbiased proteomics approach was performed to investigate pathophysiological changes in esophageal epithelium. Additionally, an RNAseq-based transcriptomic analysis in paired samples was also carried out. METHODS Total proteins were purified from esophageal endoscopic biopsies in a cohort of adult EoE patients (n = 25) and healthy esophagus controls (n = 10). Differentially accumulated (DA) proteins in EoE patients compared to control tissues were characterized to identify altered biological processes and signaling pathways. Results were also compared with a quantitative proteome dataset of the human esophageal mucosa. Next, results were contrasted with those obtained after RNAseq analysis in paired samples. Finally, we matched up protein expression with two EoE-specific mRNA panels (EDP and Eso-EoE panel). RESULTS A total of 1667 proteins were identified, of which 363 were DA in EoE. RNA sequencing in paired samples identified 1993 differentially expressed (DE) genes. Total RNA and protein levels positively correlated, especially in DE mRNA-proteins pairs. Pathway analysis of these proteins in EoE showed alterations in immune and inflammatory responses for the upregulated proteins, and in epithelial differentiation, cornification and keratinization in those downregulated. Interestingly, a set of DA proteins, including eosinophil-related and secreted proteins, were not detected at the mRNA level. Protein expression positively correlated with EDP and Eso-EoE, and corresponded with the most abundant proteins of the human esophageal proteome. CONCLUSIONS We unraveled for the first time key proteomic features involved in EoE pathogenesis. An integrative analysis of transcriptomic and proteomic datasets provides a deeper insight than transcriptomic alone into understanding complex disease mechanisms.
Collapse
Affiliation(s)
- Francisca Molina-Jiménez
- Molecular Biology Unit, Hospital Universitario de la Princesa, Madrid, Spain
- Instituto de Investigación Sanitaria Hospital Universitario de La Princesa (IIS-Princesa), Madrid, Spain
| | - Lola Ugalde-Triviño
- Molecular Biology Unit, Hospital Universitario de la Princesa, Madrid, Spain
- Instituto de Investigación Sanitaria Hospital Universitario de La Princesa (IIS-Princesa), Madrid, Spain
| | - Laura Arias-González
- Instituto de Investigación Sanitaria Hospital Universitario de La Princesa (IIS-Princesa), Madrid, Spain
- Department of Gastroenterology, Hospital General de Tomelloso, Ciudad Real, Spain
- Instituto de Investigación Sanitaria de Castilla-La Mancha (IDISCAM), Toledo, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
| | - Carlos Relaño-Rupérez
- Molecular Biology Unit, Hospital Universitario de la Princesa, Madrid, Spain
- Instituto de Investigación Sanitaria Hospital Universitario de La Princesa (IIS-Princesa), Madrid, Spain
| | - Sergio Casabona
- Instituto de Investigación Sanitaria Hospital Universitario de La Princesa (IIS-Princesa), Madrid, Spain
- Department of Gastroenterology, Hospital Universitario de La Princesa, Madrid, Spain
| | - María Teresa Pérez-Fernández
- Instituto de Investigación Sanitaria Hospital Universitario de La Princesa (IIS-Princesa), Madrid, Spain
- Department of Gastroenterology, Hospital Universitario de La Princesa, Madrid, Spain
| | - Verónica Martín-Domínguez
- Instituto de Investigación Sanitaria Hospital Universitario de La Princesa (IIS-Princesa), Madrid, Spain
- Department of Gastroenterology, Hospital Universitario de La Princesa, Madrid, Spain
| | - Jennifer Fernández-Pacheco
- Instituto de Investigación Sanitaria Hospital Universitario de La Princesa (IIS-Princesa), Madrid, Spain
- Department of Gastroenterology, Hospital Universitario de La Princesa, Madrid, Spain
| | - Emilio José Laserna-Mendieta
- Instituto de Investigación Sanitaria Hospital Universitario de La Princesa (IIS-Princesa), Madrid, Spain
- Department of Gastroenterology, Hospital General de Tomelloso, Ciudad Real, Spain
- Clinical Laboratory, Hospital Universitario de La Princesa, Madrid, Spain
| | | | - Ángel Arias-Arias
- Instituto de Investigación Sanitaria Hospital Universitario de La Princesa (IIS-Princesa), Madrid, Spain
- Instituto de Investigación Sanitaria de Castilla-La Mancha (IDISCAM), Toledo, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
- Research Unit, Hospital General La Mancha Centro, Alcázar de San Juan, Spain
| | - Ainara Cano
- Food Research, AZTI, Basque Research and Technology Alliance (BRTA), Parque Tecnológico de Bizkaia, Derio, Spain
| | - Javier Muñoz
- Cell Signalling and Clinical Proteomics Group, Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
- Ikerbasque, Basque Foundation for Science, Bilbao, Spain
| | - Alfredo J Lucendo
- Instituto de Investigación Sanitaria Hospital Universitario de La Princesa (IIS-Princesa), Madrid, Spain
- Department of Gastroenterology, Hospital General de Tomelloso, Ciudad Real, Spain
- Instituto de Investigación Sanitaria de Castilla-La Mancha (IDISCAM), Toledo, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
| | - Cecilio Santander
- Instituto de Investigación Sanitaria Hospital Universitario de La Princesa (IIS-Princesa), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
- Department of Gastroenterology, Hospital Universitario de La Princesa, Madrid, Spain
| | - Pedro Majano
- Molecular Biology Unit, Hospital Universitario de la Princesa, Madrid, Spain
- Instituto de Investigación Sanitaria Hospital Universitario de La Princesa (IIS-Princesa), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
- Department of Cellular Biology, Faculty of Biology, Universidad Complutense de Madrid, Madrid, Spain
| |
Collapse
|
36
|
Wegner E, Mickan T, Truffel S, Slotina E, Müller L, Wunderlich F, Harper A, Ritz U, Rommens PM, Gercek E, Drees P, Baranowski A. The effect of losartan on the development of post-traumatic joint stiffness in a rat model. Biomed Pharmacother 2023; 166:115291. [PMID: 37557010 DOI: 10.1016/j.biopha.2023.115291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Revised: 07/27/2023] [Accepted: 08/04/2023] [Indexed: 08/11/2023] Open
Abstract
Post-traumatic joint stiffness (PTJS) is accompanied by a multidimensional disturbance of joint architecture. Pharmacological approaches represent promising alternatives as the traumatic nature of current therapeutic standards may lead to PTJS' progression. Losartan is an auspicious candidate, as it has demonstrated an antifibrotic effect in other organs. Forty-eight Sprague Dawley rats were randomized into equally sized losartan or control groups. After a standardized knee trauma, the joint was immobilized for either 2 weeks (n = 16), 4 weeks (n = 16) or 4 weeks with re-mobilization for an additional 4 weeks (n = 16). Pharmacotherapy with losartan or placebo (30 mg/kg/day) was initiated on the day of trauma and continued for the entire course. Joint contracture was measured alongside histological and molecular biological assessments. There were no significant biomechanical changes in joint contracture over time, comparing short-term (2 weeks) with long-term losartan therapy (4 weeks). However, comparing the formation of PTJS with that of the control, there was a trend toward improvement of joint mobility of 10.5° (p 0.09) under the influence of losartan. During the re-mobilization phase, no significant effect of losartan on range of motion (ROM) was demonstrated. At a cellular level, losartan significantly reduced myofibroblast counts by up to 72 % (4 weeks, p ≤ 0.001) without effecting the capsular configuration. Differences in expression levels of profibrotic factors (TGF-β, CTGF, Il-6) were most pronounced at week 4. The antifibrotic properties of losartan are not prominent enough to completely prevent the development of PTJS after severe joint injury.
