1
|
Song J, Luo X, Xiong L, Li S, Chen Y, Liu W, Yuan Y, Ma Y, Bian J, Liu Z, Zou H. Lycium barbarum polysaccharide alleviate cadmium-induced mitochondrial dysfunction mediated pyroptosis in duck liver. Int J Biol Macromol 2025; 311:143989. [PMID: 40339840 DOI: 10.1016/j.ijbiomac.2025.143989] [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: 01/23/2025] [Revised: 04/19/2025] [Accepted: 05/05/2025] [Indexed: 05/10/2025]
Abstract
Cadmium (Cd) is a persistent environmental heavy metal pollutant that has been an important issue in toxicology research worldwide. This toxic element is non-degradable and accumulates indefinitely in the ecosystem, and gradually accumulates in living organisms through nutrient transfer, ultimately posing a significant risk to the health of animals. The hepatic system is both the main target organ for Cd bioaccumulation and the metabolic center of organisms, and is particularly susceptible to heavy metal exposure. However, the underlying mechanism of whether LBP protects against Cd-induced liver injury in male Shaoxing ducks is still unknown. In our study, male Shaoxing ducks were treated with Cdcl2 (50 mg/kg) and/or LBP (50 mg/kg) for 30 days. We found that Cd exposure reduced the growth rate, feed intake, liver weight and organ index, and increased the serum biochemical indexes (AST, ALT, ALP) in Shaoxing ducks. Further studies have shown that Cd induces liver pathological damage and macro/trace element metabolism disorders, thereby inhibiting the activity of Nrf2 antioxidant signaling pathway and further inducing oxidative stress. Finally, we found that Cd induces the destruction of liver mitochondrial structure, disrupts the normal mitochondrial biogenesis and fusion/fission disorder, and thus causes liver pyroptosis and fibrosis. In this process, we found that LBP administration significantly improved Cd-induced growth inhibition and liver damage.
Collapse
Affiliation(s)
- Jie Song
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, People's Republic of China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu 225009, People's Republic of China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu 225009, People's Republic of China
| | - Xianzu Luo
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, People's Republic of China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu 225009, People's Republic of China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu 225009, People's Republic of China
| | - Ling Xiong
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, People's Republic of China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu 225009, People's Republic of China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu 225009, People's Republic of China
| | - Sifan Li
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, People's Republic of China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu 225009, People's Republic of China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu 225009, People's Republic of China
| | - Yan Chen
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, People's Republic of China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu 225009, People's Republic of China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu 225009, People's Republic of China
| | - Wei Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, People's Republic of China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu 225009, People's Republic of China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu 225009, People's Republic of China
| | - Yan Yuan
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, People's Republic of China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu 225009, People's Republic of China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu 225009, People's Republic of China
| | - Yonggang Ma
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, People's Republic of China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu 225009, People's Republic of China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu 225009, People's Republic of China
| | - Jianchun Bian
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, People's Republic of China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu 225009, People's Republic of China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu 225009, People's Republic of China
| | - Zongping Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, People's Republic of China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu 225009, People's Republic of China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu 225009, People's Republic of China
| | - Hui Zou
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, People's Republic of China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu 225009, People's Republic of China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu 225009, People's Republic of China
| |
Collapse
|
2
|
El-Rayes B, Reddy D, Sarvesh S, Foote J, Welsch D, Cheng C, Akce M, Nagaraju GP. Paricalcitol and hydroxychloroquine modulates extracellular matrix and enhance chemotherapy efficacy in pancreatic cancer. RESEARCH SQUARE 2025:rs.3.rs-6406693. [PMID: 40386428 PMCID: PMC12083674 DOI: 10.21203/rs.3.rs-6406693/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/26/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive cancer with poor prognosis and limited therapeutic options. In a previous publication, our group defined some of the mechanisms that vitamin D analogue paricalcitol (P) and hydroxychloroquine (H) potentiated the effects of gemcitabine-based chemotherapy in PDAC. Based on this, we hypothesized that PH may potentiate 5-fluorouracil (5FU) and oxaliplatin-based chemotherapy, and this may involve a novel mechanism of extracellular matrix (ECM) modulation. The combination of PH with 5FU/oxaliplatin significantly increased the cell death, apoptosis, and S-phase cell cycle arrest as compared to untreated or 5FU/oxaliplatin treated MIA PaCa-2, HPAC and KPC cell lines. In vivo, the combination therapy inhibited PDAC growth and altered the immune landscape by activating T and NK cells. Proteomic analysis revealed significant reduction in ECM proteins, specifically integrin beta-4 (ITGB4). Confirmation of the role of ITGB4 was performed through genetic knockdown of ITGB4 which led ECM inhibition. In conclusion, the combination of PH significantly enhances the efficacy of oxaliplatin and 5FU. We identified a new mechanism of action of PH through inhibiting ITGB4 leading to ECM modulation. These results suggest that the combination of PH with cytotoxic chemotherapy should be tested in PDAC clinical trials.
