1
|
Wu S, Chen Q, Yang X, Zhang L, Huang X, Huang J, Wu J, Sun C, Zhang W, Wang J. The KSR1/MEK/ERK signaling pathway promotes the progression of intrauterine adhesions. Cell Signal 2025; 131:111730. [PMID: 40089092 DOI: 10.1016/j.cellsig.2025.111730] [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: 11/24/2024] [Revised: 02/24/2025] [Accepted: 03/07/2025] [Indexed: 03/17/2025]
Abstract
Kinase suppressor of Ras 1 (KSR1) serves as a scaffold protein within the RAS-RAF pathway and plays a role in tumorigenesis, immune regulation, cell proliferation, and apoptosis. However, the specific role of KSR1 in the formation and progression of fibrotic diseases, such as intrauterine adhesions (IUA), remains unclear. This study aims to investigate KSR1 expression in IUA and the mechanisms underlying its role in promoting IUA progression. KSR1 was found to be significantly overexpressed in the endometrium of both IUA model rats and patients with IUA. KSR1 is positively involved in the regulation of proliferation, migration, and fibrosis (FN1, Collagen I, α-SMA) in immortalized human endometrial stromal cells (THESCs). Furthermore, KSR1 knockdown was observed to inhibit the fibrosis, proliferation, and migration of transforming growth factor-β1 (TGF-β1)-induced THESCs. Further studies demonstrated that the key proteins of the MEK/ERK signaling pathway, p-MEK1 and p-ERK1/2, were significantly overexpressed in the uterus of IUA rats. In vitro rescue experiments confirmed that the MEK/ERK pathway inhibitor U0126 (An ERK inhibitor) effectively suppressed the enhanced fibrosis, proliferation, and migration induced by KSR1 overexpression. In conclusion, this study demonstrates that KSR1 promotes IUA by enhancing proliferation, migration, and fibrosis of endometrial stromal cells via the MEK/ERK signaling pathway.
Collapse
Affiliation(s)
- Shasha Wu
- Department of Obstetrics and Gynecology, University-Town Hospital of Chongqing Medical University, Chongqing 401331, China
| | - Qiuhong Chen
- Department of Obstetrics and Gynecology, University-Town Hospital of Chongqing Medical University, Chongqing 401331, China
| | - Xiao Yang
- Department of Obstetrics and Gynecology, University-Town Hospital of Chongqing Medical University, Chongqing 401331, China
| | - Lulu Zhang
- Medical Sciences Research Center, University-Town Hospital of Chongqing Medical University, Chongqing 401331, China
| | - Xiyue Huang
- Department of Obstetrics and Gynecology, University-Town Hospital of Chongqing Medical University, Chongqing 401331, China
| | - Jinglin Huang
- Department of Obstetrics and Gynecology, University-Town Hospital of Chongqing Medical University, Chongqing 401331, China
| | - Jiangling Wu
- Medical Sciences Research Center, University-Town Hospital of Chongqing Medical University, Chongqing 401331, China
| | - Congcong Sun
- Department of Obstetrics and Gynecology, University-Town Hospital of Chongqing Medical University, Chongqing 401331, China.
| | - Wenwen Zhang
- Department of Pathology, University-Town Hospital of Chongqing Medical University, Chongqing 401331, China.
| | - Jia Wang
- Department of Obstetrics and Gynecology, University-Town Hospital of Chongqing Medical University, Chongqing 401331, China.
| |
Collapse
|
2
|
Abbad L, Esteve E, Chatziantoniou C. Advances and challenges in kidney fibrosis therapeutics. Nat Rev Nephrol 2025; 21:314-329. [PMID: 39934355 DOI: 10.1038/s41581-025-00934-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/20/2025] [Indexed: 02/13/2025]
Abstract
Chronic kidney disease (CKD) is a major global health burden that affects more than 10% of the adult population. Current treatments, including dialysis and transplantation, are costly and not curative. Kidney fibrosis, defined as an abnormal accumulation of extracellular matrix in the kidney parenchyma, is a common outcome in CKD, regardless of disease aetiology, and is a major cause of loss of kidney function and kidney failure. For this reason, research efforts have focused on identifying mediators of kidney fibrosis to inform the development of effective anti-fibrotic treatments. Given the prominent role of the transforming growth factor-β (TGFβ) family in fibrosis, efforts have focused on inhibiting TGFβ signalling. Despite hopes raised by the efficacy of this approach in preclinical models, translation into clinical practice has not met expectations. Antihypertensive and antidiabetic drugs slow the decline in kidney function and could slow fibrosis but, owing to the lack of technologies for in vivo renal imaging, their anti-fibrotic effect cannot be truly assessed at present. The emergence of new drugs targeting pro-fibrotic signalling, or enabling cell repair and cell metabolic reprogramming, combined with better stratification of people with CKD and the arrival of nanotechnologies for kidney-specific drug delivery, open up new perspectives for the treatment of this major public health challenge.
Collapse
Affiliation(s)
- Lilia Abbad
- INSERM UMR S 1155, Common and Rare Kidney Diseases, Tenon Hospital, Faculty of Medicine, Sorbonne University, Paris, France
| | - Emmanuel Esteve
- INSERM UMR S 1155, Common and Rare Kidney Diseases, Tenon Hospital, Faculty of Medicine, Sorbonne University, Paris, France
| | - Christos Chatziantoniou
- INSERM UMR S 1155, Common and Rare Kidney Diseases, Tenon Hospital, Faculty of Medicine, Sorbonne University, Paris, France.
| |
Collapse
|
3
|
Nair B, Menon A, Rithwik Kalidas M, Nath LR, Calina D, Sharifi-Rad J. Modulating the JAK/STAT pathway with natural products: potential and challenges in cancer therapy. Discov Oncol 2025; 16:595. [PMID: 40268770 DOI: 10.1007/s12672-025-02369-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 04/11/2025] [Indexed: 04/25/2025] Open
Abstract
The Janus Kinase/Signal Transducer and Activator of Transcription (JAK/STAT) pathway is a critical signaling network governing cellular functions such as immune responses, proliferation, and apoptosis. Dysregulation of this pathway is strongly implicated in cancer progression. This review explores the therapeutic potential of natural products, including Curcumin, Resveratrol, Apigenin, and Epigallocatechin Gallate (EGCG), as modulators of the JAK/STAT pathway. These phytochemicals exhibit anticancer activity by inhibiting JAK/STAT phosphorylation, blocking STAT dimerization, and interfering with STAT-DNA binding. A systematic evaluation of included peer-reviewed studies highlights their promise as complementary agents to conventional cancer therapies. However, challenges such as poor bioavailability and the need for robust clinical validation remain significant hurdles. Addressing these limitations through advanced drug delivery systems and rigorous trials could unlock their full potential in cancer treatment.
Collapse
Affiliation(s)
- Bhagyalakshmi Nair
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Ponekkara, P.O., Kochi, Kerala, 682041, India
- Department of Pharmacology, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Ponekkara, P.O., Kochi, Kerala, 682041, India
| | - Anjana Menon
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Ponekkara, P.O., Kochi, Kerala, 682041, India
- Department of Pharmacy Practice, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Ponekkara, P.O., Kochi, Kerala, 682041, India
| | - M Rithwik Kalidas
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Ponekkara, P.O., Kochi, Kerala, 682041, India
- Department of Pharmacy Practice, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Ponekkara, P.O., Kochi, Kerala, 682041, India
| | - Lekshmi R Nath
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Ponekkara, P.O., Kochi, Kerala, 682041, India.
| | - Daniela Calina
- Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, 200349, Craiova, Romania.
| | - Javad Sharifi-Rad
- Universidad Espíritu Santo, Samborondón, 092301, Ecuador.
- Department of Medicine, College of Medicine, Korea University, Seoul, 02841, Republic of Korea.
| |
Collapse
|
4
|
Li X, Li S, Li N. Research Progress on Natural Products Alleviating Liver Inflammation and Fibrosis via NF-κB Pathway. Chem Biodivers 2025; 22:e202402248. [PMID: 39576739 DOI: 10.1002/cbdv.202402248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 11/21/2024] [Accepted: 11/21/2024] [Indexed: 11/24/2024]
Abstract
Liver fibrosis is a key pathological process in chronic liver diseases, regulated by various cytokines and signaling pathways. Among these, the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) signaling pathway plays a significant role in the initiation and progression of liver fibrosis. Recently, natural products have garnered attention as potential anti-fibrotic agents. This review highlights recent studies on how natural products, including flavonoids, terpenoids, polysaccharides, phenols, alkaloids, quinones, phenylpropanoids, steroids, and nitrogen compounds, mitigate liver fibrosis by modulating the NF-κB signaling pathway. Specifically, it examines how these natural products influence NF-κB activation, nuclear translocation, and downstream signaling, thereby inhibiting inflammatory responses, reducing apoptosis, and regulating hepatic stellate cell (HSC) activity, ultimately achieving therapeutic effects against liver fibrosis. A deeper understanding of the mechanisms by which natural products regulate the NF-κB signaling pathway can provide crucial theoretical foundations and valuable insights for the development of novel anti-fibrotic drugs.
Collapse
Affiliation(s)
- Xiaoying Li
- Department of Pathology, Henan Medical College, Zhengzhou, Henan, China
| | - Saifei Li
- Department of Pharmacy, Henan Medical College, Zhengzhou, Henan, China
| | - Ningning Li
- Department of Pathology, Henan Medical College, Zhengzhou, Henan, China
| |
Collapse
|
5
|
Andersen MD, Wolter K, Enemark MH, Lauridsen KL, Hamilton-Dutoit SJ, Starklint J, d'Amore F, Ludvigsen M, Honoré B, Kamper P. Proteomic profiling identifies classic Hodgkin lymphoma patients at risk of bleomycin pulmonary toxicity. Leuk Lymphoma 2025; 66:656-667. [PMID: 39625996 DOI: 10.1080/10428194.2024.2434170] [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: 08/13/2024] [Revised: 10/21/2024] [Accepted: 11/20/2024] [Indexed: 03/29/2025]
Abstract
Advances in treating classic Hodgkin lymphoma (cHL) have improved cure rates, with overall survival exceeding 80%, resulting in a growing population of survivors at risk of long-term complications, particularly cardiac and pulmonary toxicity. Bleomycin, a key component of combination chemotherapy, is associated with bleomycin-induced pulmonary toxicity (BPT). Using label-free quantification nano liquid chromatography-tandem mass spectrometry, protein expression in diagnostic lymphoma samples from patients with and without BPT was compared. Results showed differential protein expression and disrupted cellular pathways, suggesting biological differences in BPT risk. Immunohistochemical analysis revealed higher expression of JAK3, BID, and MMP9, and lower expression of CD20, TPD52, and PIK3R4 in patients with BPT. High BID and low CD20 expression were associated with inferior overall survival, while high BID and low JAK3 and CD20 expression were linked to poorer progression-free survival. These findings highlight altered protein profiles in pretreatment cHL biopsies associated with BPT development.
Collapse
Affiliation(s)
- Maja Dam Andersen
- Department of Hematology, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Katharina Wolter
- Department of Hematology, Aarhus University Hospital, Aarhus, Denmark
| | - Marie Hairing Enemark
- Department of Hematology, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | | | | | - Jørn Starklint
- Department of Medicine, Regional Hospital Goedstrup, Herning, Denmark
| | - Francesco d'Amore
- Department of Hematology, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Maja Ludvigsen
- Department of Hematology, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Bent Honoré
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Peter Kamper
- Department of Hematology, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
6
|
Patil VS, Patil CR, Patel HM, Kumar A. Exploring disulfiram mechanisms in renal fibrosis: insights from biological data and computational approaches. Front Pharmacol 2025; 16:1480732. [PMID: 40170735 PMCID: PMC11958968 DOI: 10.3389/fphar.2025.1480732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 02/03/2025] [Indexed: 04/03/2025] Open
Abstract
Background Disulfiram (DSF) is an anti-alcoholic drug that has been reported to inhibit the epithelial-to-mesenchymal transition and crosslinking during fibrosis, pyroptosis, and inflammatory NF-κB and Nrf-2 signaling pathways. However, there is insufficient evidence to support the mechanisms of DSF in preventing renal fibrosis (RF). Therefore, the current study aimed to elucidate the DSF-modulated targets and pathways in renal fibrosis. Methods The common proteins between DSF and RF were screened for protein-protein interaction, pathway enrichment, cluster, and gene ontology analysis. Molecular docking was executed for core genes using AutoDock Vina through the POAP pipeline. Molecular dynamics (MD) simulation (100 ns) was performed to infer protein-ligand stability, and conformational changes were analyzed by free energy landscape (FEL). Results A total of 78 targets were found to be common between DSF and RF, of which NFKB, PIK3CA/R1, MTOR, PTGS2, and MMP9 were the core genes. PI3K-Akt signaling followed by JAK-STAT, TNF, Ras, ErbB, p53, phospholipase D, mTOR, IL-17, NF-κB, AMPK, VEGF, and MAPK signaling pathways were modulated by DSF in RF. DSF showed a direct binding affinity with active site residues of core genes, and except for DSF with NF-κB, all other complexes, including the standard, were found to be stable during 100 ns MD simulation with minimal protein-ligand root mean squared deviation and residual fluctuations and higher compactness with broad conformational changes. Conclusion DSF protects against renal fibrosis, and this study paves the way for experimental investigation to repurpose DSF for treating RF.
Collapse
Affiliation(s)
- Vishal S. Patil
- Department of Pharmacology, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, India
| | - Chandragouda R. Patil
- Department of Pharmacology, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, India
| | - Harun M. Patel
- Department of Pharmaceutical Chemistry, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, India
| | - Anoop Kumar
- Department of Pharmacology, Delhi Pharmaceutical Sciences and Research University (DPSRU), New Delhi, India
| |
Collapse
|
7
|
Karsdal M, Cox TR, Parker AL, Willumsen N, Sand JMB, Jenkins G, Hansen HH, Oldenburger A, Geillinger-Kaestle KE, Larsen AT, Black D, Genovese F, Eckersley A, Heinz A, Nyström A, Holm Nielsen S, Bennink L, Johannsson L, Bay-Jensen AC, Orange DE, Friedman S, Røpke M, Fiore V, Schuppan D, Rieder F, Simona B, Borthwick L, Skarsfeldt M, Wennbo H, Thakker P, Stoffel R, Clarke GW, Kalluri R, Ruane D, Zannad F, Mortensen JH, Sinkeviciute D, Sundberg F, Coseno M, Thudium C, Croft AP, Khanna D, Cooreman M, Broermann A, Leeming DJ, Mobasheri A, Ricard-Blum S. Advances in Extracellular Matrix-Associated Diagnostics and Therapeutics. J Clin Med 2025; 14:1856. [PMID: 40142664 PMCID: PMC11943371 DOI: 10.3390/jcm14061856] [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: 12/20/2024] [Revised: 01/28/2025] [Accepted: 02/08/2025] [Indexed: 03/28/2025] Open
Abstract
The extracellular matrix (ECM) is the common denominator of more than 50 chronic diseases. Some of these chronic pathologies lead to enhanced tissue formation and deposition, whereas others are associated with increased tissue degradation, and some exhibit a combination of both, leading to severe tissue alterations. To develop effective therapies for diseases affecting the lung, liver, kidney, skin, intestine, musculoskeletal system, heart, and solid tumors, we need to modulate the ECM's composition to restore its organization and function. Across diverse organ diseases, there are common denominators and distinguishing factors in this fibroinflammatory axis, which may be used to foster new insights into drug development across disease indications. The 2nd Extracellular Matrix Pharmacology Congress took place in Copenhagen, Denmark, from 17 to 19 June 2024 and was hosted by the International Society of Extracellular Matrix Pharmacology. The event was attended by 450 participants from 35 countries, among whom were prominent scientists who brought together state-of-the-art research on organ diseases and asked important questions to facilitate drug development. We highlight key aspects of the ECM in the liver, kidney, skin, intestine, musculoskeletal system, lungs, and solid tumors to advance our understanding of the ECM and its central targets in drug development. We also highlight key advances in the tools and technology that enable this drug development, thereby supporting the ECM.
Collapse
Affiliation(s)
- Morten Karsdal
- Nordic Bioscience, 2730 Herlev, Denmark; (N.W.); (J.M.B.S.); (A.T.L.); (F.G.); (S.H.N.); (A.-C.B.-J.); (J.H.M.); (D.S.); (D.J.L.)
| | - Thomas R. Cox
- Garvan Institute of Medical Research, Sydney 2010, Australia; (T.R.C.); (A.L.P.)
- School of Clinical Medicine, St Vincent’s Clinical Campus, UNSW Medicine & Health, UNSW, Sydney 2010, Australia
| | - Amelia L. Parker
- Garvan Institute of Medical Research, Sydney 2010, Australia; (T.R.C.); (A.L.P.)