Collapse
Affiliation(s)
- Erik Wegner
- Department of Orthopaedics and Traumatology, Biomatics Group, University Medical Center of the Johannes Gutenberg University, Mainz 55131, Germany
| | - Tim Mickan
- Department of Orthopaedics and Traumatology, Biomatics Group, University Medical Center of the Johannes Gutenberg University, Mainz 55131, Germany
| | - Sebastian Truffel
- Department of Orthopaedics and Traumatology, Biomatics Group, University Medical Center of the Johannes Gutenberg University, Mainz 55131, Germany
| | - Ekaterina Slotina
- Department of Orthopaedics and Traumatology, Biomatics Group, University Medical Center of the Johannes Gutenberg University, Mainz 55131, Germany
| | - Lukas Müller
- Department of Diagnostic and Interventional Radiology, University Medical Center of the Johannes Gutenberg University, Mainz 55131, Germany; Mainz Research School of Translational Biomedicine, University Medical Center of the Johannes Gutenberg University, Mainz 55131, Germany
| | - Felix Wunderlich
- Department of Orthopaedics and Traumatology, Biomatics Group, University Medical Center of the Johannes Gutenberg University, Mainz 55131, Germany
| | - Austin Harper
- St. George's University School of Medicine, True Blue, St. George, Grenada
| | - Ulrike Ritz
- Department of Orthopaedics and Traumatology, Biomatics Group, University Medical Center of the Johannes Gutenberg University, Mainz 55131, Germany
| | - Pol M Rommens
- Department of Orthopaedics and Traumatology, Biomatics Group, University Medical Center of the Johannes Gutenberg University, Mainz 55131, Germany
| | - Erol Gercek
- Department of Orthopaedics and Traumatology, Biomatics Group, University Medical Center of the Johannes Gutenberg University, Mainz 55131, Germany
| | - Philipp Drees
- Department of Orthopaedics and Traumatology, Biomatics Group, University Medical Center of the Johannes Gutenberg University, Mainz 55131, Germany
| | - Andreas Baranowski
- Department of Orthopaedics and Traumatology, Biomatics Group, University Medical Center of the Johannes Gutenberg University, Mainz 55131, Germany.
| |
Collapse
|
37
|
ZHOU H, LI H, WANG H. Potential protective effects of the water-soluble Chinese propolis on experimental ulcerative colitis. J TRADIT CHIN MED 2023; 43:925-933. [PMID: 37679980 PMCID: PMC10465833 DOI: 10.19852/j.cnki.jtcm.20230727.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Accepted: 09/24/2022] [Indexed: 09/09/2023]
Abstract
OBJECTIVE To investigate the outcome of Chinese water-soluble propolis (WSP) on the inflammatory response and oxidative stress (OS) of colonic mucosa in rats with ulcerative colitis. METHODS Dextran sulfate sodium (DSS) was employed to establish the ucerative colitis (UC) rat model. Forty-eight male rats were arbitrarily separated into six groups, namely control, UC, low-dose water-soluble propolis (L-WSP), medium-dose water-soluble propolis (M-WSP), high-dose water-soluble propolis (H-WSP), and sulfasalazine (Sulfa). In this study, we adopted a method of pre-administration and reconstruction of the model that assessed the water-soluble propolis mediated protection against DSS-induced UC rats. Moreover, we examined the body weight (BW), disease activity index (DAI), bloody stool, colon length, and intestinal mucosal injury index of rats. In addition, using enzyme linked immunosorbent assays, we assessed indicators, such as, colonic myeloperoxidase (MPO), interleukin-6 (IL-6), interleukin-9 (IL-9), tumor necrosis factor-ɑ (TNF-ɑ), superoxide dismutase (SOD), malondialdehyde, and glutathione peroxidase (GSH-Px) levels. RESULTS The pro-inflammatory cytokine expression, as well as OS, was increased in the model rats. However, upon WSP intervention, both pro-inflammatory cytokine levels and OS reduced dramatically, and the therapeutic effect was dose-dependent. CONCLUSION WSP downregulates OS by enhancing the function of endogenous antioxidant enzymes like SOD and GSH-Px, that inhibit neutrophil activity, as well as diminish pro-inflammatory cytokines like TNF-ɑ, IL-6, and IL-9, along with mechanisms that attenuate intestinal inflammation in UC rat model.