Collapse
|
3
|
Blasiak J, Pawlowska E, Helotera H, Ionov M, Derwich M, Kaarniranta K. Potential of autophagy in subretinal fibrosis in neovascular age-related macular degeneration. Cell Mol Biol Lett 2025; 30:54. [PMID: 40307700 PMCID: PMC12044759 DOI: 10.1186/s11658-025-00732-8] [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: 01/13/2025] [Accepted: 04/11/2025] [Indexed: 05/02/2025] Open
Abstract
Age-related macular degeneration (AMD) is an eye disease that can lead to legal blindness and vision loss. In its advanced stages, it is classified into dry and neovascular AMD. In neovascular AMD, the formation of new blood vessels disrupts the structure of the retina and induces an inflammatory response. Treatment for neovascular AMD involves antibodies and fusion proteins targeting vascular endothelial growth factor A (VEGFA) and its receptors to inhibit neovascularization and slow vision loss. However, a fraction of patients with neovascular AMD do not respond to therapy. Many of these patients exhibit a subretinal fibrotic scar. Thus, retinal fibrosis may contribute to resistance against anti-VEGFA therapy and the cause of irreversible vision loss in neovascular AMD patients. Retinal pigment epithelium cells, choroidal fibroblasts, and retinal glial cells are crucial in the development of the fibrotic scar as they can undergo a mesenchymal transition mediated by transforming growth factor beta and other molecules, leading to their transdifferentiation into myofibroblasts, which are key players in subretinal fibrosis. Autophagy, a process that removes cellular debris and contributes to the pathogenesis of AMD, regardless of its type, may be stimulated by epithelial-mesenchymal transition and later inhibited. The mesenchymal transition of retinal cells and the dysfunction of the extracellular matrix-the two main aspects of fibrotic scar formation-are associated with impaired autophagy. Nonetheless, the causal relationship between autophagy and subretinal fibrosis remains unknown. This narrative/perspective review presents information on neovascular AMD, subretinal fibrosis, and autophagy, arguing that impaired autophagy may be significant for fibrosis-related resistance to anti-VEGFA therapy in neovascular AMD.
Collapse
Affiliation(s)
- Janusz Blasiak
- Faculty of Medicine, Collegium Medicum, Mazovian Academy in Plock, 09-402, Plock, Poland.
| | - Elzbieta Pawlowska
- Department of Pediatric Dentistry, Medical University of Lodz, 92-217, Lodz, Poland
| | - Hanna Helotera
- Department of Ophthalmology, University of Eastern Finland, 70210, Kuopio, Finland
| | - Maksim Ionov
- Faculty of Health Sciences, Mazovian Academy in Plock, 09-402, Plock, Poland
| | - Marcin Derwich
- Department of Pediatric Dentistry, Medical University of Lodz, 92-217, Lodz, Poland
| | - Kai Kaarniranta
- Department of Ophthalmology, University of Eastern Finland, 70210, Kuopio, Finland
- Department of Ophthalmology, Kuopio University Hospital, 70210, Kuopio, Finland
| |
Collapse
|
4
|
Dilger OB, Carstens MF, Bothun CE, Payne AN, Berry DJ, Sanchez-Sotelo J, Morrey ME, Thaler R, Dudakovic A, Abdel MP. Induction of cellular autophagy impairs TGF-β1-mediated extracellular matrix deposition in primary human knee fibroblasts. Bone Joint Res 2025; 14:331-340. [PMID: 40192622 PMCID: PMC11975063 DOI: 10.1302/2046-3758.144.bjr-2024-0312.r1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/09/2025] Open
Abstract
Aims To evaluate the role of autophagy in primary knee fibroblasts undergoing myofibroblast differentiation as an in vitro model of arthrofibrosis, a complication after total knee arthroplasty characterized by aberrant intra-articular scar tissue formation and limited range of motion. Methods We conducted a therapeutic screen of autophagic-modulating therapies in primary human knee fibroblasts undergoing transforming growth factor-beta 1 (TGF-β1)-mediated myofibroblast differentiation. Autophagy was induced pharmacologically with rapamycin or by amino acid deprivation. Picrosirius red staining was performed to quantify collagen deposition. Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and western blotting were conducted to evaluate fibrotic gene expression levels. Results Rapamycin, an mTOR complex 1 (mTORC1) inhibitor and autophagy inducer, reduced TGF-β1-mediated collagen deposition. Interestingly, we simultaneously report that myofibrogenic genes, including ACTA2, were highly upregulated following rapamycin-TGF-β1 treatment. When autophagy was induced through amino acid deprivation, we demonstrated suppressed extracellular matrix levels, fibrotic gene expression (e.g. ACTA2), and SMAD2 phosphorylation levels in TGF-β1-stimulated fibroblasts. Conclusion Our findings demonstrate that the induction of cellular autophagy suppresses TGF-β1-induced collagen deposition in primary human knee fibroblasts. Taken together, these data suggest that cellular autophagy may be prophylactic against the pathogenesis of arthrofibrosis.