- School of Clinical Medicine, St Vincent’s Clinical Campus, UNSW Medicine & Health, UNSW, Sydney 2010, Australia
| | - Nicholas Willumsen
- Nordic Bioscience, 2730 Herlev, Denmark; (N.W.); (J.M.B.S.); (A.T.L.); (F.G.); (S.H.N.); (A.-C.B.-J.); (J.H.M.); (D.S.); (D.J.L.)
| | - Jannie Marie Bülow Sand
- Nordic Bioscience, 2730 Herlev, Denmark; (N.W.); (J.M.B.S.); (A.T.L.); (F.G.); (S.H.N.); (A.-C.B.-J.); (J.H.M.); (D.S.); (D.J.L.)
| | - Gisli Jenkins
- Margaret Turner Warwick Centre for Fibrosing Lung Disease, National Heart and Lung Institute, NIHR Imperial Biomedical Research Centre, Imperial College London, London SW7 2AZ, UK;
| | | | | | - Kerstin E. Geillinger-Kaestle
- Department of Immunology and Respiratory Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, 88400 Biberach an der Riss, Germany;
| | - Anna Thorsø Larsen
- Nordic Bioscience, 2730 Herlev, Denmark; (N.W.); (J.M.B.S.); (A.T.L.); (F.G.); (S.H.N.); (A.-C.B.-J.); (J.H.M.); (D.S.); (D.J.L.)
| | | | - Federica Genovese
- Nordic Bioscience, 2730 Herlev, Denmark; (N.W.); (J.M.B.S.); (A.T.L.); (F.G.); (S.H.N.); (A.-C.B.-J.); (J.H.M.); (D.S.); (D.J.L.)
| | - Alexander Eckersley
- Wellcome Centre for Cell Matrix Research, Division of Musculoskeletal and Dermatological Sciences, School of Biological Sciences, University of Manchester, Manchester M13 9PL, UK;
| | - Andrea Heinz
- LEO Foundation Center for Cutaneous Drug Delivery, Department of Pharmacy, University of Copenhagen, 2100 Copenhagen, Denmark;
| | - Alexander Nyström
- Department of Dermatology, Faculty of Medicine, Medical Center—University of Freiburg, 79106 Breisgau, Germany;
| | - Signe Holm Nielsen
- Nordic Bioscience, 2730 Herlev, Denmark; (N.W.); (J.M.B.S.); (A.T.L.); (F.G.); (S.H.N.); (A.-C.B.-J.); (J.H.M.); (D.S.); (D.J.L.)
| | | | | | - Anne-Christine Bay-Jensen
- Nordic Bioscience, 2730 Herlev, Denmark; (N.W.); (J.M.B.S.); (A.T.L.); (F.G.); (S.H.N.); (A.-C.B.-J.); (J.H.M.); (D.S.); (D.J.L.)
| | - Dana E. Orange
- Hospital for Special Surgery, The Rockefeller University, New York, NY 10065, USA;
| | - Scott Friedman
- Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, New York, NY 10029, USA;
| | | | - Vincent Fiore
- Boehringer Ingelheim, 55218 Ingelheim am Rhein, Germany;
| | - Detlef Schuppan
- Institute of Translational Immunology, University Medical Center, Johannes Gutenberg University Mainz, 55131 Mainz, Germany;
| | - Florian Rieder
- Department of Inflammation and Immunity, Cleveland Clinic Foundation, Cleveland, OH 44195, USA;
| | | | - Lee Borthwick
- FibroFind Ltd., FibroFind Laboratories, Medical School, Newcastle upon Tyne NE2 4HH, UK;
- Newcastle Fibrosis Research Group, Biosciences Institute, Newcastle University, Newcastle upon Tyne NE1 7RU, UK
| | - Mark Skarsfeldt
- Nordic Bioscience, 2730 Herlev, Denmark; (N.W.); (J.M.B.S.); (A.T.L.); (F.G.); (S.H.N.); (A.-C.B.-J.); (J.H.M.); (D.S.); (D.J.L.)
| | - Haakan Wennbo
- Takeda, Translational Medicine Biomarkers Gastrointestinal & Global, Boston, MA 02110, USA; (H.W.); (P.T.)
| | - Paresh Thakker
- Takeda, Translational Medicine Biomarkers Gastrointestinal & Global, Boston, MA 02110, USA; (H.W.); (P.T.)
| | - Ruedi Stoffel
- Roche Diagnostics International Ltd., 6343 Rotkreuz, Switzerland;
| | - Graham W. Clarke
- Translational Science and Experimental Medicine, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, 431 83 Gothenburg, Sweden;
- School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King’s College, London E1 9RT, UK
| | - Raghu Kalluri
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Darren Ruane
- Janssen Immunology, Translational Sciences and Medicine, La Jolla, CA 92037, USA;
| | - Faiez Zannad
- Division of Heart Failure and Hypertension, and of the Inserm CIC, University of Lorraine, 54000 Metz, France;
| | - Joachim Høg Mortensen
- Nordic Bioscience, 2730 Herlev, Denmark; (N.W.); (J.M.B.S.); (A.T.L.); (F.G.); (S.H.N.); (A.-C.B.-J.); (J.H.M.); (D.S.); (D.J.L.)
| | - Dovile Sinkeviciute
- Nordic Bioscience, 2730 Herlev, Denmark; (N.W.); (J.M.B.S.); (A.T.L.); (F.G.); (S.H.N.); (A.-C.B.-J.); (J.H.M.); (D.S.); (D.J.L.)
| | - Fred Sundberg
- Sengenics Corporation LLC, Wilmington, DE 19801, USA; (F.S.); (M.C.)
| | - Molly Coseno
- Sengenics Corporation LLC, Wilmington, DE 19801, USA; (F.S.); (M.C.)
| | - Christian Thudium
- Nordic Bioscience, 2730 Herlev, Denmark; (N.W.); (J.M.B.S.); (A.T.L.); (F.G.); (S.H.N.); (A.-C.B.-J.); (J.H.M.); (D.S.); (D.J.L.)
| | - Adam P. Croft
- National Institute for Health and Care Research (NIHR) Birmingham Biomedical Research Centre, University of Birmingham, Birmingham B15 2TT, UK;
- Institute of Inflammation and Ageing, Queen Elizabeth Hospital, University of Birmingham, Birmingham B15 2TT, UK
| | - Dinesh Khanna
- Michigan Medicine, University of Michigan, Ann Arbor, MI 48109, USA;
| | | | - Andre Broermann
- Department of CardioMetabolic Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, 88400 Biberach an der Riss, Germany;
| | - Diana Julie Leeming
- Nordic Bioscience, 2730 Herlev, Denmark; (N.W.); (J.M.B.S.); (A.T.L.); (F.G.); (S.H.N.); (A.-C.B.-J.); (J.H.M.); (D.S.); (D.J.L.)
| | - Ali Mobasheri
- Faculty of Medicine, University of Oulu, 90570 Oulu, Finland;
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, 08406 Vilnius, Lithuania
- Faculté de Médecine, Université de Liège, 4000 Liège, Belgium
- Department of Joint Surgery, First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Sylvie Ricard-Blum
- Institut de Chimie et Biochimie Moléculaires et Supramoléculaires (ICBMS), UMR 5246 CNRS, ICBMS, University Lyon 1, 69622 Villeurbanne Cedex, France;
| |
Collapse
|
8
|
Jiang M, Hou J, Chai Q, Yin S, Liu Q. Mechanism of β-Catenin in Pulmonary Fibrosis Following SARS-CoV-2 Infection. Cells 2025; 14:394. [PMID: 40136643 PMCID: PMC11940791 DOI: 10.3390/cells14060394] [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: 01/11/2025] [Revised: 02/20/2025] [Accepted: 03/05/2025] [Indexed: 03/27/2025] Open
Abstract
Pulmonary fibrosis due to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is the leading cause of death in patients with COVID-19. β-catenin, a key molecule in the Wnt/β-catenin signaling pathway, has been shown to be involved in the development of pulmonary fibrosis (e.g., idiopathic pulmonary fibrosis, silicosis). In this study, we developed a SARS-CoV-2-infected A549-hACE2 cell model to evaluate the efficacy of the A549-hACE2 monoclonal cell line against SARS-CoV-2 infection. The A549-hACE2 cells were then subjected to either knockdown or overexpression of the effector β-catenin, and the modified cells were subsequently infected with SARS-CoV-2. Additionally, we employed transcriptomics and raw letter analysis approaches to investigate other potential effects of β-catenin on SARS-CoV-2 infection. We successfully established a model of cellular fibrosis induced by SARS-CoV-2 infection in lung-derived cells. This model can be utilized to investigate the molecular biological mechanisms and cellular signaling pathways associated with virus-induced lung fibrosis. The results of our mechanistic studies indicate that β-catenin plays a significant role in lung fibrosis resulting from SARS-CoV-2 infection. Furthermore, the inhibition of β-catenin mitigated the accumulation of mesenchymal stroma in A549-hACE2 cells. Additionally, β-catenin knockdown was found to facilitate multi-pathway crosstalk following SARS-CoV-2 infection. The fact that β-catenin overexpression did not exacerbate cellular fibrosis may be attributed to the activation of PPP2R2B.
Collapse
Affiliation(s)
| | | | | | | | - Qian Liu
- Department of Forensic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China; (M.J.); (J.H.); (Q.C.); (S.Y.)
| |
Collapse
|
9
|
Won Y, Kim HH, Jeong SH, Bhosale PB, Abusaliya A, Heo JD, Seong JK, Ahn MJ, Kim HJ, Kim GS. The Effects of Iridin and Irigenin on Cancer: Comparison with Well-Known Isoflavones in Breast, Prostate, and Gastric Cancers. Int J Mol Sci 2025; 26:2390. [PMID: 40141034 PMCID: PMC11942201 DOI: 10.3390/ijms26062390] [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: 02/03/2025] [Revised: 02/28/2025] [Accepted: 02/28/2025] [Indexed: 03/28/2025] Open
Abstract
Cancer, a worldwide problem and one of the leading causes of death due to uncontrolled cell proliferation, can be caused by various factors, such as genetic and environmental factors. Apoptosis is a programmed cell death mechanism that eliminates abnormal cells or renews cells. There are two main apoptotic pathways: intrinsic and extrinsic pathways. These pathways can be affected by various signaling pathways in cancer, such as the PI3K/AKT, MAPK, Wnt, and JAK/STAT pathways. Numerous approaches to cancer treatment have been studied, and among them, natural compounds have been actively researched. Flavonoids are natural compounds from fruits and vegetables and have been studied for their anti-cancer effects. Isoflavones, one of the subclasses of flavonoids, are usually found in soy food or legumes and are effective in several bioactive functions. The well-known isoflavones are genistein, daidzein, and glycitein. Irigenin and iridin can be extracted from the Iris family. Both irigenin and iridin are currently being studied for anti-inflammation, antioxidant, and anti-cancer by inducing apoptosis. In this review, we summarized five isoflavones, genistein, daidzein, glycitein, irigenin, and iridin and their effects on three different cancers: breast cancer, prostate cancer, and gastric cancer.
Collapse
Affiliation(s)
- Yaeram Won
- Department of Pharmacology, Institute of Medical Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea; (Y.W.); (H.-J.K.)
- Department of Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea
| | - Hun-Hwan Kim
- Research Institute of Life Science, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea; (H.-H.K.); (S.-H.J.); (P.B.B.); (A.A.)
| | - Se-Hyo Jeong
- Research Institute of Life Science, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea; (H.-H.K.); (S.-H.J.); (P.B.B.); (A.A.)
| | - Pritam Bhagwan Bhosale
- Research Institute of Life Science, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea; (H.-H.K.); (S.-H.J.); (P.B.B.); (A.A.)
| | - Abuyaseer Abusaliya
- Research Institute of Life Science, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea; (H.-H.K.); (S.-H.J.); (P.B.B.); (A.A.)
| | - Jeong-Doo Heo
- Biological Resources Research Group, Gyeongnam Department of Environment Toxicology and Chemistry, Korea Institute of Toxicology, 17 Jegok-gil, Jinju 52834, Republic of Korea;
| | - Je-Kyung Seong
- Laboratory of Developmental Biology and Goenomics, BK21 PLUS Program for Creative Veterinary Science Research, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea;
| | - Mee-Jung Ahn
- Department of Animal Science, College of Life Science, Sangji University, Wonju 26339, Republic of Korea;
| | - Hye-Jung Kim
- Department of Pharmacology, Institute of Medical Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea; (Y.W.); (H.-J.K.)
- Department of Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea
| | - Gon-Sup Kim
- Research Institute of Life Science, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea; (H.-H.K.); (S.-H.J.); (P.B.B.); (A.A.)
| |
Collapse
|
10
|
Navarro-Ledesma S. Frozen Shoulder as a Metabolic and Immune Disorder: Potential Roles of Leptin Resistance, JAK-STAT Dysregulation, and Fibrosis. J Clin Med 2025; 14:1780. [PMID: 40095902 PMCID: PMC11901274 DOI: 10.3390/jcm14051780] [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: 01/21/2025] [Revised: 03/02/2025] [Accepted: 03/06/2025] [Indexed: 03/19/2025] Open
Abstract
Frozen shoulder (FS) is a complex and multifactorial condition characterized by persistent inflammation, fibrosis, and metabolic dysregulation. Despite extensive research, the underlying drivers of FS remain poorly understood. Recent findings indicate the coexistence of pro-inflammatory and fibrosis-resolving macrophages within affected tissues, suggesting a dysregulated immune response influenced by metabolic and neuroendocrine factors. This review proposes that leptin resistance, a hallmark of metabolic syndrome and chronic inflammation, may play a central role in FS pathogenesis by impairing macrophage polarization, perpetuating inflammation, and disrupting fibrosis resolution. The JAK-STAT signaling pathway, critically modulated by leptin resistance, may further contribute to immune dysregulation by sustaining inflammatory macrophage activation and interfering with tissue remodeling. Additionally, FS shares pathogenic features with fibrotic diseases driven by TGF-β signaling, mitochondrial dysfunction, and circadian disruption, further linking systemic metabolic dysfunction to localized fibrotic pathology. Beyond immune and metabolic regulation, alterations in gut microbiota, bacterial translocation, and chronic psychosocial stress may further exacerbate systemic inflammation and neuroendocrine imbalances, intensifying JAK-STAT dysregulation and leptin resistance. By examining the intricate interplay between metabolism, immune function, and fibrotic remodeling, this review highlights targeting leptin sensitivity, JAK-STAT modulation, and mitochondrial restoration as novel therapeutic strategies for FS treatment. Future research should explore these interconnections to develop integrative interventions that address both the metabolic and immune dysregulation underlying FS, ultimately improving clinical outcomes.
Collapse
Affiliation(s)
- Santiago Navarro-Ledesma
- Department of Physiotherapy, Faculty of Health Sciences, Campus of Melilla, University of Granada, Querol Street 5, 52004 Melilla, Spain
| |
Collapse
|
11
|
Jiang Z, Zhao J, Li R, Ke Q, Wang J, Li Y, Hu S, Zeng J, Pu F, Li N, Xu P, Zhou T. Immune regulation differences in Large yellow croaker with varied resistance to Cryptocaryon irritans infection. FISH & SHELLFISH IMMUNOLOGY 2025; 158:110159. [PMID: 39900312 DOI: 10.1016/j.fsi.2025.110159] [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: 11/06/2024] [Revised: 01/07/2025] [Accepted: 01/24/2025] [Indexed: 02/05/2025]
Abstract
Large yellow croaker (Larimichthys crocea) is one of the main marine aquaculture species in China, but has faced considerable losses due to Cryptocaryon irritans infection. In this study, we successfully established a C. irritans-susceptible population of large yellow croaker by genomic selection technology. We then compared the immune genetic mechanisms of this susceptible population with those of a large yellow croaker population from eastern Fujian in response to C. irritans infection. GWAS identified 44 significant SNPs across 11 QTL regions on different chromosomes associated with C. irritans infection, with most located on chromosomes 1 and 24. Notably, the QTL region on chromosome 1 overlapped with the resistance QTL region mapped in the C. irritans-resistant population previously established by our team, underscoring its crucial role in conferring resistance to C. irritans infection. RNA-Seq analysis revealed significant differences in immune responses between the two groups, with the susceptible group specifically activating the Jak/Stat signaling pathway and upregulating interleukin-related genes, including il11a, il-5r and il-20r. A combined analysis of the GWAS and RNA-Seq data revealed that cspg4 was located in the overlapping QTL region on chromosome 1 associated with resistance. Upon infection, the expression of cspg4 was significantly higher in the susceptible group compared to the control group. As a downstream factor of interleukins, cspg4 may regulate interleukin expression by activating the Jak/Stat pathway, thereby influencing the body's normal immune defense functions. These findings provide new insights into the mechanisms of host-parasite immune responses and highlight potential therapeutic targets.
Collapse
Affiliation(s)
- Zhou Jiang
- State Key Laboratory of Mariculture Breeding, College of Ocean and Earth Sciences, Xiamen University, Xiamen, 361102, China; Fujian Key Laboratory of Genetics and Breeding of Marine Organisms, College of Ocean and Earth Sciences, Xiamen University, Xiamen, 361102, China
| | - Ji Zhao
- State Key Laboratory of Mariculture Breeding, College of Ocean and Earth Sciences, Xiamen University, Xiamen, 361102, China; State Key Laboratory of Marine Environmental Science, College of the Environment and Ecology, Xiamen University, Xiamen, 361102, China; Fujian Key Laboratory of Genetics and Breeding of Marine Organisms, College of Ocean and Earth Sciences, Xiamen University, Xiamen, 361102, China
| | - Rui Li
- State Key Laboratory of Mariculture Breeding, College of Ocean and Earth Sciences, Xiamen University, Xiamen, 361102, China; Fujian Key Laboratory of Genetics and Breeding of Marine Organisms, College of Ocean and Earth Sciences, Xiamen University, Xiamen, 361102, China
| | - Qiaozhen Ke
- State Key Laboratory of Mariculture Breeding, College of Ocean and Earth Sciences, Xiamen University, Xiamen, 361102, China; State Key Laboratory of Marine Environmental Science, College of the Environment and Ecology, Xiamen University, Xiamen, 361102, China; Fujian Key Laboratory of Genetics and Breeding of Marine Organisms, College of Ocean and Earth Sciences, Xiamen University, Xiamen, 361102, China
| | - Jiaying Wang
- State Key Laboratory of Mariculture Breeding, College of Ocean and Earth Sciences, Xiamen University, Xiamen, 361102, China; Fujian Key Laboratory of Genetics and Breeding of Marine Organisms, College of Ocean and Earth Sciences, Xiamen University, Xiamen, 361102, China
| | - Yin Li
- State Key Laboratory of Mariculture Breeding, College of Ocean and Earth Sciences, Xiamen University, Xiamen, 361102, China; Fujian Key Laboratory of Genetics and Breeding of Marine Organisms, College of Ocean and Earth Sciences, Xiamen University, Xiamen, 361102, China
| | - Shuimu Hu
- State Key Laboratory of Mariculture Breeding, College of Ocean and Earth Sciences, Xiamen University, Xiamen, 361102, China; Fujian Key Laboratory of Genetics and Breeding of Marine Organisms, College of Ocean and Earth Sciences, Xiamen University, Xiamen, 361102, China
| | - Junjia Zeng
- State Key Laboratory of Mariculture Breeding, College of Ocean and Earth Sciences, Xiamen University, Xiamen, 361102, China; Fujian Key Laboratory of Genetics and Breeding of Marine Organisms, College of Ocean and Earth Sciences, Xiamen University, Xiamen, 361102, China
| | - Fei Pu
- State Key Laboratory of Mariculture Breeding, College of Ocean and Earth Sciences, Xiamen University, Xiamen, 361102, China; Fujian Key Laboratory of Genetics and Breeding of Marine Organisms, College of Ocean and Earth Sciences, Xiamen University, Xiamen, 361102, China
| | - Ning Li
- State Key Laboratory of Mariculture Breeding, College of Ocean and Earth Sciences, Xiamen University, Xiamen, 361102, China; Fujian Key Laboratory of Genetics and Breeding of Marine Organisms, College of Ocean and Earth Sciences, Xiamen University, Xiamen, 361102, China
| | - Peng Xu
- State Key Laboratory of Mariculture Breeding, College of Ocean and Earth Sciences, Xiamen University, Xiamen, 361102, China; Fujian Key Laboratory of Genetics and Breeding of Marine Organisms, College of Ocean and Earth Sciences, Xiamen University, Xiamen, 361102, China
| | - Tao Zhou
- State Key Laboratory of Mariculture Breeding, College of Ocean and Earth Sciences, Xiamen University, Xiamen, 361102, China; Fujian Key Laboratory of Genetics and Breeding of Marine Organisms, College of Ocean and Earth Sciences, Xiamen University, Xiamen, 361102, China.