Collapse
Affiliation(s)
- Hua ZHOU
- 1 Department of Physiology, Anhui Medical College, Hefei 230601, China
| | - Hui LI
- 2 Department of Physiology, Wannan Medical College, Wuhu 241002, China
| | - Haihua WANG
- 2 Department of Physiology, Wannan Medical College, Wuhu 241002, China
| |
Collapse
|
38
|
Yu JT, Fan S, Li XY, Hou R, Hu XW, Wang JN, Shan RR, Dong ZH, Xie MM, Dong YH, Shen XY, Jin J, Wen JG, Liu MM, Wang W, Meng XM. Novel insights into STAT3 in renal diseases. Biomed Pharmacother 2023; 165:115166. [PMID: 37473682 DOI: 10.1016/j.biopha.2023.115166] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 07/07/2023] [Accepted: 07/11/2023] [Indexed: 07/22/2023] Open
Abstract
Signal transducer and activator of transcription 3 (STAT3) is a cell-signal transcription factor that has attracted considerable attention in recent years. The stimulation of cytokines and growth factors can result in the transcription of a wide range of genes that are crucial for several cellular biological processes involved in pro- and anti-inflammatory responses. STAT3 has attracted considerable interest as a result of a recent upsurge in study because of their role in directing the innate immune response and sustaining inflammatory pathways, which is a key feature in the pathogenesis of many diseases, including renal disorders. Several pathological conditions which may involve STAT3 include diabetic nephropathy, acute kidney injury, lupus nephritis, polycystic kidney disease, and renal cell carcinoma. STAT3 is expressed in various renal tissues under these pathological conditions. To better understand the role of STAT3 in the kidney and provide a theoretical foundation for STAT3-targeted therapy for renal disorders, this review covers the current work on the activities of STAT3 and its mechanisms in the pathophysiological processes of various types of renal diseases.
Collapse
Affiliation(s)
- Ju-Tao Yu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, the Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Shuai Fan
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei 230032 China; Department of Urology, Institute of Urology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei 230032 China
| | - Xiang-Yu Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, the Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Rui Hou
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, the Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Xiao-Wei Hu
- Department of Clinical Pharmacy, Anhui Provincial Children's Hospital, Hefei 230051, China
| | - Jia-Nan Wang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, the Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Run-Run Shan
- School of Life Sciences, Anhui Medical University, Hefei 230032, China
| | - Ze-Hui Dong
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, the Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Man-Man Xie
- School of Life Sciences, Anhui Medical University, Hefei 230032, China
| | - Yu-Hang Dong
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, the Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Xiao-Yu Shen
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, the Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Juan Jin
- Department of Pharmacology, School of Basic Medical Sciences, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei 230032, China
| | - Jia-Gen Wen
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, the Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Ming-Ming Liu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, the Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Wei Wang
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei 230032 China; Department of Urology, Institute of Urology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei 230032 China.
| | - Xiao-Ming Meng
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, the Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China.
| |
Collapse
|
39
|
Saleh DO, El-Nasr NMEA, Fayez AM, Ahmed KA, Mohamed RA. Uro-protective role of chrysin against cyclophosphamide-induced hemorrhagic cystitis in rats involving the turning-off NF-κB/P38-MAPK, NO/PARP-1 and STAT-3 signaling cascades. Chem Biol Interact 2023; 382:110585. [PMID: 37263553 DOI: 10.1016/j.cbi.2023.110585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 05/06/2023] [Accepted: 05/29/2023] [Indexed: 06/03/2023]
Abstract
BACKGROUND Chemotherapeutic agents are used to treat a wide range of cancer types, but they cause serious side effects which must be managed after treatment. Cyclophosphamide (CYP) is one of chemotherapeutic drugs that causes hemorrhagic cystitis (HC) induced by acrolein. OBJECTIVE The current investigation intended to uncover the role of chrysin (CHR) in CYP-induced HC in rats and explore the signaling pathway beyond this effect. ANALYSIS process: A single dose of CYP (200 mg/kg/IP) was injected, meanwhile CHR (25, 50 and 100 mg/kg, P.O) was administered respectively for 7 days prior to CYP administration and resume for 7 days afterwards. Urinary bladder tissue was then isolated from all rats to assess oxidative stress and inflammatory biomarkers. Moreover, histopathological examinations were performed. RESULTS Treatment with CHR showed a marked alleviation in oxidative stress biomarkers induced by CYP. Furthermore, CHR treatment presented a dose-dependent boost in the anti-inflammatory; IL-10 levels and a drop in the pro-inflammatory biomarkers; IL-1β, IL-6, and TNF-α. Additionally, stabilization of the PARP-1 protein expression was also detected thus preventing DNA damage. Similarly, CHR restored the urinary bladder cGMP levels. Notably, CHR treatment was accompanied with inhibition in NF-κB/p38-MAPK, NO/PARP-1 and STAT-3 signaling pathways inflammatory cascades. All these findings conformed with the histopathological examinations as well as iNOS immunostaining in the urinary bladder tissue. CONCLUSION Co-administration of CHR and CYP attained uro-protective therapeutic potential to guard against HC as well as spot the tangled mechanism of CHR in attenuating the HC induced by CYP.
Collapse
Affiliation(s)
- Dalia O Saleh
- Pharmacology Department, Medical Research and Clinical Studies Institute, National Research Centre, Giza, Egypt.