Collapse
Affiliation(s)
- Oliver B. Dilger
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Mason F. Carstens
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Cole E. Bothun
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Ashley N. Payne
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Daniel J. Berry
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Mark E. Morrey
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Roman Thaler
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Amel Dudakovic
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Matthew P. Abdel
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
5
|
Son J, Park J, Jeong JW, Lee SH, Kim JE. SIRT2 inhibition attenuates myofibroblast transition through autophagy-mediated ciliogenesis in renal epithelial cells. Int J Biochem Cell Biol 2025; 181:106754. [PMID: 39988243 DOI: 10.1016/j.biocel.2025.106754] [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: 10/05/2024] [Revised: 02/09/2025] [Accepted: 02/16/2025] [Indexed: 02/25/2025]
Abstract
Myofibroblast transition plays a crucial role in both fibrotic diseases and wound healing. Although SIRT2 regulates fibrosis, its mechanisms of action remain poorly understood. This study aimed to investigate the effects of SIRT2 inhibition on myofibroblast transition in human renal cells under quiescent conditions. HK-2 kidney proximal tubular epithelial cells were starved of serum, resulting in the formation of primary cilia. Transforming growth factor-β (TGF-β) stimulation reduced both the number of ciliated cells and ciliary length. The ciliary defects resulted from a failure in autophagy termination, leading to the accumulation of OFD1, a negative regulator of ciliogenesis, at centriolar satellites. This phenomenon was correlated with the upregulation of fibrosis-related proteins. To elucidate the role of SIRT2 in the autophagy-ciliogenesis-fibrosis axis, cells were treated with AGK2, a specific inhibitor of SIRT2. AGK2 treatment promoted the formation of both autophagosomes and autolysosomes and facilitated OFD1 degradation at the centriolar satellites, resulting in the lengthening of primary cilia. Restoration of primary cilia by AGK2 was associated with the suppression of myofibroblast transition. In conclusion, SIRT2 inhibition attenuates TGF-β-induced fibrosis by promoting autophagy-mediated ciliogenesis. This study highlights SIRT2 as a potential therapeutic target for fibrotic diseases.
Collapse
Affiliation(s)
- Juyoung Son
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Jaejung Park
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Joo-Won Jeong
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; Department of Anatomy and Neurobiology, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Seung Hyeun Lee
- Department of Precision Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Ja-Eun Kim
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; Department of Precision Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; Department of Pharmacology, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea.
| |
Collapse
|
6
|
Luo PY, Zou JR, Chen T, Zou J, Li W, Chen Q, Cheng L, Zheng LY, Qian B. Autophagy in erectile dysfunction: focusing on apoptosis and fibrosis. Asian J Androl 2025; 27:166-176. [PMID: 39028624 PMCID: PMC11949458 DOI: 10.4103/aja202433] [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: 01/12/2024] [Accepted: 04/22/2024] [Indexed: 07/21/2024] Open
Abstract
ABSTRACT In most types of erectile dysfunction, particularly in advanced stages, typical pathological features observed are reduced parenchymal cells coupled with increased tissue fibrosis. However, the current treatment methods have shown limited success in reversing these pathologic changes. Recent research has revealed that changes in autophagy levels, along with alterations in apoptosis and fibrosis-related proteins, are linked to the progression of erectile dysfunction, suggesting a significant association. Autophagy, known to significantly affect cell fate and tissue fibrosis, is currently being explored as a potential treatment modality for erectile dysfunction. However, these present studies are still in their nascent stage, and there are limited experimental data available. This review analyzes erectile dysfunction from a pathological perspective. It provides an in-depth overview of how autophagy is involved in the apoptotic processes of smooth muscle and endothelial cells and its role in the fibrotic processes occurring in the cavernosum. This study aimed to develop a theoretical framework for the potential effectiveness of autophagy in preventing and treating erectile dysfunction, thus encouraging further investigation among researchers in this area.