| |
Collapse
|
12
|
de Oliveira Silva D, Lima MA, de Freitas TM, Ferreira FCS, de Freitas AKL, Mota MRL, da Silva JA, de Freitas Clementino MA, Havt A. Comparison of the nephroprotective effect of [6]-gingerol and the [6]-gingerol-β-cyclodextrin complex against cisplatin-induced AKI. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-03922-5. [PMID: 40014121 DOI: 10.1007/s00210-025-03922-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 02/11/2025] [Indexed: 02/28/2025]
Abstract
Cisplatin is an antineoplastic agent effective for the treatment of several cancers. However, due to its nephrotoxicity, its use is limited. Acute kidney injury (AKI) induced by cisplatin is caused by inflammation, apoptosis, and oxidative stress in renal tubular cells. Natural compounds derived from ginger (Zingiber officinale) such as [6]-gingerol, possess anti-inflammatory and antioxidant properties. This study explores the protective effects of [6]-gingerol and its β-cyclodextrin complex ([6]-gingerol-β-CD) on cisplatin-induced AKI in mice. We hypothesize that complexing [6]-gingerol with β-cyclodextrin would enhance its solubility and reduce its pungency, making it more suitable for clinical use. Additionally, we aim to determine whether β-cyclodextrin complexation can improve the oral bioavailability of [6]-gingerol without compromising its therapeutic efficacy. The minimum dose of cisplatin required to induce AKI was determined to be 15 mg/kg. Female Swiss mice (n = 6-8) were divided into four groups: control, cisplatin (15 mg/kg), [6]-gingerol (12.5 mg/kg), and [6]-gingerol-β-CD (12.5 mg/kg). The treatments were administered orally for 5 days. On the third day, cisplatin was administered intraperitoneally (i.p.), and the animals were euthanized after 5 days. All biochemical parameters investigated, such as plasma urea and creatinine, urinary gamma GT, urinary protein, MPO, and GSH were altered by cisplatin. The parameters evaluated in this study were categorized into renal function, inflammation, and oxidative stress. Both [6]-gingerol and complexed gingerol effectively restored most of the evaluated parameters to control levels. Following these analyses, we investigated the JAK/STAT pathway and observed that gingerol deactivates this pathway, thereby preventing cisplatin-induced nephrotoxicity. In conclusion, β-CD complexation did not interfere with [6]-gingerol therapeutic, emphasizing its indication for future clinical tests. Further analysis showed that [6]-gingerol prevented cisplatin nephrotoxicity by deactivating JAK/STAT signaling pathway.
Collapse
Affiliation(s)
- Dayara de Oliveira Silva
- Institute of Biomedicine for Brazilian Semiarid, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Mikael Almeida Lima
- Institute of Biomedicine for Brazilian Semiarid, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Thiago Miranda de Freitas
- Institute of Biomedicine for Brazilian Semiarid, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil
| | | | | | - Mário Rogério Lima Mota
- Department of Dental Clinic, Division of Oral Pathology, Faculty of Pharmacy, Dentistry and Nursing, Federal University of Ceará, Fortaleza, CE, Brazil
| | | | | | - Alexandre Havt
- Institute of Biomedicine for Brazilian Semiarid, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil.
| |
Collapse
|
13
|
Vašková J, Kováčová G, Pudelský J, Palenčár D, Mičková H. Methylglyoxal Formation-Metabolic Routes and Consequences. Antioxidants (Basel) 2025; 14:212. [PMID: 40002398 PMCID: PMC11852113 DOI: 10.3390/antiox14020212] [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: 12/18/2024] [Revised: 02/04/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
Methylglyoxal (MGO), a by-product of glycolysis, plays a significant role in cellular metabolism, particularly under stress conditions. However, MGO is a potent glycotoxin, and its accumulation has been linked to the development of several pathological conditions due to oxidative stress, including diabetes mellitus and neurodegenerative diseases. This paper focuses on the biochemical mechanisms by which MGO contributes to oxidative stress, particularly through the formation of advanced glycation end products (AGEs), its interactions with antioxidant systems, and its involvement in chronic diseases like diabetes, neurodegeneration, and cardiovascular disorders. MGO exerts its effects through multiple signaling pathways, including NF-κB, MAPK, and Nrf2, which induce oxidative stress. Additionally, MGO triggers apoptosis primarily via intrinsic and extrinsic pathways, while endoplasmic reticulum (ER) stress is mediated through PERK-eIF2α and IRE1-JNK signaling. Moreover, the activation of inflammatory pathways, particularly through RAGE and NF-κB, plays a crucial role in the pathogenesis of these conditions. This study points out the connection between oxidative and carbonyl stress due to increased MGO formation, and it should be an incentive to search for a marker that could have prognostic significance or could be a targeted therapeutic intervention in various diseases.
Collapse
Affiliation(s)
- Janka Vašková
- Department of Medical Biology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 11 Košice, Slovakia
| | - Gabriela Kováčová
- Department of Medical and Clinical Biochemistry, Faculty of Medicine, Pavol Jozef Šafárik, 040 11 Košice, Slovakia; (G.K.)
| | - Jakub Pudelský
- Department of Medical and Clinical Biochemistry, Faculty of Medicine, Pavol Jozef Šafárik, 040 11 Košice, Slovakia; (G.K.)
| | - Drahomír Palenčár
- Department of Plastic Surgery, Faculty of Medicine, Comenius University Bratislava, 813 72 Bratislava, Slovakia
| | - Helena Mičková
- Department of Medical Biology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 11 Košice, Slovakia
| |
Collapse
|
14
|
Hassan HM, El Safadi M, Hayat MF, Al-Emam A. Prevention of fenitrothion induced hepatic toxicity by saponarin via modulating TLR4/MYD88, JAK1/STAT3 and NF-κB signaling pathways. Int J Biochem Cell Biol 2025; 179:106716. [PMID: 39645143 DOI: 10.1016/j.biocel.2024.106716] [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: 10/06/2024] [Revised: 11/27/2024] [Accepted: 12/03/2024] [Indexed: 12/09/2024]
Abstract
Fenitrothion (FEN) is an organophosphate insecticidal agent that is considered as major source of organs toxicity. Saponarin (SAP) is a naturally occurring novel flavone that exhibits a wide range of medicinal properties. The current trial was conducted to evaluate the ameliorative potential of SAP against FEN instigated liver toxicity in rats. Thirty-two male albino rats were apportioned into four groups including control, FEN (10 mg/kg), FEN (10 mg/kg) + SAP (80 mg/kg), and SAP (80 mg/kg) alone treated group. It was revealed that FEN administration upregulated the gene expression of TNF-α, TLR4, IL-1β, MYD88, IL-6, TRAF6, COX-2, NF-κB, JAK1 and STAT3 while reducing the gene expression of IκB. Moreover, the levels of reactive oxygen species (ROS) and malondialdehyde (MDA) were increased while the activities of catalase (CAT), glutathione peroxidase (GPx), superoxide dismutase (SOD), heme-oxygenase-1 (HO-1) and glutathione reductase (GSR) were decreased after FEN exposure. Furthermore, FEN administration notably escalated the levels of hepatic enzymes including alanine transaminase (ALT), aspartate aminotransferase (AST), gamma-glutamyl transferase (GGT) and alkaline phosphatase (ALP) whereas reduced the levels of total proteins and albumin. Besides, FEN intake upregulated the levels of Caspase-9, Bax and Caspase-3 while reducing the levels of Bcl-2. Hepatic histology was impaired after FEN intoxication. Nonetheless, SAP treatment remarkably protected the normal state of liver via regulating abovementioned irregularities. Our in-silico analysis confirmed that SAP hold that potential to interact with binding pocket of these proteins, highlighting its ability as a therapeutic compound to alleviate FEN-induced liver damage.
Collapse
Affiliation(s)
- Hesham M Hassan
- Department of pathology, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Mahmoud El Safadi
- Department of Chemistry, College of Science, United Arab Emirates University, P.O. Box 15551, Al Ain, Abu Dhabi , United Arab Emirates
| | - Muhammad Faisal Hayat
- Department of Zoology, Wildlife and Fisheries, University of Agriculture, Faisalabad, Pakistan.
| | - Ahmed Al-Emam
- Department of Pathology, College of Medicine, King Khalid University, Asir 61421, Saudi Arabia; Department of Forensic Medicine and Clinical Toxicology, Mansoura University, Egypt
| |
Collapse
|
15
|
Feng R, Liu H, Chen Y. Baricitinib represses the myocardial fibrosis via blocking JAK/STAT and TGF-β1 pathways in vivo and in vitro. BMC Cardiovasc Disord 2025; 25:65. [PMID: 39891042 PMCID: PMC11783835 DOI: 10.1186/s12872-025-04517-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: 03/04/2024] [Accepted: 01/22/2025] [Indexed: 02/03/2025] Open
Abstract
BACKGROUND JAK/STAT pathway is closely involved in the organ fibrotic process. The current study aimed to investigate the impact of baricitinib, an oral selective JAK1/JAK2 inhibitor, on the myocardial fibrosis in vivo and the activation of cardiac fibroblasts in vitro. METHODS The mouse myocardial fibrosis model was established by isoproterenol (ISO) treatment, then was treated by baricitinib. The activation of mouse cardiac fibroblasts was established by TGF-β1 stimulation, then was treated by baricitinib with several concentrations. Besides, JAK2 was knocked down by small interfering RNA (siRNA) in TGF-β1-stimulated mouse cardiac fibroblasts. RESULTS Baricitinib not only attenuated myocardial cell widening, inflammatory infiltration, fibrous tissue, and heart index, but also reduced collagen volume fraction, the expressions of Col1, Col3, α-SMA, Fn, MMP9, and TIMP1 in ISO-induced myocardial fibrosis mice. Meanwhile, baricitinib decreased the expressions of p-STAT3 and TGF-βRII in these mice. Interestingly, in TGF-β1-stimulated cardiac fibroblasts, baricitinib decreased the expressions of Col1, Col3, α-SMA, Fn, MMP9, and TIMP1 in a dose-dependent manner (From 10 to 2000 nM), also exhibited a dose-dependent impact on the expressions of p-STAT3 and TGF-βRII. Finally, JAK2 knockdown by siRNA downregulated the expressions of Col1, Col3, α-SMA, and Fn in TGF-β1-stimulated cardiac fibroblasts. CONCLUSION Inhibition of JAK/STAT pathway by baricitinib represses the myocardial fibrosis in vivo and in vitro, indicating baricitinib may be a treatment option for myocardial fibrosis, while further validation is needed.
Collapse
Affiliation(s)
- Renlei Feng
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
- Department of Geriatrics, Chongqing General Hospital, Chongqing University, Chongqing, 401147, China
| | - Hongli Liu
- Department of Geriatrics, Chongqing Medical University, Chongqing, 400016, China
| | - Yunqing Chen
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China.
| |
Collapse
|
16
|
Gillesberg FS, Pehrsson M, Bay-Jensen AC, Frederiksen P, Karsdal M, Deleuran BW, Kragstrup TW, Kubo S, Tanaka Y, Mortensen JH. Regulation of fibronectin and collagens type I, III and VI by TNF-α, TGF-β, IL-13, and tofacitinib. Sci Rep 2025; 15:1087. [PMID: 39774197 PMCID: PMC11707072 DOI: 10.1038/s41598-024-84151-3] [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: 08/15/2024] [Accepted: 12/20/2024] [Indexed: 01/11/2025] Open
Abstract
Understanding how inflammatory cytokines influence profibrogenic wound healing responses in fibroblasts is important for understanding the pathogenesis of fibrosis. TNF-α and IL-13 are key cytokines in Th1 and Th2 immune responses, respectively, while TGF-β1 is the principal pro-fibrotic mediator. We show that 12-day fibroblast culture with TNF-α or IL-13 induces fibrogenesis, marked by progressively increasing type III and VI collagen formation, and that TGF-β1 co-stimulation amplifies these effects. Tofacitinib substantially reduced the formation of ECM proteins in response to IL-13, while fibrogenesis in response to TNF-α or TGF-β1 was marginally inhibited. The in vitro findings were supported by clinical observations in patients with active rheumatoid arthritis, which had elevated serum type III collagen formation, indicating ongoing fibrogenesis during inflammation. After 48-60 weeks of tofacitinib treatment, type III collagen degradation, aswell as formation, were significantly decreased compared to baseline, highlighting dual anti-inflammatory and anti-fibrogenic effects of tofacitinib. In contrast, other anti-inflammatory treatments including methotrexate, adalimumab and tocilizumab demonstrated anti-inflammatory effects only. Our results highlight fibro-inflammatory profiles associated with TNF-α or IL-13 stimulation, both alone and in combination with TGF-β1, and support the use of tofacitinib as an anti-fibrogenic treatment in chronic inflammatory conditions.
Collapse
Affiliation(s)
- Frederik S Gillesberg
- Nordic Bioscience, Immunoscience, Herlev Hovedgade 205-207, Herlev, 2730, Denmark.
- Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, Aarhus C, 8000, Denmark.
| | - Martin Pehrsson
- Nordic Bioscience, Immunoscience, Herlev Hovedgade 205-207, Herlev, 2730, Denmark
| | | | - Peder Frederiksen
- Nordic Bioscience, Immunoscience, Herlev Hovedgade 205-207, Herlev, 2730, Denmark
| | - Morten Karsdal
- Nordic Bioscience, Immunoscience, Herlev Hovedgade 205-207, Herlev, 2730, Denmark
| | - Bent W Deleuran
- Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, Aarhus C, 8000, Denmark
- Department of Rheumatology, Århus University Hospital, Palle Juul-Jensens Boulevard 99, Aarhus N, 8200, Denmark
| | - Tue W Kragstrup
- Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, Aarhus C, 8000, Denmark
- Department of Rheumatology, Århus University Hospital, Palle Juul-Jensens Boulevard 99, Aarhus N, 8200, Denmark
- Diagnostic Center, Regional Hospital Silkeborg, Falkevej 1, Silkeborg, 8600, Denmark
| | - Satoshi Kubo
- The First Department of Internal Medicine, School of Medicine, University of Occupational & Environmental Health, Japan, 1-1 Iseigaoka, Yahata-nishi, 807-8555, Kitakyushu, Japan
| | - Yoshiya Tanaka
- The First Department of Internal Medicine, School of Medicine, University of Occupational & Environmental Health, Japan, 1-1 Iseigaoka, Yahata-nishi, 807-8555, Kitakyushu, Japan
| | - Joachim H Mortensen
- Nordic Bioscience, Immunoscience, Herlev Hovedgade 205-207, Herlev, 2730, Denmark
| |
Collapse
|
17
|
Özmen Ö, Şahi̇n M, Topsakal Ş, Taşan Ş, Şahi̇n U. Lycopene ameliorates diabetes-induced pancreatic, hepatic, and renal damage by modulating the JAK/STAT/SOCS signaling pathway in rats. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2025; 28:461-468. [PMID: 39968090 PMCID: PMC11831743 DOI: 10.22038/ijbms.2025.79979.17326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 08/10/2024] [Indexed: 02/20/2025]
Abstract
Objectives Emerging evidence suggests that the JAK/STAT/SOCS signaling pathway is crucial for maintaining homeostasis, and its dysregulation contributes to diabetes development. This study aimed to evaluate the roles of SOCS-1 and SOCS-3 in the pancreas, liver, and kidney and to explore the involvement of the JAK/STAT pathway in the molecular mechanisms underlying their effects on inflammation and apoptosis, as well as organ injury in a diabetes mellitus (DM) model. Additionally, we sought to elucidate the role of the JAK/STAT/SOCS pathway in mediating the effects of lycopene (LYC). Materials and Methods Forty Sprague-Dawley rats were divided into control, DM, LYC, and LYC+DM groups. Diabetes was induced in the DM groups using streptozotocin. LYC was administered to the LYC and LYC+DM groups for 30 days. After the study, pancreas, liver, and kidney tissues were analyzed using histopathological, immunohistochemical, and PCR methods. Results Significant vacuolization and degenerative changes were observed in the DM group's pancreatic islet cells. Kidney and liver tissues showed hyperemia, hemorrhage, and degenerative changes. Immunohistochemical analysis revealed increased expression of Cas-3, TNF-α, IFN-α, and IL-6, while IL-10 was significantly reduced in the DM group. PCR analysis showed elevated levels of TNF-α and Cas-3, with decreased SOCS-1 and SOCS-3 expression in the DM group. Conclusion This study highlights the therapeutic potential of targeting the JAK/STAT/SOCS pathway with lycopene, demonstrating its promise in mitigating diabetes and related complications.