| | - Nesma M E Abo El-Nasr
- Pharmacology Department, Medical Research and Clinical Studies Institute, National Research Centre, Giza, Egypt
| | - Ahmed M Fayez
- Pharmacology and Toxicology Department, School of Life and Medical Sciences, University of Hertfordshire Hosted by Global Academic Foundation, New Administrative Capital, Cairo, Egypt
| | - Kawkab A Ahmed
- Pathology Department, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt
| | - Reem A Mohamed
- Department of Pharmacology, Faculty of Pharmacy, October University for Modern Sciences and Arts (MSA), Giza, Egypt
| |
Collapse
|
40
|
Cucinotta L, Mannino D, Casili G, Repici A, Crupi L, Paterniti I, Esposito E, Campolo M. Prolyl oligopeptidase inhibition ameliorates experimental pulmonary fibrosis both in vivo and in vitro. Respir Res 2023; 24:211. [PMID: 37626373 PMCID: PMC10463606 DOI: 10.1186/s12931-023-02519-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 08/21/2023] [Indexed: 08/27/2023] Open
Abstract
BACKGROUND Pulmonary fibrosis is a progressive disease characterized by lung remodeling due to excessive deposition of extracellular matrix. Although the etiology remains unknown, aberrant angiogenesis and inflammation play an important role in the development of this pathology. In this context, recent scientific research has identified new molecules involved in angiogenesis and inflammation, such as the prolyl oligopeptidase (PREP), a proteolytic enzyme belonging to the serine protease family, linked to the pathology of many lung diseases such as pulmonary fibrosis. Therefore, the aim of this study was to investigate the effect of a selective inhibitor of PREP, known as KYP-2047, in an in vitro and in an in vivo model of pulmonary fibrosis. METHODS The in vitro model was performed using human alveolar A549 cells. Cells were exposed to lipopolysaccharide (LPS) 10 μg/ml and then, cells were treated with KYP-2047 at the concentrations of 1 μM, 10 μM and 50 μM. Cell viability was evaluated by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium (MTT) bromide colorimetric assay, while inflammatory protein expression was assessed by western blots analysis. The in vivo model was induced in mice by intra-tracheal administration of bleomycin (1 mg/kg) and then treated intraperitoneally with KYP-2047 at doses of 1, 2.5 and 5 mg/kg once daily for 12 days and then mice were sacrificed, and lung tissues were collected for analyses. RESULTS The in vitro results demonstrated that KYP-2047 preserved cell viability, reduced inflammatory process by decreasing IL-18 and TNF-α, and modulated lipid peroxidation as well as nitrosative stress. The in vivo pulmonary fibrosis has demonstrated that KYP-2047 was able to restore histological alterations reducing lung injury. Our data demonstrated that KYP-2047 significantly reduced angiogenesis process and the fibrotic damage modulating the expression of fibrotic markers. Furthermore, KYP-2047 treatment modulated the IκBα/NF-κB pathway and reduced the expression of related pro-inflammatory enzymes and cytokines. Moreover, KYP-2047 was able to modulate the JAK2/STAT3 pathway, highly involved in pulmonary fibrosis. CONCLUSION In conclusion, this study demonstrated the involvement of PREP in the pathogenesis of pulmonary fibrosis and that its inhibition by KYP-2047 has a protective role in lung injury induced by BLM, suggesting PREP as a potential target therapy for pulmonary fibrosis. These results speculate the potential protective mechanism of KYP-2047 through the modulation of JAK2/STAT3 and NF-κB pathways.
Collapse
Affiliation(s)
- Laura Cucinotta
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 7 Viale Ferdinando Stagno D'Alcontres, 31-98166, Messina, Italy
| | - Deborah Mannino
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 7 Viale Ferdinando Stagno D'Alcontres, 31-98166, Messina, Italy
| | - Giovanna Casili
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 7 Viale Ferdinando Stagno D'Alcontres, 31-98166, Messina, Italy
| | - Alberto Repici
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 7 Viale Ferdinando Stagno D'Alcontres, 31-98166, Messina, Italy
| | - Lelio Crupi
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 7 Viale Ferdinando Stagno D'Alcontres, 31-98166, Messina, Italy
| | - Irene Paterniti
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 7 Viale Ferdinando Stagno D'Alcontres, 31-98166, Messina, Italy
| | - Emanuela Esposito
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 7 Viale Ferdinando Stagno D'Alcontres, 31-98166, Messina, Italy.
| | - Michela Campolo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 7 Viale Ferdinando Stagno D'Alcontres, 31-98166, Messina, Italy
| |
Collapse
|
41
|
Peralta S, Grenier JK, Webb SM, Miller AD, Miranda IC, Parker JSL. Transcriptomic signatures of feline chronic gingivostomatitis are influenced by upregulated IL6. Sci Rep 2023; 13:13437. [PMID: 37596310 PMCID: PMC10439118 DOI: 10.1038/s41598-023-40679-4] [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: 04/23/2023] [Accepted: 08/16/2023] [Indexed: 08/20/2023] Open
Abstract
Feline chronic gingivostomatitis (FCGS) is a relatively common and debilitating disease characterized by bilateral inflammation and ulceration of the caudal oral mucosa, alveolar and buccal mucosa, and varying degrees of periodontal disease. The etiopathogenesis of FCGS remains unresolved. In this study, we performed bulk RNA-seq molecular profiling of affected tissues derived from a cohort of client-owned cats with FCGS compared to tissues from unaffected animals, to identify candidate genes and pathways that can help guide future exploration of novel clinical solutions. We complemented transcriptomic findings with immunohistochemistry and in situ hybridization assays to better understand the biological significance of the results and performed RNA-seq validation of biologically relevant differentially expressed genes using qPCR assays to demonstrate technical reproducibility. Transcriptomic profiles of oral mucosal tissues in cats with FCGS are enriched with immune- and inflammation-related genes and pathways that appear to be largely influenced by IL6, and include NFKB, JAK/STAT, IL-17 and IFN type I and II signaling, offering new opportunities to develop novel clinical applications based on a more rational understanding of the disease.
Collapse
Affiliation(s)
- Santiago Peralta
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA.
- Clinical Programs Center, College of Veterinary Medicine, Cornell University, Box 31, Ithaca, NY, 14853, USA.