Collapse
Affiliation(s)
- Pei-Yue Luo
- The First Clinical College, Gannan Medical University, Ganzhou 341000, China
- Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
- Key Laboratory of Urology and Andrology of Ganzhou, Ganzhou 341000, China
| | - Jun-Rong Zou
- Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
- Key Laboratory of Urology and Andrology of Ganzhou, Ganzhou 341000, China
| | - Tao Chen
- The First Clinical College, Gannan Medical University, Ganzhou 341000, China
- Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
- Key Laboratory of Urology and Andrology of Ganzhou, Ganzhou 341000, China
| | - Jun Zou
- The First Clinical College, Gannan Medical University, Ganzhou 341000, China
- Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
- Key Laboratory of Urology and Andrology of Ganzhou, Ganzhou 341000, China
| | - Wei Li
- The First Clinical College, Gannan Medical University, Ganzhou 341000, China
- Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
- Key Laboratory of Urology and Andrology of Ganzhou, Ganzhou 341000, China
| | - Qi Chen
- The First Clinical College, Gannan Medical University, Ganzhou 341000, China
- Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
- Key Laboratory of Urology and Andrology of Ganzhou, Ganzhou 341000, China
| | - Le Cheng
- The First Clinical College, Gannan Medical University, Ganzhou 341000, China
- Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
- Key Laboratory of Urology and Andrology of Ganzhou, Ganzhou 341000, China
| | - Li-Ying Zheng
- Department of Graduate, The First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
| | - Biao Qian
- Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
- Key Laboratory of Urology and Andrology of Ganzhou, Ganzhou 341000, China
| |
Collapse
|
7
|
Mencucci R, Cennamo M, Rosa I, Guasti D, Buzzi M, Sgambati E, Marini M, Manetti M. New insights into persistent corneal subepithelial infiltrates following epidemic keratoconjunctivitis: The first case report with ultrastructural and immunohistochemical investigations. Acta Histochem 2025; 127:152231. [PMID: 39874705 DOI: 10.1016/j.acthis.2025.152231] [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: 12/06/2024] [Revised: 01/18/2025] [Accepted: 01/21/2025] [Indexed: 01/30/2025]
Abstract
Epidemic keratoconjunctivitis (EKC) is one of the most severe clinical manifestations of human adenovirus ocular surface infection, which may lead to the formation of subepithelial infiltrates (SEIs) in the anterior corneal stroma in 20-50 % of cases. SEIs may be asymptomatic or give rise to corneal aberrations and visual impairment for months or years after acute infection, despite treatments. Here, we describe the ultrastructural and immunophenotypic features of the anterior corneal stroma of a patient who underwent superficial anterior lamellar keratoplasty (SALK) surgery to remove corneal opacities related to clinically significant and steroid-unresponsive, long-lasting SEIs after adenoviral EKC. Before femtosecond laser-assisted SALK surgical intervention, the patient underwent in vivo confocal microscopy that showed a cluster of hyperreflective inflammatory cells within the basal epithelium, associated to an abnormal sub-basal nerve plexus with a fragmented nervous component appearance. The areas corresponding to the SEIs appeared as roundish hyperreflective spots with undefined borders. Transmission electron microscopy analysis of the excised anterior corneal button revealed the presence of giant stromal cells displaying myofibroblast-like features immediately beneath the Bowman's layer. Such abnormal cells exhibited ultrastructural signs of endoplasmic reticulum stress and autophagy, and were positive for markers of activated fibroblasts/myofibroblasts at immunofluorescence analysis. The deeper stroma was instead populated by normal stromal cells (i.e., keratocytes). This case report provides the first morphological evidence that persistent SEIs could be the macroscopic expression of subepithelial giant stromal cells with myofibroblast-like characteristics. Such a novel observation might pave the way toward a better targeted therapeutic management of SEIs.
Collapse
Affiliation(s)
- Rita Mencucci
- Eye Clinic, Department of Neurosciences, Psychology, Pharmacology and Child Health, University of Florence, Largo Brambilla 3, Florence 50134, Italy
| | - Michela Cennamo
- Eye Clinic, Department of Neurosciences, Psychology, Pharmacology and Child Health, University of Florence, Largo Brambilla 3, Florence 50134, Italy
| | - Irene Rosa
- Section of Anatomy and Histology, Imaging Platform, Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla 3, Florence 50134, Italy
| | - Daniele Guasti
- Section of Anatomy and Histology, Imaging Platform, Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla 3, Florence 50134, Italy
| | - Matilde Buzzi
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, Milan 20072, Italy
| | - Eleonora Sgambati
- Department of Biosciences and Territory, University of Molise, Contrada Fonte Lappone, Pesche, Isernia 86090, Italy
| | - Mirca Marini
- Section of Anatomy and Histology, Imaging Platform, Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla 3, Florence 50134, Italy
| | - Mirko Manetti
- Section of Anatomy and Histology, Imaging Platform, Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla 3, Florence 50134, Italy.