Collapse
Affiliation(s)
- Özlem Özmen
- Department of Pathology, Faculty of Veterinary Medicine, Burdur Mehmet Akif Ersoy University, Burdur, Türkiye
| | - Melda Şahi̇n
- Department of Pathology, Faculty of Veterinary Medicine, Burdur Mehmet Akif Ersoy University, Burdur, Türkiye
| | - Şenay Topsakal
- Department of Endocrinology and Metabolism,University, Pamukkale Faculty of Medicine, Denizli, Türkiye
| | - Şerife Taşan
- Department of Pathology, Faculty of Veterinary Medicine, Burdur Mehmet Akif Ersoy University, Burdur, Türkiye
| | - Uğur Şahi̇n
- Genetic Research Unit, Innovative Technologies Application and Research Center, Süleyman Demirel University, Isparta, Türkiye
| |
Collapse
|
18
|
Di X, Li Y, Wei J, Li T, Liao B. Targeting Fibrosis: From Molecular Mechanisms to Advanced Therapies. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410416. [PMID: 39665319 PMCID: PMC11744640 DOI: 10.1002/advs.202410416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 10/27/2024] [Indexed: 12/13/2024]
Abstract
As the final stage of disease-related tissue injury and repair, fibrosis is characterized by excessive accumulation of the extracellular matrix. Unrestricted accumulation of stromal cells and matrix during fibrosis impairs the structure and function of organs, ultimately leading to organ failure. The major etiology of fibrosis is an injury caused by genetic heterogeneity, trauma, virus infection, alcohol, mechanical stimuli, and drug. Persistent abnormal activation of "quiescent" fibroblasts that interact with or do not interact with the immune system via complicated signaling cascades, in which parenchymal cells are also triggered, is identified as the main mechanism involved in the initiation and progression of fibrosis. Although the mechanisms of fibrosis are still largely unknown, multiple therapeutic strategies targeting identified molecular mechanisms have greatly attenuated fibrotic lesions in clinical trials. In this review, the organ-specific molecular mechanisms of fibrosis is systematically summarized, including cardiac fibrosis, hepatic fibrosis, renal fibrosis, and pulmonary fibrosis. Some important signaling pathways associated with fibrosis are also introduced. Finally, the current antifibrotic strategies based on therapeutic targets and clinical trials are discussed. A comprehensive interpretation of the current mechanisms and therapeutic strategies targeting fibrosis will provide the fundamental theoretical basis not only for fibrosis but also for the development of antifibrotic therapies.
Collapse
Affiliation(s)
- Xingpeng Di
- Department of Urology and Institute of UrologyWest China HospitalSichuan UniversityChengduP.R. China
| | - Ya Li
- Department of Urology and Institute of UrologyWest China HospitalSichuan UniversityChengduP.R. China
| | - Jingwen Wei
- Department of Urology and Institute of UrologyWest China HospitalSichuan UniversityChengduP.R. China
| | - Tianyue Li
- Department of Urology and Institute of UrologyWest China HospitalSichuan UniversityChengduP.R. China
| | - Banghua Liao
- Department of Urology and Institute of UrologyWest China HospitalSichuan UniversityChengduP.R. China
| |
Collapse
|
19
|
Saraswat I, Goel A. Herbal Remedies for Hepatic Inflammation: Unravelling Pathways and Mechanisms for Therapeutic Intervention. Curr Pharm Des 2025; 31:128-139. [PMID: 39350422 DOI: 10.2174/0113816128348771240925100639] [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: 08/03/2024] [Accepted: 09/16/2024] [Indexed: 02/18/2025]
Abstract
Inflammation is a universal response of mammalian tissue to harm, comprising reactions to injuries, pathogens, and foreign particles. Liver inflammation is commonly associated with hepatocyte necrosis and apoptosis. These forms of liver cell injury initiate a sequence of events independent of the etiological basis for the inflammation and can result in hepatic disorders. It is also common for liver cancer. This review fundamentally focuses on the molecular pathways involved in hepatic inflammation. This review aims to explore the molecular pathways involved in hepatic inflammation, focusing on arachidonic acid, NF-κB, MAPK, PI3K/Akt, and JAK/STAT pathways. It investigates active compounds in herbal plants and their pharmacological characteristics. The review proposes a unique therapeutic blueprint for managing hepatic inflammation and diseases by modifying these pathways with herbal remedies.
Collapse
Affiliation(s)
- Istuti Saraswat
- Department of Biotechnology, GLA University, 17 km Stone, NH-2 Mathura-Delhi Road Mathura, Chaumuhan, Uttar Pradesh, India
| | - Anjana Goel
- Department of Biotechnology, GLA University, 17 km Stone, NH-2 Mathura-Delhi Road Mathura, Chaumuhan, Uttar Pradesh, India
| |
Collapse
|
20
|
Neblett MF, Ducharme MT, Meridew JA, Haak AJ, Girard S, Tschumperlin DJ, Stewart EA. Evaluation of the In Vivo Efficacy of the JAK Inhibitor AZD1480 in Uterine Leiomyomas Using a Patient-derived Xenograft Murine Model. Reprod Sci 2024:10.1007/s43032-024-01775-6. [PMID: 39738934 DOI: 10.1007/s43032-024-01775-6] [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: 09/10/2024] [Accepted: 12/17/2024] [Indexed: 01/02/2025]
Abstract
Uterine leiomyomas are common noncancerous hormonally-dependent neoplasms comprised of uterine smooth-muscle cells and fibroblasts. Despite their significant impact on morbidity, effective non-hormonal medical treatments are lacking. In vitro studies have identified the Janus kinase/signal transducer and activator of transcription (JAK/STAT) signaling pathway as a promising target in leiomyoma cells. Our objective was to evaluate the efficacy of AZD1480, a JAK 1/2 inhibitor, in treating uterine leiomyomas using a patient-derived xenograft murine model. Ovariectomized immunodeficient mice received an estrogen and progesterone pellet and were subsequently implanted with human leiomyoma tissue surgically resected from premenopausal women not on hormonal medication. Mice were divided into treatment (n = 6) and vehicle control (n = 6) groups receiving either 50 mg/kg of AZD1480 or vehicle via oral gavage for 5 days/week for 28 days. Our results demonstrate a significant AZD1480-mediated reduction in both xenograft volume (59.5% vs. 0.3%; treated vs. control, p < .0001) and weight (56.0% vs. 31.2%; p = 0.03) compared to controls. Moreover, xenografts from the treated group exhibited a significant decrease in cell density(p = 0.01). Levels of pSTAT3-positive cells (4.1% vs. 10.3%), Ki67-positive cells (4.1% vs. 6.5%), and fibrillar collagen (19.8% vs. 29.5%) declined but did not reach statistical significance, whereas AZD1480 treatment significantly reduced blood vessel formation in the xenografts (20.1 vs 45.6 per FOV; p = 0.01). These findings suggest JAK inhibition as a potential treatment for uterine leiomyomas by targeting angiogenesis. However, further studies are warranted to explore alternative JAK inhibitors, examine downstream effects, optimize dosing, and establish clinical efficacy and safety.
Collapse
Affiliation(s)
- Michael F Neblett
- Department of Obstetrics and Gynecology, Mayo Clinic, Rochester, MN, USA
- Mayo Clinic Alix School of Medicine, Rochester, MN, USA
| | | | - Jeffrey A Meridew
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Andrew J Haak
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Sylvie Girard
- Department of Obstetrics and Gynecology, Mayo Clinic, Rochester, MN, USA
- Mayo Clinic Alix School of Medicine, Rochester, MN, USA
- Department of Immunology, Mayo Clinic, Rochester, MN, USA
| | - Daniel J Tschumperlin
- Mayo Clinic Alix School of Medicine, Rochester, MN, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Elizabeth A Stewart
- Department of Obstetrics and Gynecology, Mayo Clinic, Rochester, MN, USA.
- Mayo Clinic Alix School of Medicine, Rochester, MN, USA.
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
21
|
Fang X, Zhang W, Liu C, Liu Y, Tan W, Wang Z, Wang X. Inhibition of peritendinous adhesion through targeting JAK2-STAT3 signaling pathway: The therapeutic potential of AG490. Int Immunopharmacol 2024; 143:113582. [PMID: 39527887 DOI: 10.1016/j.intimp.2024.113582] [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: 09/11/2024] [Revised: 11/04/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
Peritendinous adhesion is a common complication following tendon injury repair, posing a significant clinical challenge that requires urgent attention. The primary cause of peritendinous adhesion is the excessive deposition of collagen matrix due to the abnormal proliferation of fibroblasts in an inflammatory state. Janus kinase2 (JAK2) and signal transducer and activator of transcription 3 (STAT3) are key signaling molecules involved in cell proliferation and fibrosis development in various organs. However, the role of the JAK-2 and STAT3 signaling pathways in peritendinous adhesion fibrosis remains unclear. In our study, we first observed upregulation of p-JAK2 and p-STAT3 proteins in human peritendinous adhesion specimens and rat peritendinous adhesion models. In vitro, the JAK2/STAT3 pathway inhibitor AG490 effectively inhibited TGF-β1-induced fibroblast proliferation. Wound healing and transwell assays demonstrated that AG490 suppressed TGF-β1-induced fibroblast migration. Furthermore, we found that AG490 decreased the expression of pro-inflammatory factors, including IL-1β and TNF-α, as well as extracellular matrix (ECM) proteins in fibroblasts under TGF-β1 stimulation. In vivo, histological staining showed that AG490 prevented fibrous tissue formation in a rat model of tendon injury. Moreover, AG490 inhibited the overexpression of pro-inflammatory factors IL-1β and TNF-α, as well as ECM in the peritendinous adhesions. In conclusion, AG490 inhibited fibrosis and inflammation in injured tendons by targeting the JAK2-STAT3 signaling pathway, presenting a promising strategy for the prophylaxis of peritendinous adhesions.
Collapse
Affiliation(s)
- Xue Fang
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China; Hubei Clinical Medical Research Center of Trauma and Microsurgery, Wuhan, Hubei, China
| | - Wang Zhang
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China; Hubei Clinical Medical Research Center of Trauma and Microsurgery, Wuhan, Hubei, China
| | - Changhuan Liu
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China; Hubei Clinical Medical Research Center of Trauma and Microsurgery, Wuhan, Hubei, China
| | - Yuping Liu
- Sichuan University West China Second University Hospital, Department of Anesthesiology, Chengdu, Sichuan, China
| | - Wei Tan
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China; Hubei Clinical Medical Research Center of Trauma and Microsurgery, Wuhan, Hubei, China
| | - Zheng Wang
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China; Hubei Clinical Medical Research Center of Trauma and Microsurgery, Wuhan, Hubei, China.
| | - Xin Wang
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China; Hubei Clinical Medical Research Center of Trauma and Microsurgery, Wuhan, Hubei, China; Elderly Hip Fracture Diagnosis and Treatment Center, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China.
| |
Collapse
|
22
|
Meng L, Gu T, Yu P, Zhang Z, Wei Z. The role of microglia in Neuroinflammation associated with cardiopulmonary bypass. Front Cell Neurosci 2024; 18:1496520. [PMID: 39742156 PMCID: PMC11685197 DOI: 10.3389/fncel.2024.1496520] [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: 09/17/2024] [Accepted: 11/28/2024] [Indexed: 01/03/2025] Open
Abstract
Cardiopulmonary bypass (CPB) and deep hypothermic circulatory arrest (DHCA) are indispensable core techniques in cardiac surgery. Numerous studies have shown that cardiopulmonary bypass and deep hypothermic circulatory arrest are associated with the occurrence of neuroinflammation, accompanied by the activation of microglia. Microglia, as macrophages in the central nervous system, play an irreplaceable role in neuroinflammation. Current research on neuroinflammation induced by microglia activation mainly focuses on neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, neuropathic pain, acquired brain injury, and others. However, there is relatively limited research on microglia and neuroinflammation under conditions of cardiopulmonary bypass and deep hypothermic circulatory arrest. The close relationship between cardiopulmonary bypass, deep hypothermic circulatory arrest, and cardiac surgery underscores the importance of identifying targets for intervening in neuroinflammation through microglia. This could greatly benefit cardiac surgery patients during cardiopulmonary bypass and the perioperative period, significantly improving patient prognosis. This review article provides the first comprehensive discussion on the signaling pathways associated with neuroinflammation triggered by microglia activation, the impact of cardiopulmonary bypass on microglia, as well as the current status and advancements in cardiopulmonary bypass animal models. It provides new insights and methods for the treatment of neuroinflammation related to cardiopulmonary bypass and deep hypothermic circulatory arrest, holding significant importance for clinical treatment by cardiac surgeons, management strategies by cardiopulmonary bypass physicians, and the development of neurologically related medications.
Collapse
Affiliation(s)
- Lingda Meng
- Department of Cardiac Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Tianxiang Gu
- Department of Cardiac Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Peng Yu
- Department of Cardiac Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Zhiwei Zhang
- Department of Cardiac Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Zhijing Wei
- Department of Trauma Center, The First Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
23
|
Ronan G, Yang J, Zorlutuna P. Small Extracellular Vesicles Isolated from Cardiac Tissue Matrix or Plasma Display Distinct Aging-Related Changes in Cargo Contributing to Chronic Cardiovascular Disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.06.627231. [PMID: 39713371 PMCID: PMC11661072 DOI: 10.1101/2024.12.06.627231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Aging is a major risk factor for cardiovascular disease, the leading cause of death worldwide, and numerous other diseases, but the mechanisms of these aging-related effects remain elusive. Chronic changes in the microenvironment and paracrine signaling behaviors have been implicated, but remain understudied. Here, for the first time, we directly compare extracellular vesicles obtained from young and aged patients to identify therapeutic or disease-associated agents, and directly compare vesicles isolated from heart tissue matrix (TEVs) or plasma (PEVs). While young EVs showed notable overlap of miRNA cargo, aged EVs differed substantially, indicating differential age-related changes between TEVs and PEVs. TEVs overall were uniquely enriched in miRNAs which directly or indirectly demonstrate cardioprotective effects, with 45 potential therapeutic agents implicated in our analysis. Both populations also showed increased predisposition to disease with aging, though through different mechanisms. PEVs were largely correlated with chronic systemic inflammation, while TEVs were more related to cardiac homeostasis and local inflammation. From this, 17 protein targets unique to TEVs were implicated as aging-related changes which likely contribute to the development of cardiovascular disease.
Collapse
Affiliation(s)
- George Ronan
- Bioengineering Graduate Program, University of Notre Dame, Notre Dame, IN, 46556, USA
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Jun Yang
- Bioengineering Graduate Program, University of Notre Dame, Notre Dame, IN, 46556, USA
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Pinar Zorlutuna
- Bioengineering Graduate Program, University of Notre Dame, Notre Dame, IN, 46556, USA
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN, 46556, USA
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN, 46556, USA
- Harper Cancer Research Institute, University of Notre Dame, Notre Dame, 46556, USA
| |
Collapse
|
24
|
Singh K, Witek M, Brahmbhatt J, McEntire J, Thirunavukkarasu K, Oladipupo SS. Stage-Dependent Fibrotic Gene Profiling of WISP1-Mediated Fibrogenesis in Human Fibroblasts. Cells 2024; 13:2005. [PMID: 39682753 PMCID: PMC11640464 DOI: 10.3390/cells13232005] [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: 11/03/2024] [Revised: 11/28/2024] [Accepted: 11/29/2024] [Indexed: 12/18/2024] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is the most common interstitial lung disease with unknown etiology, characterized by chronic inflammation and tissue scarring. Although, Pirfenidone and Nintedanib slow the disease progression, no currently available drugs or therapeutic interventions address the underlying cause, highlighting the unmet medical need. A matricellular protein, Wnt-1-induced secreted protein 1 (WISP1), also referred to as CCN4 (cellular communication network factor 4), is a secreted multi-modular protein implicated in multi-organ fibrosis. Although the precise mechanism of WISP1-mediated fibrosis remains unclear, emerging evidence indicates that WISP1 is profibrotic in nature. While WISP1-targeting therapy is applied in the clinic for fibrosis, detailed interrogation of WISP1-mediated fibrogenic molecular and biological pathways is lacking. Here, for the first time, using NanoString® technology, we identified a novel WISP1-associated profibrotic gene signature and molecular pathways potentially involved in the initiation and progression of fibrosis in primary human dermal and lung fibroblasts from both healthy individuals and IPF patients. Our data demonstrate that WISP1 is upregulated in IPF-lung fibroblasts as compared to healthy control. Furthermore, our results confirm that WISP1 is downstream of the transforming growth factor-β (TGFβ), and it induces fibroblast cell proliferation. Additionally, WISP1 induced IL6 and CCL2 in fibroblasts. We also developed a novel, combined TGFβ and WISP1 in vitro system to demonstrate a role for WISP1 in the progression of fibrosis. Overall, our findings uncover not only similarities but also striking differences in the molecular profile of WISP1 in human fibroblasts, both during the initiation and progression phases, as well as in disease-specific context.
Collapse
Affiliation(s)
- Kirti Singh
- Biotherapeutics Enabling Biology, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46225, USA; (K.S.); (J.M.)
| | - Marta Witek
- Protein Optimization, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46225, USA; (M.W.); (J.B.)
| | - Jaladhi Brahmbhatt
- Protein Optimization, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46225, USA; (M.W.); (J.B.)
| | - Jacquelyn McEntire
- Biotherapeutics Enabling Biology, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46225, USA; (K.S.); (J.M.)
| | - Kannan Thirunavukkarasu
- Immunology Research, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46225, USA;
| | - Sunday S. Oladipupo
- Biotherapeutics Enabling Biology, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46225, USA; (K.S.); (J.M.)
| |
Collapse
|
25
|
Zhang Q, McDermott GC, Juge PA, Chang SH, Vanni KM, Qian G, Bade KJ, Mueller KT, Kowalski EN, Saavedra AA, Sparks JA. Disease-modifying antirheumatic drugs and risk of incident interstitial lung disease among patients with rheumatoid arthritis: A systematic review and meta-analysis. Semin Arthritis Rheum 2024; 69:152561. [PMID: 39413452 PMCID: PMC11606763 DOI: 10.1016/j.semarthrit.2024.152561] [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: 05/25/2024] [Revised: 08/13/2024] [Accepted: 09/16/2024] [Indexed: 10/18/2024]
Abstract
OBJECTIVE To investigate the association of disease-modifying antirheumatic drugs (DMARDs) and risk of incident interstitial lung disease (ILD) among patients with rheumatoid arthritis (RA) using a systematic literature review and meta-analysis. METHODS We performed a systematic literature review and meta-analysis of studies examining the association of DMARDs with incident RA-ILD. PubMed, Embase, Web of Science, and Cochrane Library were searched from inception to November 2023 for randomized controlled trials (RCTs), observational studies, and post-marketing surveillance studies that investigated adults with RA and compared DMARDs of interest with placebo, no DMARDs, or other DMARDs. The outcome was incident ILD. We summarized the literature on DMARDs and incident RA-ILD risk. Among studies with sufficient quality, we performed meta-analyses to obtain odds ratios (OR) and 95 % confidence intervals (95 %CI) using the Mantel-Haenszel method. RESULTS Among 3,612 studies, we identified a total of 40 papers that encompassed 486,465 patients with RA and 3,928 incident ILD outcomes that were included in the final systematic review and meta-analysis. Among the studies, 24 were RCTs, 4 were prospective cohort studies, 9 were retrospective cohort studies, 2 were case-control studies, and 1 was a post-marketing surveillance study. The pooled analysis from RCTs revealed no statistically significant difference in the odds of ILD development for any specific DMARD across all comparisons examined. The largest identified RCT (Oral Surveillance trial) of tofacitinib (n = 2,911) vs. tumor necrosis factor inhibitor (TNFi, n = 1,451) found no relationship with incident ILD (OR 0.94, 95 %CI 0.52 to 1.69, p = 0.828). In 7 observational studies, the use of methotrexate (MTX) yielded a pooled OR for ILD of 0.49 (95 %CI 0.32 to 0.76, p < 0.001) compared to those not using MTX. In a single observational study, tofacitinib users had an OR for ILD of 0.36 (95 %CI 0.15 to 0.87, p = 0.024) compared to TNFi users. CONCLUSION Observational data suggest no increased risk for any DMARD for incident RA-ILD risk, and perhaps a potential protective role of MTX and tofacitinib. However, these studies may be susceptible to bias, and no specific DMARD showed associations with incident RA-ILD in RCTs. Further well-designed prospective studies are warranted for definitive conclusions on the potential relationship between DMARDs and RA-ILD risk.