| | - Jennifer K Grenier
- Transcriptional Regulation and Expression Facility, Biotechnology Resource Center, Institute of Biotechnology, Cornell University, Ithaca, NY, 14853, USA
| | - Suzin M Webb
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA
| | - Andrew D Miller
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA
| | - Ileana C Miranda
- Laboratory of Comparative Pathology, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, and The Rockefeller University, New York, NY, 10065, USA
| | - John S L Parker
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA
| |
Collapse
|
42
|
Xu F, Lu S, Jia X, Zhou Y. Bromodomain protein 4 mediates the roles of TGFβ1-induced Stat3 signaling in mouse liver fibrogenesis. Toxicol Lett 2023; 385:42-50. [PMID: 37634812 DOI: 10.1016/j.toxlet.2023.08.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 07/30/2023] [Accepted: 08/24/2023] [Indexed: 08/29/2023]
Abstract
Epigenetic reader Bromodomain protein 4 (BrD4) functions as a global genomic regulator to direct hepatic stellate cell (HSC) activation (a key step in liver fibrogenesis) and liver fibrosis. The pivotal pro-fibrotic cytokine transforming growth factor-β1 (TGFβ1) signals through both Smad and Stat3 to elicit a wide array of biological effects. Stat3 is widely acknowledged as a regulator of gene transcription and is involved in fibrosis of multiple tissues. The present study focused on BrD4 function implication in the roles of TGFβ1-induced Stat3 signaling in HSC activation and liver fibrosis by using heterozygous TGFβ1 knockout mice and HSC culture. Results showed that Stat3 was required for TGFβ1-induced BrD4 expression in HSCs. BrD4 expression paralleled Stat3 activation in activated HSCs in human cirrhotic livers. BrD4 was involved in the roles of TGFβ1-induced Stat3 in HSC activation and liver fibrogenesis. Smad3 bound to phosphorylated-Stat3 and contributed to TGFβ1-induced Stat3 signaling. BrD4 expression induced by Stat3 signaling required the early-immediate gene Egr1. Egr1 had a positive feedback on Stat3 activation. In conclusion, a network consisting of Stat3 signaling, Smad3 signaling, Egr1, and BrD4 was involved in the effects of TGFβ1 on liver fibrosis, providing new toxicological mechanisms for TGFβ1 in liver fibrogenesis.
Collapse
Affiliation(s)
- Feifan Xu
- Department of Clinical Laboratory, Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), 500 Yonghe Road, Nantong 226011, Jiangsu, China
| | - Sidan Lu
- Department of Biochemistry & Molecular Biology, Medical School, Nantong University, Qi xiou Road 19, Nantong 226001, Jiangsu, China
| | - Xin Jia
- Department of Biochemistry & Molecular Biology, Medical School, Nantong University, Qi xiou Road 19, Nantong 226001, Jiangsu, China
| | - Yajun Zhou
- Department of Biochemistry & Molecular Biology, Medical School, Nantong University, Qi xiou Road 19, Nantong 226001, Jiangsu, China.
| |
Collapse
|
43
|
Palomino Lago E, Jelbert ER, Baird A, Lam PY, Guest DJ. Equine induced pluripotent stem cells are responsive to inflammatory cytokines before and after differentiation into musculoskeletal cell types. In Vitro Cell Dev Biol Anim 2023; 59:514-527. [PMID: 37582999 PMCID: PMC10520172 DOI: 10.1007/s11626-023-00800-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 07/19/2023] [Indexed: 08/17/2023]
Abstract
Persistent inflammation is associated with the poor regeneration of musculoskeletal tissues. Embryonic stem cells (ESCs) have an attenuated response to inflammatory cytokines, but there are mixed reports on the response of induced pluripotent stem cells (iPSCs) to inflammation. Horses provide a relevant large animal model for studying musculoskeletal tissue diseases and the testing of novel therapies. The aim of this study was to determine if equine iPSCs are responsive to the inflammatory cytokines IL-1β, TNFα and IFN-γ in their undifferentiated state, or following differentiation into tendon and cartilage-like cells. We demonstrated that in undifferentiated iPSCs, the cytokines induce NF-κB P65 and STAT1 nuclear translocation which leads to cell death, decreased OCT4 expression and increased expression of inflammatory genes. Following differentiation towards cartilage-like cells exposure to the cytokines resulted in STAT1 nuclear translocation, changes in cartilage gene expression and increased expression of matrix metalloproteinases (MMPs) and inflammatory genes. Exposure of iPSC-derived tendon-like cells to the cytokines resulted nuclear translocation of NF-κB P65 and STAT1, altered tendon gene expression, increased MMP expression and increased expression of inflammatory genes. Equine iPSCs are therefore capable of responding to inflammatory stimulation and this may have relevance for their future clinical application.
Collapse
Affiliation(s)
- Esther Palomino Lago
- Department of Clinical Sciences and Services, The Royal Veterinary College, Hawkshead Lane, North Mymms, Hatfield, AL9 7TA, Herts, UK
| | - Elizabeth R Jelbert
- Department of Clinical Sciences and Services, The Royal Veterinary College, Hawkshead Lane, North Mymms, Hatfield, AL9 7TA, Herts, UK
| | - Arabella Baird
- Animal Health Trust, Lanwades Park, Kentford, Newmarket, CB8 7UU, UK
| | - Pak Y Lam
- Department of Clinical Sciences and Services, The Royal Veterinary College, Hawkshead Lane, North Mymms, Hatfield, AL9 7TA, Herts, UK
| | - Deborah J Guest
- Department of Clinical Sciences and Services, The Royal Veterinary College, Hawkshead Lane, North Mymms, Hatfield, AL9 7TA, Herts, UK.
| |
Collapse
|
44
|
Scortegagna M, Du Y, Bradley LM, Wang K, Molinolo A, Ruppin E, Murad R, Ronai ZA. Ubiquitin Ligases Siah1a/2 Control Alveolar Macrophage Functions to Limit Carcinogen-Induced Lung Adenocarcinoma. Cancer Res 2023; 83:2016-2033. [PMID: 37078793 PMCID: PMC10330299 DOI: 10.1158/0008-5472.can-23-0258] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 03/22/2023] [Accepted: 04/17/2023] [Indexed: 04/21/2023]
Abstract
Cellular components of the tumor microenvironment, including myeloid cells, play important roles in the progression of lung adenocarcinoma (LUAD) and its response to therapy. Here, we characterize the function of the ubiquitin ligases Siah1a/2 in regulating the differentiation and activity of alveolar macrophages (AM) and assess the implication of Siah1a/2 control of AMs for carcinogen-induced LUAD. Macrophage-specific genetic ablation of Siah1a/2 promoted accumulation of AMs with an immature phenotype and increased expression of protumorigenic and pro-inflammatory Stat3 and β-catenin gene signatures. Administration of urethane to wild-type mice promoted enrichment of immature-like AMs and lung tumor development, which was enhanced by macrophage-specific Siah1a/2 ablation. The profibrotic gene signature seen in Siah1a/2-ablated immature-like macrophages was associated with increased tumor infiltration of CD14+ myeloid cells and poorer survival of patients with LUAD. Single-cell RNA-seq confirmed the presence of a cluster of immature-like AMs expressing a profibrotic signature in lungs of patients with LUAD, a signature enhanced in smokers. These findings identify Siah1a/2 in AMs as gatekeepers of lung cancer development. SIGNIFICANCE The ubiquitin ligases Siah1a/2 control proinflammatory signaling, differentiation, and profibrotic phenotypes of alveolar macrophages to suppress lung carcinogenesis.