| |
Collapse
|
8
|
Tang J, Liu J, Zhou Z, Cui X, Tu H, Jia J, Chen B, Dai X, Liu O. Oral submucous fibrosis: pathogenesis and therapeutic approaches. Int J Oral Sci 2025; 17:8. [PMID: 39890798 PMCID: PMC11785813 DOI: 10.1038/s41368-024-00344-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 11/14/2024] [Accepted: 12/24/2024] [Indexed: 02/03/2025] Open
Abstract
Oral submucous fibrosis (OSF), characterized by excessive deposition of extracellular matrix (ECM) that causes oral mucosal tissue sclerosis, and even cancer transformation, is a chronic, progressive fibrosis disease. However, despite some advancements in recent years, no targeted antifibrotic strategies for OSF have been approved; likely because the complicated mechanisms that initiate and drive fibrosis remain to be determined. In this review, we briefly introduce the epidemiology and etiology of OSF. Then, we highlight how cell-intrinsic changes in significant structural cells can drive fibrotic response by regulating biological behaviors, secretion function, and activation of ECM-producing myofibroblasts. In addition, we also discuss the role of innate and adaptive immune cells and how they contribute to the pathogenesis of OSF. Finally, we summarize strategies to interrupt key mechanisms that cause OSF, including modulation of the ECM, inhibition of inflammation, improvement of vascular disturbance. This review will provide potential routes for developing novel anti-OSF therapeutics.
Collapse
Affiliation(s)
- Jianfei Tang
- Hunan Key Laboratory of Oral Health Research & Hunan 3D Printing Engineering Research Center of Oral Care & Hunan Clinical Research Center of Oral Major Diseases and Oral Health & Academician Workstation for Oral-Maxilofacial and Regenerative Medicine & Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, China
| | - Junjie Liu
- Hunan Key Laboratory of Oral Health Research & Hunan 3D Printing Engineering Research Center of Oral Care & Hunan Clinical Research Center of Oral Major Diseases and Oral Health & Academician Workstation for Oral-Maxilofacial and Regenerative Medicine & Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, China
| | - Zekun Zhou
- Hunan Key Laboratory of Oral Health Research & Hunan 3D Printing Engineering Research Center of Oral Care & Hunan Clinical Research Center of Oral Major Diseases and Oral Health & Academician Workstation for Oral-Maxilofacial and Regenerative Medicine & Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, China
| | - Xinyan Cui
- Hunan Key Laboratory of Oral Health Research & Hunan 3D Printing Engineering Research Center of Oral Care & Hunan Clinical Research Center of Oral Major Diseases and Oral Health & Academician Workstation for Oral-Maxilofacial and Regenerative Medicine & Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, China
| | - Hua Tu
- Hunan Key Laboratory of Oral Health Research & Hunan 3D Printing Engineering Research Center of Oral Care & Hunan Clinical Research Center of Oral Major Diseases and Oral Health & Academician Workstation for Oral-Maxilofacial and Regenerative Medicine & Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, China
| | - Jia Jia
- Hunan Key Laboratory of Oral Health Research & Hunan 3D Printing Engineering Research Center of Oral Care & Hunan Clinical Research Center of Oral Major Diseases and Oral Health & Academician Workstation for Oral-Maxilofacial and Regenerative Medicine & Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, China
| | - Baike Chen
- Hunan Key Laboratory of Oral Health Research & Hunan 3D Printing Engineering Research Center of Oral Care & Hunan Clinical Research Center of Oral Major Diseases and Oral Health & Academician Workstation for Oral-Maxilofacial and Regenerative Medicine & Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, China
| | - Xiaohan Dai
- Hunan Key Laboratory of Oral Health Research & Hunan 3D Printing Engineering Research Center of Oral Care & Hunan Clinical Research Center of Oral Major Diseases and Oral Health & Academician Workstation for Oral-Maxilofacial and Regenerative Medicine & Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, China.
| | - Ousheng Liu
- Hunan Key Laboratory of Oral Health Research & Hunan 3D Printing Engineering Research Center of Oral Care & Hunan Clinical Research Center of Oral Major Diseases and Oral Health & Academician Workstation for Oral-Maxilofacial and Regenerative Medicine & Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, China.
| |
Collapse
|
9
|
Zeng Y, Feng Q. ULK1 methylation promotes TGF-β1-induced endometrial fibrosis via the FOXP1/DNMT1 axis. Kaohsiung J Med Sci 2025; 41:e12915. [PMID: 39629895 PMCID: PMC11724166 DOI: 10.1002/kjm2.12915] [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: 07/16/2024] [Revised: 11/12/2024] [Accepted: 11/14/2024] [Indexed: 01/30/2025] Open
Abstract
Intrauterine adhesion (IUA) is the second most common cause of secondary infertility in women and can also lead to menstrual abnormalities and multiple adverse pregnancy outcomes. Therefore, elucidating the mechanism of its development is crucial for the prevention and treatment of IUA. This study will investigate the function and mechanism of forkhead box P1 (FOXP1)/DNA methyltransferase 1 (DNMT1)/unc-51-like autophagy activating kinase 1 (ULK1) in IUA. Expression levels of key genes were detected using western blot and quantitative - real time reverse transcription polymerase chain reaction. Cell proliferation was detected by CCK-8 and EdU staining. Transcriptional regulation relationships were detected by dual luciferase reporter gene and chromatin immunoprecipitation (ChIP) assay. Methylation station of ULK1 was detected by methylmion specific PCR (MSP). Fibrosis and pathological changes in the uterine cavity tissues were detected by Masson and hematoxylin and eosin staining. It was observed that the expression of FOXP1 and DNMT1 increased in transforming growth factor (TGF)-β1-induced cells, while ULK1 expression decreased. Downregulation of FOXP1 could inhibit human endometrial stromal cells proliferation and autophagy, as well as decrease the expression of fibrogenic factors (collagen type I alpha 1 chain [COL1A1], fibronectin [FN], and alpha-smooth muscle actin [α-SMA]). The results of MSP and ChIP experiments showed that DNMT1 promotes methylation of the ULK1 promoter region and inhibits its transcription. In an animal model, knockdown of FOXP1 alleviated pathological fibrosis and uterine adhesions. Knockdown of FOXP1 can inhibit endometrial fibrosis in IUA rats; FOXP1 could be a potential target for the treatment of IUA.