Collapse
Affiliation(s)
- Qianru Zhang
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, MA, USA; Department of Rheumatology and Immunology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China; Harvard Medical School, Boston, Massachusetts, USA
| | - Gregory C McDermott
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, MA, USA; Harvard Medical School, Boston, Massachusetts, USA
| | - Pierre-Antoine Juge
- INSERM UMR 1152, Université Paris Cité, Paris, Île-de-France, France; Service de Rhumatologie, Hôpital Bichat - Claude-Bernard, AP-HP, Paris, Île-de-France, France
| | - Sung Hae Chang
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, MA, USA; Soonchunhyang University, Division of Rheumatology, Department of Internal Medicine, Cheonan, Korea, Rep. of (South Korea)
| | - Kathleen Mm Vanni
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, MA, USA
| | - Grace Qian
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, MA, USA
| | - Katarina J Bade
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, MA, USA
| | - Kevin T Mueller
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, MA, USA
| | - Emily N Kowalski
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, MA, USA
| | - Alene A Saavedra
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, MA, USA
| | - Jeffrey A Sparks
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, MA, USA; Harvard Medical School, Boston, Massachusetts, USA.
| |
Collapse
|
26
|
Li P, Huang D. Targeting the JAK-STAT pathway in colorectal cancer: mechanisms, clinical implications, and therapeutic potential. Front Cell Dev Biol 2024; 12:1507621. [PMID: 39659524 PMCID: PMC11628519 DOI: 10.3389/fcell.2024.1507621] [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: 10/08/2024] [Accepted: 11/11/2024] [Indexed: 12/12/2024] Open
Abstract
Colorectal cancer (CRC) remains one of the most prevalent and fatal malignancies worldwide, consistently ranking among the top three in terms of incidence and mortality. Despite notable advancements in early detection and therapeutic interventions, survival outcomes for advanced-stage CRC are still dismal, largely due to issues such as drug resistance and metastasis. Recent research has increasingly implicated the JAK-STAT signaling pathway as a pivotal contributor to CRC pathogenesis. This evolutionarily conserved pathway plays a key role in transmitting extracellular signals to the nucleus, thereby modulating gene expression involved in numerous fundamental biological processes. In CRC, dysregulation of the JAK-STAT pathway is frequently observed and is strongly associated with tumor progression, including processes such as cellular proliferation, apoptosis, metastasis, immune evasion, and the sustenance of cancer stem cells. Given its integral role in CRC advancement, the JAK-STAT pathway has gained recognition as a viable therapeutic target. Extensive evidence from preclinical and clinical models supports the efficacy and safety of targeting components of the JAK-STAT pathway, presenting new therapeutic possibilities for patients with CRC, particularly in addressing drug resistance and enhancing treatment outcomes. This review offers a detailed exploration of the JAK-STAT pathway, focusing on its regulatory mechanisms in CRC-related malignancies. Moreover, it examines the association between JAK-STAT protein expression, clinical features, prognosis, and its therapeutic potential in CRC management.
Collapse
Affiliation(s)
- Penghui Li
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, Henan, China
| | - Di Huang
- Department of Child Health Care, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
27
|
Lei X, Zou F, Tang X, He F, Wang J, Cheng S, Lei X. CD3D silencing alleviates diabetic nephropathy via inhibition of JAK/STAT pathway. FASEB J 2024; 38:e70169. [PMID: 39530557 DOI: 10.1096/fj.202401879r] [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: 08/13/2024] [Revised: 10/17/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024]
Abstract
Diabetic nephropathy (DN) is a severe microvascular complication of diabetes that poses a significant burden to global health. This investigation aims to illustrate the functional role of CD3D and its relevant mechanisms in DN progression. The pivotal genes between the GSE47183 and GSE30528 datasets were identified using bioinformatics methods. The effects of CD3D silencing on renal damage, inflammatory response, and lipid metabolism were validated in DN mice. Furthermore, the impacts of CD3D knockdown on cell viability, apoptotic rate, inflammation, and lipid levels were investigated in HK-2 cells under high glucose (HG) conditions. Additionally, RO8191 was employed to investigate the role of CD3D in the JAK/STAT pathway in HG-treated cells. A total of 5 focal genes were identified through bioinformatics and were found to be upregulated in renal tissues from DN mice. CD3D silencing mitigated pathological damage to kidneys, reduced inflammatory response, and decreased lipid accumulation in DN mice. HG stimulation restrained viability, increased apoptosis, promoted the release of inflammatory cytokines, and affected expressions of hallmarks related to lipid metabolism in HG-treated cells; these changes were partially abolished by CD3D knockdown. Mechanistically, CD3D downregulation ameliorated HG-induced injury in HK-2 cells by blocking the JAK/STAT pathway. This study underscores that CD3D silencing has significant potential as a promising candidate in the treatment of DN.
Collapse
Affiliation(s)
- Xianghong Lei
- Department of Nephrology, the First Affiliated Hospital of Gannan Medical University, Ganzhou City, China
| | - Fangqin Zou
- Department of Nephrology, the First Affiliated Hospital of Gannan Medical University, Ganzhou City, China
| | - Xianhu Tang
- Department of Nephrology, the First Affiliated Hospital of Gannan Medical University, Ganzhou City, China
| | - Fengxia He
- Department of Nephrology, the First Affiliated Hospital of Gannan Medical University, Ganzhou City, China
| | - Jiyang Wang
- Department of Nephrology, the First Affiliated Hospital of Gannan Medical University, Ganzhou City, China
| | - Shengyu Cheng
- Department of Nephrology, the First Affiliated Hospital of Gannan Medical University, Ganzhou City, China
| | - Xiangxin Lei
- School of Information Science and Engineering, East China University of Science and Technology, Shanghai, China
| |
Collapse
|
28
|
Chen H, Xu X, Li J, Xue Y, Li X, Zhang K, Jiang H, Liu X, Li M. Decoding tumor-fibrosis interplay: mechanisms, impact on progression, and innovative therapeutic strategies. Front Pharmacol 2024; 15:1491400. [PMID: 39534084 PMCID: PMC11555290 DOI: 10.3389/fphar.2024.1491400] [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: 09/04/2024] [Accepted: 10/09/2024] [Indexed: 11/16/2024] Open
Abstract
Malignant tumors are a category of diseases that possess invasive and metastatic capabilities, with global incidence and mortality rates remaining high. In recent years, the pivotal role of fibrosis in tumor progression, drug resistance, and immune evasion has increasingly been acknowledged. Fibrosis enhances the proliferation, migration, and invasion of tumor cells by modifying the composition and structure of the extracellular matrix, thereby offering protection for immune evasion by tumor cells. The activation of cancer-associated fibroblasts (CAFs) plays a significant role in this process, as they further exacerbate the malignant traits of tumors by secreting a variety of cytokines and growth factors. Anti-fibrotic tumor treatment strategies, including the use of anti-fibrotic drugs and inhibition of fibrosis-related signaling pathways such as Transforming Growth Factor-β (TGF-β), have demonstrated potential in delaying tumor progression and improving the effectiveness of chemotherapy, targeted therapy, and immunotherapy. In the future, by developing novel drugs that target the fibrotic microenvironment, new therapeutic options may be available for patients with various refractory tumors.
Collapse
Affiliation(s)
- Huiguang Chen
- Institute of Infection, Immunology, and Tumor Microenvironment, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Xuexin Xu
- Institute of Infection, Immunology, and Tumor Microenvironment, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Jingxian Li
- Institute of Infection, Immunology, and Tumor Microenvironment, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Yu Xue
- Institute of Infection, Immunology, and Tumor Microenvironment, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Xin Li
- Institute of Infection, Immunology, and Tumor Microenvironment, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Kaiyu Zhang
- Institute of Infection, Immunology, and Tumor Microenvironment, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Haihui Jiang
- Institute of Infection, Immunology, and Tumor Microenvironment, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Xiaoliu Liu
- Institute of Infection, Immunology, and Tumor Microenvironment, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
- Department of Anatomy, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Mingzhe Li
- Department of Anatomy, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| |
Collapse
|
29
|
Cao Y, Tan YJ, Huang D. Molecular Mechanism of 5,6-Dihydroxyflavone in Suppressing LPS-Induced Inflammation and Oxidative Stress. Int J Mol Sci 2024; 25:10694. [PMID: 39409020 PMCID: PMC11477439 DOI: 10.3390/ijms251910694] [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: 08/26/2024] [Revised: 10/01/2024] [Accepted: 10/02/2024] [Indexed: 10/20/2024] Open
Abstract
5,6-dihydroxyflavone (5,6-DHF), a flavonoid that possesses potential anti-inflammatory and antioxidant activities owing to its special catechol motif on the A ring. However, its function and mechanism of action against inflammation and cellular oxidative stress have not been elucidated. In the current study, 5,6-DHF was observed inhibiting lipopolysaccharide (LPS)-induced nitric oxide (NO) and cytoplasmic reactive oxygen species (ROS) production with the IC50 of 11.55 ± 0.64 μM and 0.8310 ± 0.633 μM in murine macrophages, respectively. Meanwhile, 5,6-DHF suppressed the overexpression of pro-inflammatory mediators such as proteins and cytokines and eradicated the accumulation of mitochondrial ROS (mtROS). The blockage of the activation of cell surface toll-like receptor 4 (TLR4), impediment of the phosphorylation of c-Jun N-terminal kinase (JNK) and p38 from the mitogen-activated protein kinases (MAPK) pathway, Janus kinase 2 (JAK2) and signal transducer and activator of transcription 3 (STAT3) from the JAK-STAT pathway, and p65 from nuclear factor-κB (NF-κB) pathways were involved in the process of 5,6-DHF suppressing inflammation. Furthermore, 5,6-DHF acted as a cellular ROS scavenger and heme-oxygenase 1 (HO-1) inducer in relieving cellular oxidative stress. Importantly, 5,6-DHF exerted more potent anti-inflammatory activity than its close structural relatives, such as baicalein and chrysin. Overall, our findings pave the road for further research on 5,6-DHF in animal models.
Collapse
Affiliation(s)
- Yujia Cao
- Department of Food Science and Technology, National University of Singapore, Singapore 117542, Singapore;
| | - Yee-Joo Tan
- Infectious Diseases Translational Research Program, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117545, Singapore;
| | - Dejian Huang
- Department of Food Science and Technology, National University of Singapore, Singapore 117542, Singapore;
- National University of Singapore (Suzhou) Research Institute, 377 Linquan Street, Suzhou 215123, China
| |
Collapse
|
30
|
Chen L, Kong X, Zhou R, Hu J, Zhou R, Song Z, Tang Z, Wang M. Proteomics reveals the pharmacological mechanism of flavonoids from Astragali Complanati Semen in preventing chronic liver injury. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 133:155910. [PMID: 39059265 DOI: 10.1016/j.phymed.2024.155910] [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: 04/02/2024] [Revised: 07/19/2024] [Accepted: 07/20/2024] [Indexed: 07/28/2024]
Abstract
BACKGROUND Total flavonoids from Astragali Complanati Semen (TFACS), the main active ingredients in Astragali Complanati Semen (ACS), have been shown to have a protective effect on chronic liver injury (CLI), but the hepatoprotective targets and signalling pathways involved are unclear. PURPOSE The aim of our study was to identify the anti-CLI targets and signalling pathways of TFACS and to comprehensively elucidate its mechanism of action via proteomics analysis combined with in vivo and in vitro experiments. METHODS A CLI mouse model was generated via intraperitoneal injection of carbon tetrachloride (CCl4) (CCl4: olive oil = 1:4, 2 ml/kg, twice a week for 6 weeks). The hepatoprotective effect of TFACS was assessed by observing the pathological structure of the liver and analysing indicators of liver function. The key pathways and targets related to the hepatoprotective effect of TFACS were identified via 4D-label-free quantitative proteomics technology and further verified via in vivo indicator validation and in vitro cell experiments. RESULTS TFACS administration significantly normalized the histopathological structure and function of the liver, decreased the levels of inflammatory factors and oxidative stress indicators, and reduced the iron staining area and the levels of hepcidin and iron in the liver compared with those in the CLI model. A total of 424 differentially expressed proteins (DEPs) were identified between the TFACS and model groups, and these DEPs were enriched in the focal adhesion, PI3K-Akt, and ferroptosis pathways. Akt1, Pik3ca, NF-κB p65, Itga5, Itgb5, Itga6, Prkca, Fn1, Tfrc, and Vdac3 were identified as key targets of TFACS. TFACS administration significantly reversed the changes in the gene and protein expression of the key targets compared with those in the model group. In addition, TFACS treatment significantly reduced the levels of inflammatory cytokines and inhibited Akt1, NF-κB p65 and FAK activation in lipopolysaccharide (LPS)-induced RAW 264.7 macrophages. In an erastin-induced l-O2 ferroptosis cell model, treatment with TFACS normalized the mitochondrial structure, reduced the protein levels of Tfrc and Vdac3, inhibited lipid peroxidation, and reduced the amount of Fe2+ in the mitochondria. CONCLUSION TFACS protected against CLI, and its mechanism of action may be related to inhibition of the focal adhesion, PI3K/Akt and ferroptosis signalling pathways.
Collapse
Affiliation(s)
- Lin Chen
- Co-construction Collaborative Innovation Center for Chinese Medicine Resources Industrialization by Shaanxi & Education Ministry, State Key Laboratory of Research, & Development of Characteristic Qin Medicine Resources (Cultivation), Shaanxi Innovative Drug Research Center, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi 712083, PR China
| | - Xin Kong
- College of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an, Shaanxi 712046, PR China
| | - Ruina Zhou
- College of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an, Shaanxi 712046, PR China
| | - Jinhang Hu
- Co-construction Collaborative Innovation Center for Chinese Medicine Resources Industrialization by Shaanxi & Education Ministry, State Key Laboratory of Research, & Development of Characteristic Qin Medicine Resources (Cultivation), Shaanxi Innovative Drug Research Center, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi 712083, PR China
| | - Rui Zhou
- Co-construction Collaborative Innovation Center for Chinese Medicine Resources Industrialization by Shaanxi & Education Ministry, State Key Laboratory of Research, & Development of Characteristic Qin Medicine Resources (Cultivation), Shaanxi Innovative Drug Research Center, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi 712083, PR China
| | - Zhongxing Song
- Co-construction Collaborative Innovation Center for Chinese Medicine Resources Industrialization by Shaanxi & Education Ministry, State Key Laboratory of Research, & Development of Characteristic Qin Medicine Resources (Cultivation), Shaanxi Innovative Drug Research Center, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi 712083, PR China
| | - Zhishu Tang
- Co-construction Collaborative Innovation Center for Chinese Medicine Resources Industrialization by Shaanxi & Education Ministry, State Key Laboratory of Research, & Development of Characteristic Qin Medicine Resources (Cultivation), Shaanxi Innovative Drug Research Center, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi 712083, PR China; Beijing University of Chinese Medicine, Beijing 100700, PR China.
| | - Mei Wang
- Co-construction Collaborative Innovation Center for Chinese Medicine Resources Industrialization by Shaanxi & Education Ministry, State Key Laboratory of Research, & Development of Characteristic Qin Medicine Resources (Cultivation), Shaanxi Innovative Drug Research Center, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi 712083, PR China; Academic Development Office, Wangjing Hospital of China Academy of Chinese Medical Sciences, Beijing 100700, PR China.
| |
Collapse
|
31
|
Li DJ, Berry CE, Wan DC, Longaker MT. Clinical, mechanistic, and therapeutic landscape of cutaneous fibrosis. Sci Transl Med 2024; 16:eadn7871. [PMID: 39321265 DOI: 10.1126/scitranslmed.adn7871] [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: 12/29/2023] [Accepted: 09/03/2024] [Indexed: 09/27/2024]
Abstract
When dysregulated, skin fibrosis can lead to a multitude of pathologies. We provide a framework for understanding the wide clinical spectrum, mechanisms, and management of cutaneous fibrosis encompassing a variety of matrix disorders, fibrohistiocytic neoplasms, injury-induced scarring, and autoimmune scleroses. Underlying such entities are common mechanistic pathways that leverage morphogenic signaling, immune activation, and mechanotransduction to modulate fibroblast function. In light of the limited array of available treatments for cutaneous fibrosis, scientific insights have opened new therapeutic and investigative avenues for conditions that still lack effective interventions.