Collapse
Affiliation(s)
- Marzia Scortegagna
- NCI-designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla CA
| | - Yuanning Du
- NCI-designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla CA
| | - Linda M. Bradley
- NCI-designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla CA
| | - Kun Wang
- Cancer Data Science Laboratory, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | | | - Eytan Ruppin
- Cancer Data Science Laboratory, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Rabi Murad
- NCI-designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla CA
| | - Ze’ev A. Ronai
- NCI-designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla CA
| |
Collapse
|
45
|
Bonente D, Bianchi L, De Salvo R, Nicoletti C, De Benedetto E, Bacci T, Bini L, Inzalaco G, Franci L, Chiariello M, Tosi GM, Bertelli E, Barone V. Co-Expression of Podoplanin and CD44 in Proliferative Vitreoretinopathy Epiretinal Membranes. Int J Mol Sci 2023; 24:ijms24119728. [PMID: 37298679 DOI: 10.3390/ijms24119728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 05/30/2023] [Accepted: 06/01/2023] [Indexed: 06/12/2023] Open
Abstract
Epiretinal membranes (ERMs) are sheets of tissue that pathologically develop in the vitreoretinal interface leading to progressive vision loss. They are formed by different cell types and by an exuberant deposition of extracellular matrix proteins. Recently, we reviewed ERMs' extracellular matrix components to better understand molecular dysfunctions that trigger and fuel the onset and development of this disease. The bioinformatics approach we applied delineated a comprehensive overview on this fibrocellular tissue and on critical proteins that could really impact ERM physiopathology. Our interactomic analysis proposed the hyaluronic-acid-receptor cluster of differentiation 44 (CD44) as a central regulator of ERM aberrant dynamics and progression. Interestingly, the interaction between CD44 and podoplanin (PDPN) was shown to promote directional migration in epithelial cells. PDPN is a glycoprotein overexpressed in various cancers and a growing body of evidence indicates its relevant function in several fibrotic and inflammatory pathologies. The binding of PDPN to partner proteins and/or its ligand results in the modulation of signaling pathways regulating proliferation, contractility, migration, epithelial-mesenchymal transition, and extracellular matrix remodeling, all processes that are vital in ERM formation. In this context, the understanding of the PDPN role can help to modulate signaling during fibrosis, hence opening a new line of therapy.
Collapse
Affiliation(s)
- Denise Bonente
- Department of Life Sciences, University of Siena, Via A. Moro 2, 53100 Siena, Italy
- Department of Molecular and Developmental Medicine, University of Siena, Via A. Moro 2, 53100 Siena, Italy
| | - Laura Bianchi
- Section of Functional Proteomics, Department of Life Sciences, University of Siena, Via A. Moro 2, 53100 Siena, Italy
| | - Rossana De Salvo
- Section of Functional Proteomics, Department of Life Sciences, University of Siena, Via A. Moro 2, 53100 Siena, Italy
| | - Claudio Nicoletti
- Department of Molecular and Developmental Medicine, University of Siena, Via A. Moro 2, 53100 Siena, Italy
| | - Elena De Benedetto
- Department of Medicine, Surgery and Neuroscience, University of Siena, Viale Mario Bracci 16, 53100 Siena, Italy
| | - Tommaso Bacci
- Department of Medicine, Surgery and Neuroscience, University of Siena, Viale Mario Bracci 16, 53100 Siena, Italy
| | - Luca Bini
- Section of Functional Proteomics, Department of Life Sciences, University of Siena, Via A. Moro 2, 53100 Siena, Italy
| | - Giovanni Inzalaco
- Core Research Laboratory (CRL), Istituto per lo Studio, la Prevenzione e la Rete Oncologica (ISPRO), Via Fiorentina 1, 53100 Siena, Italy
- Istituto di Fisiologia Clinica (IFC), Consiglio Nazionale delle Ricerche (CNR), Via Fiorentina 1, 53100 Siena, Italy
- Department of Medical Biotechnologies, University of Siena, Viale Mario Bracci 16, 53100 Siena, Italy
| | - Lorenzo Franci
- Core Research Laboratory (CRL), Istituto per lo Studio, la Prevenzione e la Rete Oncologica (ISPRO), Via Fiorentina 1, 53100 Siena, Italy
- Istituto di Fisiologia Clinica (IFC), Consiglio Nazionale delle Ricerche (CNR), Via Fiorentina 1, 53100 Siena, Italy
| | - Mario Chiariello
- Core Research Laboratory (CRL), Istituto per lo Studio, la Prevenzione e la Rete Oncologica (ISPRO), Via Fiorentina 1, 53100 Siena, Italy
- Istituto di Fisiologia Clinica (IFC), Consiglio Nazionale delle Ricerche (CNR), Via Fiorentina 1, 53100 Siena, Italy
| | - Gian Marco Tosi
- Department of Medicine, Surgery and Neuroscience, University of Siena, Viale Mario Bracci 16, 53100 Siena, Italy
| | - Eugenio Bertelli
- Department of Molecular and Developmental Medicine, University of Siena, Via A. Moro 2, 53100 Siena, Italy
| | - Virginia Barone
- Department of Molecular and Developmental Medicine, University of Siena, Via A. Moro 2, 53100 Siena, Italy
| |
Collapse
|
46
|
Peralta S, Grenier JK, Webb SM, Miller AD, Miranda IC, Parker JSL. Transcriptomic signatures of feline chronic gingivostomatitis are influenced by upregulated IL6. RESEARCH SQUARE 2023:rs.3.rs-2852140. [PMID: 37205490 PMCID: PMC10187381 DOI: 10.21203/rs.3.rs-2852140/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Feline chronic gingivostomatitis (FCGS) is a relatively common and debilitating disease characterized by bilateral inflammation and ulceration of the caudal oral mucosa, alveolar and buccal mucosa, and varying degrees of periodontal disease. The etiopathogenesis of FCGS remains unresolved. In this study, we performed bulk RNA-seq molecular profiling of affected tissues derived from a cohort of client-owned cats with FCGS compared to tissues from unaffected animals, to identify candidate genes and pathways that can help guide future exploration of novel clinical solutions. We complemented transcriptomic findings with immunohistochemistry and in situ hybridization assays to better understand the biological significance of the results and performed RNA-seq validation of selected differentially expressed genes using qPCR assays to demonstrate technical reproducibility. Transcriptomic profiles of oral mucosal tissues in cats with FCGS are enriched with immune- and inflammation-related genes and pathways that appear to be largely influenced by IL6 , and include NFKB, JAK/STAT, IL-17 and IFN type I and II signaling, offering new opportunities to develop novel clinical applications based on a more rational understanding of the disease.