Collapse
Affiliation(s)
- Yuhua Zeng
- Health Management Medicine CenterThe Third Xiangya Hospital of Central South UniversityChangshaChina
| | - Qing Feng
- Department of Obstetrics and GynecologyThe Third Xiangya Hospital of Central South UniversityChangshaChina
| |
Collapse
|
10
|
Xu J, Wang Y, Shao Z, Zhou Y, Bin X, Liu L, Huang W, Wang X, Hu Y, Li K. Adipose-derived stem cell exosomes attenuates myofibroblast transformation via inhibiting autophagy through TGF-β/Smad2 axis in oral submucosal fibrosis. J Nanobiotechnology 2024; 22:780. [PMID: 39702233 DOI: 10.1186/s12951-024-03067-2] [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: 10/13/2024] [Accepted: 12/09/2024] [Indexed: 12/21/2024] Open
Abstract
Oral submucous fibrosis (OSF) is a precancerous condition that poses substantial health risks. OSF is mainly caused by betel nut chewing behavior, but its pathogenesis is still unclear and there is no effective treatment strategy. The transformation of fibroblasts to myofibroblast is the key pathological change in the development of OSF. We isolated fibroblasts from human oral mucosa and induced them into myofibroblasts by arecoline, during which autophagy was significantly activated. Here, we found that adipose-derived stem cell exosomes (ADSCs-EXO) could inhibit autophagy to regulate myofibroblast phenotype, and transcriptome sequencing analysis suggested that this process is closely related to the TGF-β pathway. The interplay between autophagy and TGF-β pathway was examined through modulation the two with autophagy activators and inhibitors, TGF-β receptor activators and inhibitors. Results showed that in vitro, the TGF-β/Smad2 pathway augmented autophagy and promoted myofibroblast transformation. The transcriptome information of ADSCs-EXO showed that it contains a large number of miRNAs. Among them, miR-125a-5p could target Smad2. In vivo, injection of ADSCs-EXO alleviated OSF in mice, during which TGF-β and autophagy signals were inhibited. We suggested that ADSCs-EXO could inhibit myofibroblast transformation via inhibiting autophagy through TGF-β/Smad2 axis in OSF, providing new insights for autophagy-based intervention strategies.
Collapse
Affiliation(s)
- Jinhao Xu
- Department of Oral & Maxillofacial Surgery, Xiangya Stomatological Hospital and Xiangya School of Stomatology, Central South University, Changsha, 410000, China
- Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Xiangya Stomatological Hospital and Xiangya School of Stomatology, Central South University, Changsha, Hunan, 410008, China
| | - Yujing Wang
- Department of Oral & Maxillofacial Surgery, Xiangya Stomatological Hospital and Xiangya School of Stomatology, Central South University, Changsha, 410000, China
- Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Xiangya Stomatological Hospital and Xiangya School of Stomatology, Central South University, Changsha, Hunan, 410008, China
| | - Zifei Shao
- Department of Oral & Maxillofacial Surgery, Xiangya Stomatological Hospital and Xiangya School of Stomatology, Central South University, Changsha, 410000, China
- Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Xiangya Stomatological Hospital and Xiangya School of Stomatology, Central South University, Changsha, Hunan, 410008, China
| | - Yuxi Zhou
- Department of Oral & Maxillofacial Surgery, Xiangya Stomatological Hospital and Xiangya School of Stomatology, Central South University, Changsha, 410000, China
- Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Xiangya Stomatological Hospital and Xiangya School of Stomatology, Central South University, Changsha, Hunan, 410008, China
| | - Xin Bin
- Department of Oral & Maxillofacial Surgery, Xiangya Stomatological Hospital and Xiangya School of Stomatology, Central South University, Changsha, 410000, China
- Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Xiangya Stomatological Hospital and Xiangya School of Stomatology, Central South University, Changsha, Hunan, 410008, China
| | - Lian Liu
- Department of Oral & Maxillofacial Surgery, Xiangya Stomatological Hospital and Xiangya School of Stomatology, Central South University, Changsha, 410000, China
- Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Xiangya Stomatological Hospital and Xiangya School of Stomatology, Central South University, Changsha, Hunan, 410008, China
| | - Weiman Huang
- Department of Oral & Maxillofacial Surgery, Xiangya Stomatological Hospital and Xiangya School of Stomatology, Central South University, Changsha, 410000, China
- Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Xiangya Stomatological Hospital and Xiangya School of Stomatology, Central South University, Changsha, Hunan, 410008, China
| | - Xidi Wang
- Department of Oral & Maxillofacial Surgery, Xiangya Stomatological Hospital and Xiangya School of Stomatology, Central South University, Changsha, 410000, China
- Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Xiangya Stomatological Hospital and Xiangya School of Stomatology, Central South University, Changsha, Hunan, 410008, China
| | - Yanjia Hu
- Department of Oral & Maxillofacial Surgery, Xiangya Stomatological Hospital and Xiangya School of Stomatology, Central South University, Changsha, 410000, China
- Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Xiangya Stomatological Hospital and Xiangya School of Stomatology, Central South University, Changsha, Hunan, 410008, China
| | - Kun Li
- Department of Oral & Maxillofacial Surgery, Xiangya Stomatological Hospital and Xiangya School of Stomatology, Central South University, Changsha, 410000, China.
- Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Xiangya Stomatological Hospital and Xiangya School of Stomatology, Central South University, Changsha, Hunan, 410008, China.
| |
Collapse
|
11
|
Zeng M, Zhu Z, Yuan W, Tang Z, Qing Z, Lu Q, Wu X, He J, Li Y, Li Z. Verapamil inhibits inflammation and promotes autophagy to alleviate ureteral scar by regulation of CaMK IIδ/STAT3 axis. Ren Fail 2024; 46:2387432. [PMID: 39177245 PMCID: PMC11346332 DOI: 10.1080/0886022x.2024.2387432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 07/08/2024] [Accepted: 07/29/2024] [Indexed: 08/24/2024] Open
Abstract
BACKGROUND Ureteral stricture (US) is a pathological stenosis in the urinary tract characterized by increased collagen synthesis and inflammation. Autophagy activation has been shown to ameliorate tissue fibrosis and protect against fibrotic diseases. Verapamil has beneficial therapeutic benefits on fibrotic disorders. The pharmacological effects of verapamil on fibroblast autophagy in US and the underlying mechanism need to be investigated further. METHODS US patients were recruited to isolate scar tissues, hematoxylin-eosin (HE) and Masson trichrome staining were performed to analyze histopathological changes. The US animal model was established and administered with verapamil (0.05 mg/kg) in the drinking water. Transforming growth factor (TGF)-β1 was adopted to facilitate collagen synthesis in fibroblasts. The mRNA and protein expressions were examined by qRT-PCR, western blot, immunofluorescence and immunohistochemistry. ELISA was adopted to measure interleukin (IL)-1β and IL-6 levels. Molecular interaction experiments like dual luciferase reporter and chromatin immunoprecipitation (ChIP) assays were performed to analyze the interaction between signal transducers and activators of transcription 3 (STAT3) and RNA polymerase II associated factor 1 (PAF1). RESULTS Herein, our results revealed that verapamil activated TGF-β1-treated fibroblast autophagy and inhibited inflammation and fibrosis by repressing Ca2+⁄calmodulin-dependent protein kinase II (CaMK II) δ-mediated STAT3 activation. Our following tests revealed that STAT3 activated PAF1 transcription. PAF1 upregulation abrogated the regulatory effect of verapamil on fibroblast autophagy and fibrosis during US progression. Finally, verapamil mitigated US in vivo by activating fibroblast autophagy. CONCLUSION Taken together, verapamil activated TGF-β1-treated fibroblast autophagy and inhibited fibrosis by repressing the CaMK IIδ/STAT3/PAF1 axis.