Collapse
Affiliation(s)
- Dayan J Li
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Dermatology, Stanford University School of Medicine, Redwood City, CA 94063, USA
| | - Charlotte E Berry
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Derrick C Wan
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Michael T Longaker
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
32
|
Hadifar S, Masoudzadeh N, Heydari H, Mashayekhi Goyonlo V, Kerachian M, Daneshpazhooh M, Sadeghnia A, Tootoonchi N, Erfanian Salim R, Rafati S, Harandi AM. Intralesional gene expression profile of JAK-STAT signaling pathway and associated cytokines in Leishmania tropica-infected patients. Front Immunol 2024; 15:1436029. [PMID: 39364404 PMCID: PMC11446769 DOI: 10.3389/fimmu.2024.1436029] [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: 05/21/2024] [Accepted: 09/02/2024] [Indexed: 10/05/2024] Open
Abstract
Background The JAK-STAT signaling pathway is a central cascade of signal transduction for the myriad of cytokines in which dysregulation has been implicated in progression of inflammatory and infectious diseases. However, the involvement of this pathway in human cutaneous leishmaniasis (CL) due to Leishmania (L.) tropica warrants further investigation. Methods This study sought to investigate differential gene expression of several cytokines and their associated jak-stat genes in the lesions of L. tropica-infected patients byquantitative Real-Time PCR. Further, the expression of five inhibitory immune checkpoint genes was evaluated. Results Results showed that the gene expression levelsof both Th1 (ifng, il12, il23) and Th2 (il4, il10) types cytokines were increased in the lesion of studied patients. Further, elevated expression levels of il35, il21, il27 and il24 genes were detected in the lesions of CL patients. Notably, the expression of the majority of genes involved in JAK/STAT signaling pathway as well as checkpoint genes including pdl1, ctla4 and their corresponding receptors was increased. Conclusion Our finding revealed dysregulation of cytokines and related jak-stat genes in the lesion of CL patients. These results highlight the need for further exploration of the functional importance of these genes in the pathogenesis of, and immunity to, CL.
Collapse
Affiliation(s)
- Shima Hadifar
- Department of Immunotherapy and Leishmania Vaccine Research, Pasteur Institute of Iran, Tehran, Iran
| | - Nasrin Masoudzadeh
- Department of Immunotherapy and Leishmania Vaccine Research, Pasteur Institute of Iran, Tehran, Iran
| | - Hossein Heydari
- Department of Immunotherapy and Leishmania Vaccine Research, Pasteur Institute of Iran, Tehran, Iran
| | | | - Mohammadali Kerachian
- Cutaneous Leishmaniasis Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maryam Daneshpazhooh
- Autoimmune Bullous Disease Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | | | | | | | - Sima Rafati
- Department of Immunotherapy and Leishmania Vaccine Research, Pasteur Institute of Iran, Tehran, Iran
| | - Ali M. Harandi
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
33
|
Zhang Q, Fu S, Li X, Wang H, Wang J. STAT1 as a potential therapeutic target to treat bladder cancer. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2024; 17:298-307. [PMID: 39399655 PMCID: PMC11470427 DOI: 10.62347/hycn1717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Accepted: 07/10/2024] [Indexed: 10/15/2024]
Abstract
BACKGROUND Previous studies have reported that STAT1 (Signal Transducer and Activator of Transcription 1) is associated with multiple tumor progression. This study aimed to investigate the role and related mechanisms of STAT1 in bladder cancer. METHODS STAT1 expression in bladder cancer tissues and human bladder cancer cell lines was assessed by reverse transcription-quantitative polymerase chain reaction (RT-qPCR). The bladder cancer cell line T24 was transfected with overexpressing lentivirus targeting STAT1. Cell proliferation, invasion, and apoptosis were measured by Cell Counting Kit-8, Transwell assays, and flow cytometric analysis. Furthermore, RNA-Seq was performed to identify the downstream signaling pathways. Finally, the signaling pathway-related molecules were determined by RT-qPCR and western blot assays. RESULTS The overexpression of STAT1 inhibited bladder cancer cell proliferation and invasion while enhancing apoptosis. Moreover, the overexpression of STAT1 in bladder cancer cells delayed tumor tumorigenesis in vitro. Mechanistically, RNA-Seq analysis revealed that the JAK-STAT signaling pathway was up-regulated, especially SOCS1 (suppressor of cytokine signaling 1) and SOCS3 (suppressor of cytokine signaling 3) in STAT1-sufficient cells. CONCLUSIONS These results indicate the potential of STAT1 as a therapeutic target in bladder cancer.
Collapse
Affiliation(s)
- Qin Zhang
- Department of Gynaecology, The Second Affiliated Hospital of Kunming Medical UniversityKunming 650101, Yunnan, China
| | - Shi Fu
- Department of Gynaecology, The Second Affiliated Hospital of Kunming Medical UniversityKunming 650101, Yunnan, China
| | - Xiaotao Li
- Department of Andriatrics, The Second Affiliated Hospital of Kunming Medical UniversityKunming 650101, Yunnan, China
| | - Haifeng Wang
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Yunnan Institute of UrologyKunming 650101, Yunnan, China
| | - Jiansong Wang
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Yunnan Institute of UrologyKunming 650101, Yunnan, China
| |
Collapse
|
34
|
Abdel-Wahab BA, Zafaar D, Habeeb MS, El-Shoura EAM. Nicorandil mitigates arsenic trioxide-induced lung injury via modulating vital signalling pathways SIRT1/PGC-1α/TFAM, JAK1/STAT3, and miRNA-132 expression. Br J Pharmacol 2024; 181:3215-3231. [PMID: 38741475 DOI: 10.1111/bph.16414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 01/13/2024] [Accepted: 03/01/2024] [Indexed: 05/16/2024] Open
Abstract
BACKGROUND AND PURPOSE Nicorandil, a selective opener of potassium channels, used to treat angina, has drawn attention for its potential in mitigating lung injury, positioning it as a promising therapeutic approach to treat drug-induced lung toxicity. This study aimed to explore the protective role of nicorandil in arsenic trioxide (ATO)-induced lung injury and to elucidate the underlying mechanistic pathways. EXPERIMENTAL APPROACH We assessed the effects of nicorandil (15 mg·kg-1, p.o.) in a rat model of pulmonary injury induced by ATO (5 mg·kg-1, i.p.). The assessment included oxidative stress biomarkers, inflammatory cytokine levels, and other biomarkers, including sirtuin-1, sirtuin-3, STAT3, TFAM, and JAK in lung tissue. Histological examination using H&E staining and molecular investigations using western blotting and PCR techniques were conducted. KEY RESULTS In our model of lung injury, treatment with nicorandil ameliorated pathological changes as seen with H&E staining, reduced tissue levels of toxicity markers, and exerted significant antioxidant and anti-inflammatory actions. On a molecular level, treatment with nicorandil down-regulated JAK, STAT3, PPARγ, Nrf2, VEGF, p53, and micro-RNA 132 while up-regulating Sirt1, 3, TFAM, AMPK, and ERR-α in lung tissue. CONCLUSIONS AND IMPLICATIONS The results presented here show nicorandil as a significant agent in attenuating lung injury induced by ATO in a rodent model. Nonetheless, further clinical studies are warranted to strengthen these findings.
Collapse
Affiliation(s)
- Basel A Abdel-Wahab
- Department of Pharmacology, College of Pharmacy, Najran University, Najran, Saudi Arabia
| | - Dalia Zafaar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Modern University of Technology, and Information, Cairo, Egypt
| | | | - Ehab A M El-Shoura
- Department of Clinical Pharmacy, Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut, Egypt
| |
Collapse
|
35
|
Huang H, Peng Z, Yuan Q. Research progress in anti-renal fibrosis drugs. ZHONG NAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF CENTRAL SOUTH UNIVERSITY. MEDICAL SCIENCES 2024; 49:1353-1362. [PMID: 39788524 PMCID: PMC11628227 DOI: 10.11817/j.issn.1672-7347.2024.240284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Indexed: 01/12/2025]
Abstract
Renal fibrosis is the common pathological basis for the progressive development of chronic kidney disease (CKD) caused by various etiologies. It is characterized by the persistent deposition of extracellular matrix, leading to renal tissue damage and impaired renal function, and ultimately progressing to kidney failure. Current clinical treatments for CKD mainly focus on managing the primary diseases, with no specific drugs targeting renal fibrosis. The pathogenesis of renal fibrosis is complex, and there are currently no drugs available to reverse it. A comprehensive overview of the pathogenesis of renal fibrosis, alongside a summary of current anti-fibrotic therapies, including some that are already used clinically to slow renal function progression, new drugs in clinical trials, and emerging targeted therapies, could provide new theoretical foundations and perspectives for the treatment of renal fibrosis.
Collapse
Affiliation(s)
- Hanwei Huang
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha 410008.
- Hunan Provincial Key Laboratory of Organ Fibrosis, Changsha 410008, China.
| | - Zhangzhe Peng
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha 410008
- Hunan Provincial Key Laboratory of Organ Fibrosis, Changsha 410008, China
| | - Qiongjing Yuan
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha 410008.
- Hunan Provincial Key Laboratory of Organ Fibrosis, Changsha 410008, China.
| |
Collapse
|
36
|
Harris DD, Sabe SA, Broadwin M, Stone C, Bellam K, Malhotra A, Abid MR, Sellke FW. Dipeptidyl peptidase 4 inhibitor sitagliptin decreases myocardial fibrosis and modulates myocardial insulin signaling in a swine model of chronic myocardial ischemia. PLoS One 2024; 19:e0307922. [PMID: 39074126 PMCID: PMC11285952 DOI: 10.1371/journal.pone.0307922] [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: 03/14/2024] [Accepted: 07/09/2024] [Indexed: 07/31/2024] Open
Abstract
Although both clinical data and animal models suggest cardiovascular benefits following administration of Dipeptidyl Peptidase 4 (DPP-4) inhibitors, the underlying mechanisms remain unclear. We therefore sought to evaluate the effect of the DPP-4 inhibitor sitagliptin on myocardial fibrosis, and insulin signaling in chronic myocardial ischemia using a swine model. An ameroid constrictor placement on the left coronary circumflex artery of thirteen Yorkshire swine to model chronic myocardial ischemia. After two weeks of recovery, swine were assigned to one of two groups: control (CON, n = 8), or sitagliptin 100mg daily (SIT, n = 5). After 5 weeks of treatment, the swine underwent terminal harvest with collection of myocardial tissue. Fibrosis was quantified using Masson's trichrome. Protein expression was quantified by Immunoblotting. Trichrome stain demonstrated a significant decrease in perivascular and interstitial fibrosis in the SIT group relative to CON (all p<0.05). Immunoblot showed a reduction in Jak2, the pSTAT3 to STAT 3 Ratio, pSMAD 2/3, and SMAD 2/3, and an increase in STAT 3 in the SIT group relative to CON (all p<0.05). SIT treatment was associated with increased expression of insulin receptor one and decreased expression of makers for insulin resistance, including phospho-PKC- alpha, RBP-4, SIRT1, and PI3K (p<0.05). Sitagliptin results in a reduction in perivascular and interstitial fibrosis and increased insulin sensitivity in chronically ischemic swine myocardium. This likely contributes to the improved cardiovascular outcomes seen with DPP-4 inhibitors.
Collapse
Affiliation(s)
- Dwight D. Harris
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI, United States of America
| | - Sharif A. Sabe
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI, United States of America
| | - Mark Broadwin
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI, United States of America
| | - Chris Stone
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI, United States of America
| | - Krishna Bellam
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI, United States of America
| | - Akshay Malhotra
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI, United States of America
| | - M. Ruhul Abid
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI, United States of America
| | - Frank W. Sellke
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI, United States of America
| |
Collapse
|
37
|
Zhuang Z, Jia W, Wu L, Li Y, Lu Y, Xu M, Bai H, Bi Y, Wang Z, Chen S, Jiang Y, Chang G. Threonine Deficiency Increases Triglyceride Deposition in Primary Duck Hepatocytes by Reducing STAT3 Phosphorylation. Int J Mol Sci 2024; 25:8142. [PMID: 39125712 PMCID: PMC11312044 DOI: 10.3390/ijms25158142] [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: 06/02/2024] [Revised: 06/24/2024] [Accepted: 06/26/2024] [Indexed: 08/12/2024] Open
Abstract
Liver lipid metabolism disruption significantly contributes to excessive fat buildup in waterfowl. Research suggests that the supplementation of Threonine (Thr) in the diet can improve liver lipid metabolism disorder, while Thr deficiency can lead to such metabolic disorders in the liver. The mechanisms through which Thr regulates lipid metabolism remain unclear. STAT3 (signal transducer and activator of transcription 3), a crucial transcription factor in the JAK-STAT (Janus kinase-signal transducer and activator of transcription) pathway, participates in various biological processes, including lipid and energy metabolism. This research investigates the potential involvement of STAT3 in the increased lipid storage seen in primary duck hepatocytes as a result of a lack of Thr. Using small interfering RNA and Stattic, a specific STAT3 phosphorylation inhibitor, we explored the impact of STAT3 expression patterns on Thr-regulated lipid synthesis metabolism in hepatocytes. Through transcriptome sequencing, we uncovered pathways related to lipid synthesis and metabolism jointly regulated by Thr and STAT3. The results showed that Thr deficiency increases lipid deposition in primary duck hepatocytes (p < 0.01). The decrease in protein and phosphorylation levels of STAT3 directly caused this deposition (p < 0.01). Transcriptomic analysis revealed that Thr deficiency and STAT3 knockdown jointly altered the mRNA expression levels of pathways related to long-chain fatty acid synthesis and energy metabolism (p < 0.05). Thr deficiency, through mediating STAT3 inactivation, upregulated ELOVL7, PPARG, MMP1, MMP13, and TIMP4 mRNA levels, and downregulated PTGS2 mRNA levels (p < 0.01). In summary, these results suggest that Thr deficiency promotes lipid synthesis, reduces lipid breakdown, and leads to lipid metabolism disorders and triglyceride deposition by downregulating STAT3 activity in primary duck hepatocytes.
Collapse
Affiliation(s)
- Zhong Zhuang
- Key Laboratory for Animal Genetics & Molecular Breeding of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (Z.Z.); (W.J.); (L.W.); (Y.L.); (Y.L.); (M.X.); (Y.B.); (Z.W.); (S.C.); (G.C.)
| | - Wenqian Jia
- Key Laboratory for Animal Genetics & Molecular Breeding of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (Z.Z.); (W.J.); (L.W.); (Y.L.); (Y.L.); (M.X.); (Y.B.); (Z.W.); (S.C.); (G.C.)
| | - Lei Wu
- Key Laboratory for Animal Genetics & Molecular Breeding of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (Z.Z.); (W.J.); (L.W.); (Y.L.); (Y.L.); (M.X.); (Y.B.); (Z.W.); (S.C.); (G.C.)
| | - Yongpeng Li
- Key Laboratory for Animal Genetics & Molecular Breeding of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (Z.Z.); (W.J.); (L.W.); (Y.L.); (Y.L.); (M.X.); (Y.B.); (Z.W.); (S.C.); (G.C.)
| | - Yijia Lu
- Key Laboratory for Animal Genetics & Molecular Breeding of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (Z.Z.); (W.J.); (L.W.); (Y.L.); (Y.L.); (M.X.); (Y.B.); (Z.W.); (S.C.); (G.C.)
| | - Minghong Xu
- Key Laboratory for Animal Genetics & Molecular Breeding of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (Z.Z.); (W.J.); (L.W.); (Y.L.); (Y.L.); (M.X.); (Y.B.); (Z.W.); (S.C.); (G.C.)
| | - Hao Bai
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou 225009, China;
| | - Yulin Bi
- Key Laboratory for Animal Genetics & Molecular Breeding of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (Z.Z.); (W.J.); (L.W.); (Y.L.); (Y.L.); (M.X.); (Y.B.); (Z.W.); (S.C.); (G.C.)
| | - Zhixiu Wang
- Key Laboratory for Animal Genetics & Molecular Breeding of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (Z.Z.); (W.J.); (L.W.); (Y.L.); (Y.L.); (M.X.); (Y.B.); (Z.W.); (S.C.); (G.C.)
| | - Shihao Chen
- Key Laboratory for Animal Genetics & Molecular Breeding of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (Z.Z.); (W.J.); (L.W.); (Y.L.); (Y.L.); (M.X.); (Y.B.); (Z.W.); (S.C.); (G.C.)
| | - Yong Jiang
- Key Laboratory for Animal Genetics & Molecular Breeding of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (Z.Z.); (W.J.); (L.W.); (Y.L.); (Y.L.); (M.X.); (Y.B.); (Z.W.); (S.C.); (G.C.)
| | - Guobin Chang
- Key Laboratory for Animal Genetics & Molecular Breeding of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (Z.Z.); (W.J.); (L.W.); (Y.L.); (Y.L.); (M.X.); (Y.B.); (Z.W.); (S.C.); (G.C.)
| |
Collapse
|
38
|
Yang J, Zhang YN, Wang RX, Hao CZ, Qiu Y, Chi H, Luan WS, Tang H, Zhang XJ, Sun X, Sheps JA, Ling V, Cao M, Wang JS. ZFYVE19 deficiency: a ciliopathy involving failure of cell division, with cell death. J Med Genet 2024; 61:750-758. [PMID: 38816193 PMCID: PMC11287636 DOI: 10.1136/jmg-2023-109779] [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: 11/30/2023] [Accepted: 04/04/2024] [Indexed: 06/01/2024]
Abstract
BACKGROUND AND AIMS Variants in ZFYVE19 underlie a disorder characterised by progressive portal fibrosis, portal hypertension and eventual liver decompensation. We aim to create an animal model to elucidate the pathogenic mechanism. METHODS Zfyve19 knockout (Zfyve19-/- ) mice were generated and exposed to different liver toxins. Their livers were characterised at the tissue, cellular and molecular levels. Findings were compared with those in wild-type mice and in ZFYVE19-deficient patients. ZFYVE19 knockout and knockdown retinal pigment epithelial-1 cells and mouse embryonic fibroblasts were generated to study cell division and cell death. RESULTS The Zfyve19-/- mice were normal overall, particularly with respect to hepatobiliary features. However, when challenged with α-naphthyl isothiocyanate, Zfyve19-/- mice developed changes resembling those in ZFYVE19-deficient patients, including elevated serum liver injury markers, increased numbers of bile duct profiles with abnormal cholangiocyte polarity and biliary fibrosis. Failure of cell division, centriole and cilia abnormalities, and increased cell death were observed in knockdown/knockout cells. Increased cell death and altered mRNA expression of cell death-related signalling pathways was demonstrated in livers from Zfyve19-/- mice and patients. Transforming growth factor-β (TGF-β) and Janus kinase-Signal Transducer and Activator of Transcription 3 (JAK-STAT3) signalling pathways were upregulated in vivo, as were chemokines such as C-X-C motif ligands 1, 10 and 12. CONCLUSIONS Our findings demonstrated that ZFYVE19 deficiency is a ciliopathy with novel histological features. Failure of cell division with ciliary abnormalities and cell death activates macrophages and may thus lead to biliary fibrosis via TGF-β pathway in the disease.