Collapse
Affiliation(s)
| | | | | | | | - Ileana C Miranda
- Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, The Rockefeller University
| | | |
Collapse
|
47
|
Al-Hetty HRAK, Abdulameer SJ, Alkubaisy SA, Zaid SA, Jalil AT, Jasim IK. STAT3 signaling in pancreatic ductal adenocarcinoma: a candidate therapeutic target. Pathol Res Pract 2023; 245:154425. [PMID: 37019018 DOI: 10.1016/j.prp.2023.154425] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 03/19/2023] [Accepted: 03/21/2023] [Indexed: 04/05/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive cancer with a poor prognosis which is lethal in over 90% of cases despite the standard therapies. Mainly activated by Janus kinase 2 (JAK2), signal transducer and activator of transcription 3 (STAT3) is a key transcription factor, capable of exerting the expression of multitude of genes involved in survival. Moreover, STAT3 activity is regulated by the interleukin 28 receptor α (IL28RA) and glutathione s-transferase mu-3 (GSTM3), up-regulation of both contributes to the invasiveness of pancreatic cancer cells. In this regard, STAT3 overactivity has an important pathogenic role in the development of PDAC as it is associated with enhanced cell proliferation, survival, angiogenesis, and metastasis. STAT3-associated expression of vascular endothelial growth factor (VEGF) and matrix metalloproteinase 3 and 9 are implicated in the angiogenic and metastatic behavior of the PDAC. Multitude of evidence underline the protective role of STAT3 inhibition against PDAC both in cell cultures and in tumor grafts. However, specific inhibition of STAT3 was not feasible until recently, when a selective potent chemical STAT3 inhibitor, termed N4, were developed and it turned out to be highly effective against PDAC in vitro, as well as in vivo. This review aims to discuss the most recent advances in our understanding of STAT3 role in the pathogenesis of PDAC and its therapeutic applications.
Collapse
|
48
|
Zou X, Huang Z, Zhan Z, Yuan M, Zhang Y, Liu T, Hu X, Fan W, Chen P, Qin H, Zhang S, Xia Y, Zheng S, Pan Z, Huang P. The alcohol extracts of Sceptridium ternatum (Thunb.) Lyon exert anti-pulmonary fibrosis effect through targeting SETDB1/STAT3/p-STAT3 signaling. JOURNAL OF ETHNOPHARMACOLOGY 2023; 313:116520. [PMID: 37120058 DOI: 10.1016/j.jep.2023.116520] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 04/04/2023] [Accepted: 04/18/2023] [Indexed: 05/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Pulmonary fibrosis (PF) is a pathological process of irreversible scarring of lung tissues, with limited treatment means. Sceptridium ternatum (Thunb.) Lyon (STE) is a traditional Chinese herbal medicine that has a traditional use in relieving cough and asthma, resolving phlegm, clearing heat, and detoxicating in China. However, its role in PF has not been reported. AIM OF THE STUDY This study aims to investigate the protective role of STE in PF and the underlying mechanisms. MATERIALS AND METHODS Sprague-Dawley (SD) rats were divided into control group, PF model group, positive drug (pirfenidone) group and STE group. After 28 days of STE administration in bleomycin (BLM)-induced PF rats, living Nuclear Magnetic Resonance Imaging (NMRI) was used to observe the structural changes of lung tissues. H&E and Masson's trichrome staining were used to observe PF-associated pathological alteration, and immunohistochemistry (IHC) staining, western blotting, and qRT-PCR were used to detect the expression of PF-related marker proteins in the lung tissues. ELISA was used to detect PF-associated biochemical criteria in the lung tissue homogenates. The proteomics technology was used to screen the different proteins. Co-immunoprecipitation, western blotting, and IHC staining were used to confirm the underlying targets of STE as well as its downstream signaling. UPLC-Triple-TOF/MS assay was used to explore the effective components in the alcohol extracts of STE. Autodock vina was used to detect the potential binding between the above effective components and SETDB1. RESULTS STE prevented PF by inhibiting the activation of lung fibroblasts and ECM deposition in BLM-induced PF rats. Mechanism analyses demonstrated that STE could inhibit the up-regulation of SETDB1 induced by BLM and TGF-β1, which further blocked the binding of SETDB1 and STAT3 as well as the phosphorylation of STAT3, ultimately preventing the activation and proliferation of lung fibroblasts. CONCLUSION STE played a preventive role in PF by targeting the SETBD1/STAT3/p-STAT3 pathway, which may be a potential therapeutic agent for PF.