Collapse
Affiliation(s)
- Jing Yang
- Department of Pediatrics, Jinshan Hospital of Fudan University, Shanghai, China
- The Center for Liver Diseases, Children's Hospital of Fudan University, Shanghai, China
- Department of Pediatric Gastroenterology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Ya-Nan Zhang
- Department of Pediatrics, Jinshan Hospital of Fudan University, Shanghai, China
- The Center for Liver Diseases, Children's Hospital of Fudan University, Shanghai, China
| | - Ren-Xue Wang
- BC Cancer Agency, Vancouver, British Columbia, Canada
| | - Chen-Zhi Hao
- The Center for Liver Diseases, Children's Hospital of Fudan University, Shanghai, China
| | - Yiling Qiu
- Department of Pediatrics, Jinshan Hospital of Fudan University, Shanghai, China
- The Center for Liver Diseases, Children's Hospital of Fudan University, Shanghai, China
| | - Hao Chi
- Department of Pediatrics, Jinshan Hospital of Fudan University, Shanghai, China
| | - Wei-Sha Luan
- Department of Pediatrics, Jinshan Hospital of Fudan University, Shanghai, China
| | - HongYi Tang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiu-Juan Zhang
- Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - XuXu Sun
- Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | | | - Victor Ling
- BC Cancer Agency, Vancouver, British Columbia, Canada
| | - Muqing Cao
- Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jian-She Wang
- The Center for Liver Diseases, Children's Hospital of Fudan University, Shanghai, China
- Shanghai Key Laboratory of Birth Defect, Shanghai, China
| |
Collapse
|
39
|
Kundra S, Kaur R, Pasricha C, Kumari P, Gurjeet Singh T, Singh R. Pathological insights into activin A: Molecular underpinnings and therapeutic prospects in various diseases. Int Immunopharmacol 2024; 139:112709. [PMID: 39032467 DOI: 10.1016/j.intimp.2024.112709] [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: 02/13/2024] [Revised: 05/14/2024] [Accepted: 07/15/2024] [Indexed: 07/23/2024]
Abstract
Activin A (Act A) is a member of the TGFβ (transforming growth factor β) superfamily. It communicates via the Suppressor of Mothers against Decapentaplegic Homolog (SMAD2/3) proteins which govern processes such as cell proliferation, wound healing, apoptosis, and metabolism. Act A produces its action by attaching to activin receptor type IIA (ActRIIA) or activin receptor type IIB (ActRIIB). Increasing circulating Act A increases ActRII signalling, which on phosphorylation initiates the ALK4 (activin receptor-like kinase 4) type 1 receptor which further turns on the SMAD pathway and hinders cell functioning. Once triggered, this route leads to gene transcription, differentiation, apoptosis, and extracellular matrix (ECM) formation. Act A also governs the immunological and inflammatory responses of the body, as well as cell death. Moreover, Act A levels have been observed to elevate in several disorders like renal fibrosis, CKD, asthma, NAFLD, cardiovascular diseases, cancer, inflammatory conditions etc. Here, we provide an update on the recent studies relevant to the role of Act A in the modulation of various pathological disorders, giving an overview of the biology of Act A and its signalling pathways, and discuss the possibility of incorporating activin-A targeting as a novel therapeutic approach for the control of various disorders. Pathways such as SMAD signaling, in which SMAD moves to the nucleus by making a complex and leads to tissue fibrosis in CKD, STAT3, which drives renal fibroblast activity and the production of ECM, Kidney injury molecule (KIM-1) in the synthesis, deposition of ECM proteins, SERCA2a (sarcoplasmic reticulum Ca2+ ATPase) in cardiac dysfunction, and NF-κB (Nuclear factor kappa-light-chain-enhancer of activated B cells) in inflammation are involved in Act A signaling, have also been discussed.
Collapse
Affiliation(s)
- Sejal Kundra
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Rupinder Kaur
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Chirag Pasricha
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Pratima Kumari
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | | | - Ravinder Singh
- Chitkara College of Pharmacy, Chitkara University, Punjab, India.
| |
Collapse
|
40
|
Wu XF, Xu Q, Wang A, Wang BZ, Lan XY, Li WY, Liu Y. Relationship between Indel Variants within the JAK2 Gene and Growth Traits in Goats. Animals (Basel) 2024; 14:1994. [PMID: 38998106 PMCID: PMC11240706 DOI: 10.3390/ani14131994] [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: 05/29/2024] [Revised: 06/28/2024] [Accepted: 07/05/2024] [Indexed: 07/14/2024] Open
Abstract
Janus kinase 2 (JAK2) plays a critical role in myoblast proliferation and fat deposition in animals. Our previous RNA-Seq analyses identified a close association between the JAK2 gene and muscle development. To date, research delving into the relationship between the JAK2 gene and growth traits has been sparse. In this study, we sought to investigate the relationship between novel mutations within the JAK2 gene and goat growth traits. Herein, two novel InDel (Insertion/Deletion) polymorphisms within the JAK2 gene were detected in 548 goats, and only two genotypes were designated as ID (Insertion/Deletion) and DD (Deletion/Deletion). The results indicate that the two InDels, the del19008 locus in intron 2 and del72416 InDel in intron 6, showed significant associations with growth traits (p < 0.05). Compared to Nubian and Jianzhou Daer goats, the del72416 locus displayed a more pronounced effect in the Fuqing breed group. In the Nubian breed (NB) group, both InDels showed a marked influence on body height (BH). There were strong linkages observed for these two InDels between the Fuqing (FQ) and Jianzhou (JZ) populations. The DD-ID diplotype was associated with inferior growth traits in chest width (ChW) and cannon circumference (CaC) in the FQ goats compared to the other diplotypes. In the NB population, the DD-DD diplotype exhibited a marked negative impact on BH and HuWI (hucklebone width index), in contrast to the other diplotypes. In summary, our findings suggest that the two InDel polymorphisms within the JAK2 gene could serve as valuable molecular markers for enhancing goat growth traits in breeding programs.
Collapse
Affiliation(s)
- Xian-Feng Wu
- Fujian Provincial Key Laboratory of Animal Genetics and Breeding/Institute of Animal Husbandry and Veterinary, Fujian Academy of Agricultural Sciences, Fuzhou 350013, China
| | - Qian Xu
- Fujian Provincial Key Laboratory of Animal Genetics and Breeding/Institute of Animal Husbandry and Veterinary, Fujian Academy of Agricultural Sciences, Fuzhou 350013, China
| | - Ao Wang
- College of Animal Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Ben-Zhi Wang
- College of Animal Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Xian-Yong Lan
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling, Xianyang 712100, China
| | - Wen-Yang Li
- Fujian Provincial Key Laboratory of Animal Genetics and Breeding/Institute of Animal Husbandry and Veterinary, Fujian Academy of Agricultural Sciences, Fuzhou 350013, China
| | - Yuan Liu
- Fujian Provincial Key Laboratory of Animal Genetics and Breeding/Institute of Animal Husbandry and Veterinary, Fujian Academy of Agricultural Sciences, Fuzhou 350013, China
| |
Collapse
|
41
|
Chen J, Zhang T, Liu D, Yang F, Feng Y, Wang A, Wang Y, He X, Luo F, Li J, Tan H, Jiang L. General Semi-Solid Freeze Casting for Uniform Large-Scale Isotropic Porous Scaffolds: An Application for Extensive Oral Mucosal Reconstruction. SMALL METHODS 2024; 8:e2301518. [PMID: 38517272 DOI: 10.1002/smtd.202301518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 02/29/2024] [Indexed: 03/23/2024]
Abstract
Ice-templated porous biomaterials possess transformative potential in regenerative medicine; yet, scaling up ice-templating processes for broader applications-owing to inconsistent pore formation-remains challenging. This study reports an innovative semi-solid freeze-casting technique that draws inspiration from semi-solid metal processing (SSMP) combined with ice cream-production routines. This versatile approach allows for the large-scale assembly of various materials, from polymers to inorganic particles, into isotropic 3D scaffolds featuring uniformly equiaxed pores throughout the centimeter scale. Through (cryo-)electron microscopy, X-ray tomography, and finite element modeling, the structural evolution of ice grains/pores is elucidated, demonstrating how the method increases the initial ice nucleus density by pre-fabricating a semi-frozen slurry, which facilitates a transition from columnar to equiaxed grain structures. For a practical demonstration, as-prepared scaffolds are integrated into a bilayer tissue patch using biodegradable waterborne polyurethane (WPU) for large-scale oral mucosal reconstruction in minipigs. Systematic analyses, including histology and RNA sequencing, prove that the patch modulates the healing process toward near-scarless mucosal remodeling via innate and adaptive immunomodulation and activation of pro-healing genes converging on matrix synthesis and epithelialization. This study not only advances the field of ice-templating fabrication but sets a promising precedent for scaffold-based large-scale tissue regeneration.
Collapse
Affiliation(s)
- Jinlin Chen
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Med-X center of materials, Sichuan University, Chengdu, Sichuan, 610065, P. R. China
| | - Tianyu Zhang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
| | - Dan Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
| | - Fan Yang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
| | - Yuan Feng
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Med-X center of materials, Sichuan University, Chengdu, Sichuan, 610065, P. R. China
| | - Ao Wang
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Med-X center of materials, Sichuan University, Chengdu, Sichuan, 610065, P. R. China
| | - Yanchao Wang
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, 610000, P. R. China
| | - Xueling He
- Editorial Board of Journal of Sichuan University (Medical Sciences), Sichuan University, Chengdu, Sichuan, 610000, P. R. China
| | - Feng Luo
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Med-X center of materials, Sichuan University, Chengdu, Sichuan, 610065, P. R. China
| | - Jiehua Li
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Med-X center of materials, Sichuan University, Chengdu, Sichuan, 610065, P. R. China
| | - Hong Tan
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Med-X center of materials, Sichuan University, Chengdu, Sichuan, 610065, P. R. China
| | - Lu Jiang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
| |
Collapse
|
42
|
Song C, Wang G, Liu M, Han S, Dong M, Peng M, Wang W, Wang Y, Xu Y, Liu L. Deciphering the SOX4/MAPK1 regulatory axis: a phosphoproteomic insight into IQGAP1 phosphorylation and pancreatic Cancer progression. J Transl Med 2024; 22:602. [PMID: 38943117 PMCID: PMC11212360 DOI: 10.1186/s12967-024-05377-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Accepted: 06/06/2024] [Indexed: 07/01/2024] Open
Abstract
OBJECTIVE This study aims to elucidate the functional role of IQGAP1 phosphorylation modification mediated by the SOX4/MAPK1 regulatory axis in developing pancreatic cancer through phosphoproteomics analysis. METHODS Proteomics and phosphoproteomics data of pancreatic cancer were obtained from the Clinical Proteomic Tumor Analysis Consortium (CPTAC) database. Differential analysis, kinase-substrate enrichment analysis (KSEA), and independent prognosis analysis were performed on these datasets. Subtype analysis of pancreatic cancer patients was conducted based on the expression of prognostic-related proteins, and the prognosis of different subtypes was evaluated through prognosis analysis. Differential analysis of proteins in different subtypes was performed to identify differential proteins in the high-risk subtype. Clinical correlation analysis was conducted based on the expression of prognostic-related proteins, pancreatic cancer typing results, and clinical characteristics in the pancreatic cancer proteomics dataset. Functional pathway enrichment analysis was performed using GSEA/GO/KEGG, and most module proteins correlated with pancreatic cancer were selected using WGCNA analysis. In cell experiments, pancreatic cancer cells were grouped, and the expression levels of SOX4, MAPK1, and the phosphorylation level of IQGAP1 were detected by RT-qPCR and Western blot experiments. The effect of SOX4 on MAPK1 promoter transcriptional activity was assessed using a dual-luciferase assay, and the enrichment of SOX4 on the MAPK1 promoter was examined using a ChIP assay. The proliferation, migration, and invasion functions of grouped pancreatic cancer cells were assessed using CCK-8, colony formation, and Transwell assays. In animal experiments, the impact of SOX4 on tumor growth and metastasis through the regulation of MAPK1-IQGAP1 phosphorylation modification was studied by constructing subcutaneous and orthotopic pancreatic cancer xenograft models, as well as a liver metastasis model in nude mice. RESULTS Phosphoproteomics and proteomics data analysis revealed that the kinase MAPK1 may play an important role in pancreatic cancer progression by promoting IQGAP1 phosphorylation modification. Proteomics analysis classified pancreatic cancer patients into two subtypes, C1 and C2, where the high-risk C2 subtype was associated with poor prognosis, malignant tumor typing, and enriched tumor-related pathways. SOX4 may promote the occurrence of the high-risk C2 subtype of pancreatic cancer by regulating MAPK1-IQGAP1 phosphorylation modification. In vitro cell experiments confirmed that SOX4 promoted IQGAP1 phosphorylation modification by activating MAPK1 transcription while silencing SOX4 inhibited the proliferation, migration, and invasion of pancreatic cancer cells by reducing the phosphorylation level of MAPK1-IQGAP1. In vivo, animal experiments further confirmed that silencing SOX4 suppressed the growth and metastasis of pancreatic cancer by reducing the phosphorylation level of MAPK1-IQGAP1. CONCLUSION The findings of this study suggest that SOX4 promotes the phosphorylation modification of IQGAP1 by activating MAPK1 transcription, thereby facilitating the growth and metastasis of pancreatic cancer.
Collapse
Affiliation(s)
- Chao Song
- Department of Pancreatic Surgery, Affiliated Zhongshan Hospital, Fudan University, No.180 Fenglin Road, Xuhui District, Shanghai, PR China
- Department of General Surgery, Qingpu Branch, Affiliated Zhongshan Hospital of Fudan University, Qingpu Branch, No. 1158 Park Road East, Qingpu District, Shanghai, PR China
- Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, PR China
| | - Ganggang Wang
- Department of Hepatobiliary Surgery, Pudong Hospital, Fudan University, Shanghai, China
| | - Mengmeng Liu
- Department of Gastroenterology, Qingpu Branch, Affiliated Zhongshan Hospital of Fudan University, Shanghai, PR China
| | - Siyang Han
- Department of Pancreatic Surgery, Affiliated Zhongshan Hospital, Fudan University, No.180 Fenglin Road, Xuhui District, Shanghai, PR China
| | - Meiyuan Dong
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, PR China
| | - Maozhen Peng
- Department of Pancreatic Surgery, Affiliated Zhongshan Hospital, Fudan University, No.180 Fenglin Road, Xuhui District, Shanghai, PR China
| | - Wenquan Wang
- Department of Pancreatic Surgery, Affiliated Zhongshan Hospital, Fudan University, No.180 Fenglin Road, Xuhui District, Shanghai, PR China
| | - Yicun Wang
- Department of General Surgery, Qingpu Branch, Affiliated Zhongshan Hospital of Fudan University, Qingpu Branch, No. 1158 Park Road East, Qingpu District, Shanghai, PR China.
| | - Yaolin Xu
- Department of Pancreatic Surgery, Affiliated Zhongshan Hospital, Fudan University, No.180 Fenglin Road, Xuhui District, Shanghai, PR China.
| | - Liang Liu
- Department of Pancreatic Surgery, Affiliated Zhongshan Hospital, Fudan University, No.180 Fenglin Road, Xuhui District, Shanghai, PR China.
- Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, PR China.
| |
Collapse
|
43
|
Ziętara KJ, Wróblewska K, Zajączkowska M, Taczała J, Lejman M. The Role of the JAK-STAT Pathway in Childhood B-Cell Acute Lymphoblastic Leukemia. Int J Mol Sci 2024; 25:6844. [PMID: 38999955 PMCID: PMC11241568 DOI: 10.3390/ijms25136844] [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: 05/14/2024] [Revised: 06/16/2024] [Accepted: 06/17/2024] [Indexed: 07/14/2024] Open
Abstract
B-cell lymphoblastic leukemia is a hematologic neoplasm that poses a serious health concern in childhood. Genetic aberrations, such as mutations in the genes IL-7, IL7R, JAK1, JAK2, TLSP, CRLF2, and KTM2A or gene fusions involving BCR::ABL1, ETV6::RUNX1, and PAX5::JAK2, often correlate with the onset of this disease. These aberrations can lead to malfunction of the JAK-STAT signaling pathway, which is implicated in various important biological processes, including those related to immunology. Understanding the mechanisms underlying the malfunction of the JAK-STAT pathway holds potential for research on drugs targeting its components. Available drugs that interfere with the JAK-STAT pathway include fludarabine, ruxolitinib, and fedratinib.
Collapse
Affiliation(s)
- Karolina Joanna Ziętara
- Student Scientific Society, Independent Laboratory of Genetic Diagnostics, Medical University of Lublin, 20-093 Lublin, Poland; (K.J.Z.); (K.W.); (M.Z.)
| | - Kinga Wróblewska
- Student Scientific Society, Independent Laboratory of Genetic Diagnostics, Medical University of Lublin, 20-093 Lublin, Poland; (K.J.Z.); (K.W.); (M.Z.)
| | - Monika Zajączkowska
- Student Scientific Society, Independent Laboratory of Genetic Diagnostics, Medical University of Lublin, 20-093 Lublin, Poland; (K.J.Z.); (K.W.); (M.Z.)
| | - Joanna Taczała
- Faculty of Medicine, Medical University of Warsaw, 02-091 Warszawa, Poland;
| | - Monika Lejman
- Independent Laboratory of Genetic Diagnostics, Medical University of Lublin, 20-059 Lublin, Poland
| |
Collapse
|
44
|
Meng J, Yang W, Li C, Li F. Synergistic anticancer effects of SMYD2 inhibitor BAY-598 and doxorubicin in non-small cell lung cancer. Heliyon 2024; 10:e32015. [PMID: 38947456 PMCID: PMC11214464 DOI: 10.1016/j.heliyon.2024.e32015] [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: 01/29/2024] [Revised: 05/26/2024] [Accepted: 05/27/2024] [Indexed: 07/02/2024] Open
Abstract
Background Non-small cell lung Cancer (NSCLC) persists as a lethal neoplastic manifestation, exhibiting a diminished 5-year survival rate, partially attributable to chemotherapeutic resistance. This investigative endeavor aimed to elucidate the synergistic antineoplastic effects and underlying mechanisms of the SMYD2 inhibitor BAY-598 and the chemotherapeutic agent doxorubicin (DOX) in NSCLC. Methods The human non-small cell lung cancer cell lines A549 and H460 were subjected to treatment regimens involving BAY-598 and/or DOX. Cellular viability, apoptotic events, invasive capacity, and migratory potential were evaluated through the implementation of CCK-8 assays, flow cytometric analyses, and Transwell assays, respectively. Protein expression levels were quantified via Western blot analyses. An in vivo xenograft murine model was established to assess therapeutic efficacy. Results BAY-598 and DOX synergistically suppressed the viability, invasiveness, and migratory capabilities of NSCLC cells. Co-treatment Promoting cell apoptosis and cell cycle arrest. Additionally, Furthermore, co-administration significantly inhibited cell migration and invasion. Mechanistic studies revealed coordinately inhibited JAK-STAT signaling upon combination treatment. In vivo study further validated the synergistic antitumor efficacy of BAY-598 and DOX against NSCLC xenografts. Conclusions Our findings demonstrate that BAY-598 potentiates the anti-cancer effects of DOX in non-small cell lung cancer cells by modulating the JAK/STAT signaling pathway as a synergistic strategy. The combination holds promise as an emerging therapeutic strategy for NSCLC. Further optimization and validation are warranted to promote its translational potential.
Collapse
Affiliation(s)
- Jiaqi Meng
- Department of Pulmonary and Critical Care Medicine, The First Hospital of Nanchang, The Third Affiliated Hospital of Nanchang University, Nanchang, 330008, Jiangxi, China
- Medical Department of Graduate School, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Weichang Yang
- Medical Department of Graduate School, Nanchang University, Nanchang, 330006, Jiangxi, China
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Can Li
- Department of Pulmonary and Critical Care Medicine, The First Hospital of Nanchang, The Third Affiliated Hospital of Nanchang University, Nanchang, 330008, Jiangxi, China
| | - Fengyuan Li
- Department of Pulmonary and Critical Care Medicine, The First Hospital of Nanchang, The Third Affiliated Hospital of Nanchang University, Nanchang, 330008, Jiangxi, China
- Medical Department of Graduate School, Nanchang University, Nanchang, 330006, Jiangxi, China
| |
Collapse
|
45
|
Liang F, Liang X, Pan L, Jin Q, Deng J, Hong M, Wei W, Hao Z, Ren H, Wang H, Chen X. Immunophenotype of myeloid granulocytes in Chinese patients with BCR::ABL1-negative myeloproliferative neoplasms. Clin Exp Med 2024; 24:106. [PMID: 38771542 PMCID: PMC11108956 DOI: 10.1007/s10238-024-01363-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 04/28/2024] [Indexed: 05/22/2024]
Abstract
Typical BCR::ABL1-negative myeloproliferative neoplasms (MPN) are mainly referred to as polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofbrosis (PMF). Granulocytes in MPN patients are involved in their inflammation and form an important part of the pathophysiology of MPN patients. It has been shown that the immunophenotype of granulocytes in MPN patients is altered. We used flow cytometry to explore the immunophenotype of MPN patients and correlate it with clinical parameters. The results showed that PMF patients and PV patients had higher CD15+CD11b+ granulocytes than ET patients and normal controls. When grouped by gene mutation, changes in the granulocyte immunophenotype of MPN patients were independent of the JAK2V617F and CALR mutations. There was no significant heterogeneity in immunophenotype between ET patients and Pre-PMF, and between Overt-PMF and Pre-PMF patients. Granulocytes from some MPN patients showed an abnormal CD13/CD16 phenotype with a significant increase in mature granulocytes on molecular and cytomorphological grounds, and this abnormal pattern occurred significantly more frequently in PMF patients than in ET patients. CD15-CD11b- was negatively correlated with WBC and Hb and positively correlated with DIPSS score, whereas high CD10+ granulocytes were significantly and negatively associated with prognostic system IPSS and DIPSS scores in PMF patients. In conclusion, this study demonstrates the landscape of bone marrow granulocyte immunophenotypes in MPN patients. MPN patients, especially those with PMF, have a significant granulocyte developmental overmaturation phenotype. CD10+ granulocytes may be involved in the prognosis of PMF patients.
Collapse
Affiliation(s)
- Fengting Liang
- Key Laboratory of Molecular Diagnosis and Treatment of Hematologic Diseases of Shanxi Province, Taiyuan, China
| | - Xuelan Liang
- Key Laboratory of Molecular Diagnosis and Treatment of Hematologic Diseases of Shanxi Province, Taiyuan, China
| | | | - Qianni Jin
- Key Laboratory of Molecular Diagnosis and Treatment of Hematologic Diseases of Shanxi Province, Taiyuan, China
| | - Ju Deng
- Key Laboratory of Molecular Diagnosis and Treatment of Hematologic Diseases of Shanxi Province, Taiyuan, China
| | - Minglin Hong
- Key Laboratory of Molecular Diagnosis and Treatment of Hematologic Diseases of Shanxi Province, Taiyuan, China
| | - Wei Wei
- Key Laboratory of Molecular Diagnosis and Treatment of Hematologic Diseases of Shanxi Province, Taiyuan, China
| | - Zhuanghui Hao
- Key Laboratory of Molecular Diagnosis and Treatment of Hematologic Diseases of Shanxi Province, Taiyuan, China
| | - Huanying Ren
- Key Laboratory of Molecular Diagnosis and Treatment of Hematologic Diseases of Shanxi Province, Taiyuan, China
| | - Hongwei Wang
- Key Laboratory of Molecular Diagnosis and Treatment of Hematologic Diseases of Shanxi Province, Taiyuan, China.
| | - Xiuhua Chen
- Key Laboratory of Molecular Diagnosis and Treatment of Hematologic Diseases of Shanxi Province, Taiyuan, China.
| |
Collapse
|
46
|
Zhu L, Litts B, Wang Y, Rein JA, Atzrodt CL, Chinnarasu S, An J, Thorson AS, Xu Y, Stafford JM. Ablation of IFNγ in myeloid cells suppresses liver inflammation and fibrogenesis in mice with hepatic small heterodimer partner (SHP) deletion. Mol Metab 2024; 83:101932. [PMID: 38589002 PMCID: PMC11035112 DOI: 10.1016/j.molmet.2024.101932] [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: 01/30/2024] [Revised: 03/21/2024] [Accepted: 03/29/2024] [Indexed: 04/10/2024] Open
Abstract
BACKGROUND Metabolic dysfunction-associated steatotic liver disease (MASLD) is a common complication of obesity and, in severe cases, progresses to metabolic dysfunction-associated steatohepatitis (MASH). Small heterodimer partner (SHP) is an orphan member of the nuclear receptor superfamily and regulates metabolism and inflammation in the liver via a variety of pathways. In this study, we investigate the molecular foundation of MASH progression in mice with hepatic SHP deletion and explore possible therapeutic means to reduce MASH. METHODS Hepatic SHP knockout mice (SHPΔhep) and their wild-type littermates (SHPfl/fl) of both sexes were fed a fructose diet for 14 weeks and subjected to an oral glucose tolerance test. Then, plasma lipids were determined, and liver lipid metabolism and inflammation pathways were analyzed with immunoblotting, RNAseq, and qPCR assays. To explore possible therapeutic intersections of SHP and inflammatory pathways, SHPΔhep mice were reconstituted with bone marrow lacking interferon γ (IFNγ-/-) to suppress inflammation. RESULTS Hepatic deletion of SHP in mice fed a fructose diet decreased liver fat and increased proteins for fatty acid oxidation and liver lipid uptake, including UCP1, CPT1α, ACDAM, and SRBI. Despite lower liver fat, hepatic SHP deletion increased liver inflammatory F4/80+ cells and mRNA levels of inflammatory cytokines (IL-12, IL-6, Ccl2, and IFNγ) in both sexes and elevated endoplasmic reticulum stress markers of Cox2 and CHOP in female mice. Liver bulk RNAseq data showed upregulation of genes whose protein products regulate lipid transport, fatty acid oxidation, and inflammation in SHPΔhep mice. The increased inflammation and fibrosis in SHPΔhep mice were corrected with bone marrow-derived IFNγ-/- myeloid cell transplantation. CONCLUSION Hepatic deletion of SHP improves fatty liver but worsens hepatic inflammation possibly by driving excess fatty acid oxidation, which is corrected by deletion of IFNγ specifically in myeloid cells. This suggests that hepatic SHP limits fatty acid oxidation during fructose diet feeding but, in doing so, prevents pro-MASH pathways. The IFNγ-mediated inflammation in myeloid cells appears to be a potential therapeutic target to suppress MASH.
Collapse
Affiliation(s)
- Lin Zhu
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, USA
| | - Bridget Litts
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, USA
| | - Yu Wang
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jeffrey A Rein
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, USA
| | | | | | - Julia An
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, USA
| | - Ariel S Thorson
- Department of Molecular Physiology & Biophysics, Vanderbilt University, USA
| | - Yaomin Xu
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - John M Stafford
- Tennessee Valley Health System, Veterans Affairs, Nashville, TN, USA; Department of Molecular Physiology & Biophysics, Vanderbilt University, USA; Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, USA.
| |
Collapse
|
47
|
Shin J, Miaskowski C, Wong ML, Yates P, Olshen AB, Roy R, Dokiparthi V, Cooper B, Paul S, Conley YP, Levine JD, Hammer MJ, Kober K. Perturbations in inflammatory pathways are associated with shortness of breath profiles in oncology patients receiving chemotherapy. Support Care Cancer 2024; 32:250. [PMID: 38532105 PMCID: PMC11484916 DOI: 10.1007/s00520-024-08446-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Accepted: 03/18/2024] [Indexed: 03/28/2024]
Abstract
PURPOSE One plausible mechanistic hypothesis is the potential contribution of inflammatory mechanisms to shortness of breath. This study was aimed to evaluate for associations between the occurrence of shortness of breath and perturbations in inflammatory pathways. METHODS Patients with cancer reported the occurrence of shortness of breath six times over two cycles of chemotherapy. Latent class analysis was used to identify subgroups of patients with distinct shortness of breath occurrence profiles (i.e., none (70.5%), decreasing (8.2%), increasing (7.8%), high (13.5%)). Using an extreme phenotype approach, whole transcriptome differential gene expression and pathway impact analyses were performed to evaluate for perturbed signaling pathways associated with shortness of breath between the none and high classes. Two independent samples (RNA-sequencing (n = 293) and microarray (n = 295) methodologies) were evaluated. Fisher's combined probability method was used to combine these results to obtain a global test of the null hypothesis. In addition, an unweighted knowledge network was created using the specific pathway maps to evaluate for interconnections among these pathways. RESULTS Twenty-nine Kyoto Encyclopedia of Genes and Genomes inflammatory signaling pathways were perturbed. The mitogen-activated protein kinase signaling pathway node had the highest closeness, betweenness, and degree scores. In addition, five common respiratory disease-related pathways, that may share mechanisms with cancer-related shortness of breath, were perturbed. CONCLUSIONS Findings provide preliminary support for the hypothesis that inflammation contribute to the occurrence of shortness of breath in patients with cancer. In addition, the mechanisms that underlie shortness of breath in oncology patients may be similar to other respiratory diseases.
Collapse
Affiliation(s)
- Joosun Shin
- School of Nursing, University of California, 2 Koret Way - N631Y, San Francisco, CA, 94143-0610, USA
- Dana-Farber Cancer Institute, Boston, MA, USA
| | - Christine Miaskowski
- School of Nursing, University of California, 2 Koret Way - N631Y, San Francisco, CA, 94143-0610, USA
- School of Medicine, University of California, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Melisa L Wong
- School of Medicine, University of California, San Francisco, CA, USA
| | - Patsy Yates
- Faculty of Health, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Adam B Olshen
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Ritu Roy
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Vasuda Dokiparthi
- School of Nursing, University of California, 2 Koret Way - N631Y, San Francisco, CA, 94143-0610, USA
| | - Bruce Cooper
- School of Nursing, University of California, 2 Koret Way - N631Y, San Francisco, CA, 94143-0610, USA
| | - Steven Paul
- School of Nursing, University of California, 2 Koret Way - N631Y, San Francisco, CA, 94143-0610, USA
| | - Yvette P Conley
- School of Nursing, Univeristy of Pittsburgh, 3500 Victoria St, Pittsburgh, 15213, PA, USA
| | - Jon D Levine
- School of Medicine, University of California, San Francisco, CA, USA
| | | | - Kord Kober
- School of Nursing, University of California, 2 Koret Way - N631Y, San Francisco, CA, 94143-0610, USA.
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA.
| |
Collapse
|
48
|
Wang JN, Li Y. Exploring the molecular mechanisms between lymphoma and myelofibrosis. Am J Transl Res 2024; 16:730-737. [PMID: 38586105 PMCID: PMC10994807 DOI: 10.62347/nwjo7078] [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: 10/30/2023] [Accepted: 02/28/2024] [Indexed: 04/09/2024]
Abstract
Lymphoma is a heterogeneous malignant tumor with an increasing annual incidence. As the lymphoma progresses, bone marrow (BM) invasion gradually appears. Myelofibrosis (MF) can accompany a variety of hematological malignancies, including lymphoma, and multiple myeloma. The prognosis of lymphoma patients with myelofibrosis is poor, and a fundamental reason is that there are few studies on the correlation and pathogenesis of the two diseases. In this review, we examine the potential pathogenesis and the correlation of the two diseases.
Collapse
Affiliation(s)
- Jun-Nuan Wang
- Hebei Medical UniversityShijiazhuang, Hebei, The People’s Republic of China
- Department of Hematology, Hebei General HospitalShijiazhuang, Hebei, The People’s Republic of China
| | - Yan Li
- Department of Hematology, Hebei General HospitalShijiazhuang, Hebei, The People’s Republic of China
| |
Collapse
|
49
|
Gawish RA, Samy EM, Aziz MM. Ferulic acid protects against gamma-radiation induced liver injury via regulating JAK/STAT/Nrf2 pathways. Arch Biochem Biophys 2024; 753:109895. [PMID: 38244663 DOI: 10.1016/j.abb.2024.109895] [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: 11/25/2023] [Revised: 01/15/2024] [Accepted: 01/17/2024] [Indexed: 01/22/2024]
Abstract
This study aims to evaluate the effect and underlying mechanism of ferulic acid (FA) in alleviating the acute liver injury by ionizing radiation (IR) in vivo. Rats were divided into 4groups (Groups: control, 6Gy irradiated (IRR), FA (50 mg/kg) and FA + IRR). The results showed that FA can effectively inhibit liver damage and restore the structure and function of the liver. In mechanism, FA prevented IR-induced liver fibrosis and blocked the JAK/STAT signaling pathway to effectively inhibit the hepatic inflammatory response; and inhibited IR-induced oxidative stress (OS) by upregulating the Nrf2 signaling pathway and promoting the synthesis of several antioxidants. Moreover, FA inhibited ferroptosis in the liver by stimulating the expression of GPX4 and SLC7A11. FA reduced lipid peroxidation by downregulation of the reactive oxygen species (ROS) production and iron aggregation, thus inhibiting ferroptosis and alleviating IR-induced liver injury. In conclusion, the current study suggests the potential complex mechanisms underlying the mitigating impact of FA in IR-induced ferroptotic liver damage.
Collapse
Affiliation(s)
- Rania A Gawish
- Drug Radiation Research Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority, Cairo, Egypt.
| | - Esraa M Samy
- Drug Radiation Research Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority, Cairo, Egypt.
| | - Maha M Aziz
- Drug Radiation Research Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority, Cairo, Egypt.
| |
Collapse
|
50
|
Jiang H, Yang J, Li T, Wang X, Fan Z, Ye Q, Du Y. JAK/STAT3 signaling in cardiac fibrosis: a promising therapeutic target. Front Pharmacol 2024; 15:1336102. [PMID: 38495094 PMCID: PMC10940489 DOI: 10.3389/fphar.2024.1336102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 01/18/2024] [Indexed: 03/19/2024] Open
Abstract
Cardiac fibrosis is a serious health problem because it is a common pathological change in almost all forms of cardiovascular diseases. Cardiac fibrosis is characterized by the transdifferentiation of cardiac fibroblasts (CFs) into cardiac myofibroblasts and the excessive deposition of extracellular matrix (ECM) components produced by activated myofibroblasts, which leads to fibrotic scar formation and subsequent cardiac dysfunction. However, there are currently few effective therapeutic strategies protecting against fibrogenesis. This lack is largely because the molecular mechanisms of cardiac fibrosis remain unclear despite extensive research. The Janus kinase/signal transducer and activator of transcription (JAK/STAT) signaling cascade is an extensively present intracellular signal transduction pathway and can regulate a wide range of biological processes, including cell proliferation, migration, differentiation, apoptosis, and immune response. Various upstream mediators such as cytokines, growth factors and hormones can initiate signal transmission via this pathway and play corresponding regulatory roles. STAT3 is a crucial player of the JAK/STAT pathway and its activation is related to inflammation, malignant tumors and autoimmune illnesses. Recently, the JAK/STAT3 signaling has been in the spotlight for its role in the occurrence and development of cardiac fibrosis and its activation can promote the proliferation and activation of CFs and the production of ECM proteins, thus leading to cardiac fibrosis. In this manuscript, we discuss the structure, transactivation and regulation of the JAK/STAT3 signaling pathway and review recent progress on the role of this pathway in cardiac fibrosis. Moreover, we summarize the current challenges and opportunities of targeting the JAK/STAT3 signaling for the treatment of fibrosis. In summary, the information presented in this article is critical for comprehending the role of the JAK/STAT3 pathway in cardiac fibrosis, and will also contribute to future research aimed at the development of effective anti-fibrotic therapeutic strategies targeting the JAK/STAT3 signaling.
Collapse
Affiliation(s)
- Heng Jiang
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Junjie Yang
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Tao Li
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Xinyu Wang
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Zhongcai Fan
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Qiang Ye
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Yanfei Du
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| |
Collapse
|