Collapse
Affiliation(s)
- Xiaozhou Zou
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, China; Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Hangzhou, 310014, China.
| | - Zhongjie Huang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310014, China.
| | - Zibo Zhan
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310014, China.
| | - Mengnan Yuan
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, China; Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Hangzhou, 310014, China.
| | - Yiwen Zhang
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, China; Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Hangzhou, 310014, China.
| | - Ting Liu
- Department of Pharmacy, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China.
| | - Xiaoping Hu
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, China; Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Hangzhou, 310014, China.
| | - Weijiao Fan
- Clinical Research Institute, Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital, (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, 310014, China.
| | - Pengcheng Chen
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, China; Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Hangzhou, 310014, China.
| | - Hui Qin
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, China; Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Hangzhou, 310014, China.
| | - Su Zhang
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, China; Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Hangzhou, 310014, China.
| | - Yuxuan Xia
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, China; Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Hangzhou, 310014, China.
| | - Shuilian Zheng
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, China; Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Hangzhou, 310014, China.
| | - Zongfu Pan
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, China; Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Hangzhou, 310014, China.
| | - Ping Huang
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, China; Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Hangzhou, 310014, China.
| |
Collapse
|
49
|
Shao B, Ren SH, Wang ZB, Wang HD, Zhang JY, Qin H, Zhu YL, Sun CL, Xu YN, Li X, Wang H. CD73 mediated host purinergic metabolism in intestine contributes to the therapeutic efficacy of a novel mesenchymal-like endometrial regenerative cells against experimental colitis. Front Immunol 2023; 14:1155090. [PMID: 37180168 PMCID: PMC10167049 DOI: 10.3389/fimmu.2023.1155090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 04/14/2023] [Indexed: 05/15/2023] Open
Abstract
Background The disruption of intestinal barrier functions and the dysregulation of mucosal immune responses, mediated by aberrant purinergic metabolism, are involved in the pathogenesis of inflammatory bowel diseases (IBD). A novel mesenchymal-like endometrial regenerative cells (ERCs) has demonstrated a significant therapeutic effect on colitis. As a phenotypic marker of ERCs, CD73 has been largely neglected for its immunosuppressive function in regulating purinergic metabolism. Here, we have investigated whether CD73 expression on ERCs is a potential molecular exerting its therapeutic effect against colitis. Methods ERCs either unmodified or with CD73 knockout (CD73-/-ERCs), were intraperitoneally administered to dextran sulfate sodium (DSS)-induced colitis mice. Histopathological analysis, colon barrier function, the proportion of T cells, and maturation of dendritic cells (DCs) were investigated. The immunomodulatory effect of CD73-expressing ERCs was evaluated by co-culture with bone marrow-derived DCs under LPS stimulation. FACS determined DCs maturation. The function of DCs was detected by ELISA and CD4+ cell proliferation assays. Furthermore, the role of the STAT3 pathway in CD73-expressing ERCs-induced DC inhibition was also elucidated. Results Compared with untreated and CD73-/-ERCs-treated groups, CD73-expressing ERCs effectively attenuated body weight loss, bloody stool, shortening of colon length, and pathological damage characterized by epithelial hyperplasia, goblet cell depletion, the focal loss of crypts and ulceration, and the infiltration of inflammatory cells. Knockout of CD73 impaired ERCs-mediated colon protection. Surprisingly, CD73-expressing ERCs significantly decreased the populations of Th1 and Th17 cells but increased the proportions of Tregs in mouse mesenteric lymph nodes. Furthermore, CD73-expressing ERCs markedly reduced the levels of pro-inflammatory cytokines (IL-6, IL-1β, TNF-α) and increased anti-inflammatory factors (IL-10) levels in the colon. CD73-expressing ERCs inhibited the antigen presentation and stimulatory function of DCs associated with the STAT-3 pathway, which exerted a potent therapeutic effect against colitis. Conclusions The knockout of CD73 dramatically abrogates the therapeutic ability of ERCs for intestinal barrier dysfunctions and the dysregulation of mucosal immune responses. This study highlights the significance of CD73 mediates purinergic metabolism contributing to the therapeutic effects of human ERCs against colitis in mice.
Collapse
Affiliation(s)
- Bo Shao
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Shao-hua Ren
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Zhao-bo Wang
- School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Hong-da Wang
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Jing-yi Zhang
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Hong Qin
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Yang-lin Zhu
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Cheng-lu Sun
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Yi-ni Xu
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiang Li
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Hao Wang
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
50
|
Niu W, Zhu M, Wang M, Zhang G, Zheng C, Bao Y, Li Y, Zhang N, Wang J, He H, Wang Y. Discovery and development of benzene sulfonamide derivatives as anti-hepatic fibrosis agents. Bioorg Med Chem Lett 2023; 88:129290. [PMID: 37080476 DOI: 10.1016/j.bmcl.2023.129290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 04/14/2023] [Accepted: 04/15/2023] [Indexed: 04/22/2023]
Abstract
A novel benzene sulfonamide compound named IMB16-4 exhibits excellent anti-hepatic fibrosis activity in a recent study. To develop potential anti-hepatic fibrosis agents, a series of benzene sulfonamide derivatives were designed and synthesized based on the scaffold of the lead compound IMB16-4. As it turned out, most of the derivatives displayed potential anti-hepatic fibrosis activity, among which, compounds 11a, 11b, 11d, 13a, 36b, and 47b exhibited inhibition rates of 42.3%, 48.7%, 42.4%, 40.0%, 39.4%, and 49.3%, respectively, which were equivalent to the control IMB16-4 with an inhibition rate of 35.9%, Costunolide with an inhibition rate of 45.4%, and much more potent than that of Epigallocatechin gallate (EGCG) with an inhibition rate of 25.3%. Especially, compounds 46a, 46b, and 46c exhibited excellent anti-hepatic fibrosis activity with inhibition rates of 61.7%, 54.8%, and 60.7%, which were almost 1.5-fold inhibition rates of IMB16-4. In addition, compounds 46a, 46b, and 46c exhibited remarkable inhibitory activity in the gene expression of COL1A1, MMP-2, and the protein expression of COL1A1, FN, α-SMA, and TIMP-1 by inhibiting the JAK1-STAT1/3 pathway. These findings furnished valuable inspiration for the further development of anti-hepatic fibrosis agents.
Collapse
Affiliation(s)
- Weiping Niu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Mei Zhu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Minghua Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Guoning Zhang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Chenghong Zheng
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Yunyang Bao
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Yiming Li
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Na Zhang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Juxian Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Hongwei He
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Yucheng Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China.
| |
Collapse
|