Collapse
Affiliation(s)
- Mingqiang Zeng
- Department of Urology, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan Province, P.R. China
- Hunan Provincial Institute of Geriatrics, Research Center for Lower Urinary Tract and Pelvic Floor Functional Diseases, Changsha, Hunan Province, P.R. China
| | - Zhiwei Zhu
- Department of Urology, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan Province, P.R. China
| | - Wuxiong Yuan
- Department of Urology, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan Province, P.R. China
| | - Zhengyan Tang
- Provincial Laboratory for Diagnosis and Treatment of Genitourinary System Disease, Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan Province, P.R. China
| | - Zhibiao Qing
- Department of Urology, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan Province, P.R. China
| | - Qiang Lu
- Department of Urology, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan Province, P.R. China
| | - Xuecheng Wu
- Department of Urology, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan Province, P.R. China
| | - Junhuan He
- Department of Urology, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan Province, P.R. China
| | - Yuanwei Li
- Department of Urology, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan Province, P.R. China
| | - Zhuo Li
- Department of Urology, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan Province, P.R. China
| |
Collapse
|
12
|
Chu Y, Yuan X, Tao Y, Yang B, Luo J. Autophagy in Muscle Regeneration: Mechanisms, Targets, and Therapeutic Perspective. Int J Mol Sci 2024; 25:11901. [PMID: 39595972 PMCID: PMC11593790 DOI: 10.3390/ijms252211901] [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: 09/30/2024] [Revised: 10/31/2024] [Accepted: 11/04/2024] [Indexed: 11/28/2024] Open
Abstract
Autophagy maintains the stability of eukaryotic cells by degrading unwanted components and recycling nutrients and plays a pivotal role in muscle regeneration by regulating the quiescence, activation, and differentiation of satellite cells. Effective muscle regeneration is vital for maintaining muscle health and homeostasis. However, under certain disease conditions, such as aging, muscle regeneration can fail due to dysfunctional satellite cells. Dysregulated autophagy may limit satellite cell self-renewal, hinder differentiation, and increase susceptibility to apoptosis, thereby impeding muscle regeneration. This review explores the critical role of autophagy in muscle regeneration, emphasizing its interplay with apoptosis and recent advances in autophagy research related to diseases characterized by impaired muscle regeneration. Additionally, we discuss new approaches involving autophagy regulation to promote macrophage polarization, enhancing muscle regeneration. We suggest that utilizing cell therapy and biomaterials to modulate autophagy could be a promising strategy for supporting muscle regeneration. We hope that this review will provide new insights into the treatment of muscle diseases and promote muscle regeneration.
Collapse
Affiliation(s)
- Yun Chu
- Department of Intensive Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.C.); (Y.T.); (B.Y.)
| | - Xinrun Yuan
- Department of Emergency, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China;
| | - Yiming Tao
- Department of Intensive Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.C.); (Y.T.); (B.Y.)
| | - Bin Yang
- Department of Intensive Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.C.); (Y.T.); (B.Y.)
| | - Jinlong Luo
- Department of Emergency, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China;
| |
Collapse
|
13
|
Song L, Qiu Q, Ju F, Zheng C. Mechanisms of doxorubicin-induced cardiac inflammation and fibrosis; therapeutic targets and approaches. Arch Biochem Biophys 2024; 761:110140. [PMID: 39243924 DOI: 10.1016/j.abb.2024.110140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 08/28/2024] [Accepted: 09/04/2024] [Indexed: 09/09/2024]
Abstract
Doxorubicin plays a pivotal role in the treatment of various malignancies. Despite its efficacy, the cardiotoxicity associated with doxorubicin limits its clinical utility. The cardiotoxic nature of doxorubicin is attributed to several mechanisms, including its interference with mitochondrial function, the generation of reactive oxygen species (ROS), and the subsequent damage to cardiomyocyte DNA, proteins, and lipids. Furthermore, doxorubicin disrupts the homeostasis of cardiac-specific transcription factors and signaling pathways, exacerbating cardiac dysfunction. Oxidative stress, cell death, and other severe changes, such as mitochondrial dysfunction, activation of pro-oxidant enzymes, the renin-angiotensin system (RAS), endoplasmic reticulum (ER) stress, and infiltration of immune cells in the heart after treatment with doxorubicin, may cause inflammatory and fibrotic responses. Fibrosis and inflammation can lead to a range of disorders in the heart, resulting in potential cardiac dysfunction and disease. Various adjuvants have shown potential in preclinical studies to mitigate these challenges associated with cardiac inflammation and fibrosis. Antioxidants, plant-based products, specific inhibitors, and cardioprotective drugs may be recommended to alleviate cardiotoxicity. This review explores the complex mechanisms of doxorubicin-induced heart inflammation and fibrosis, identifies possible cellular and molecular targets, and investigates potential substances that could help reduce these harmful effects.
Collapse
Affiliation(s)
- Linghua Song
- Department of Pharmacy, Yantai Mountain Hospital, Yantai City, Shandong Province, 264001, China
| | - Qingzhuo Qiu
- Medical Imaging Department of Qingdao Women and Children's Hospital, 266000, China
| | - Fei Ju
- Department of Critical Care, Medicine East Hospital of Qingdao Municipal Hospital, 266000, China
| | - Chunyan Zheng
- Cadre Health Office of Zibo Central Hospital in Shandong Province, 255000, China.
| |
Collapse
|
14
|
Yong M, Zhou H, Zeng Y, Yao Y, Zhu H, Hu J. Reduced expression of SMAD7 and consequent reduction of autophagy promotes endometrial stromal-myofibroblast transition and fibrosis. Mol Hum Reprod 2024; 30:gaae036. [PMID: 39412480 DOI: 10.1093/molehr/gaae036] [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: 05/24/2024] [Revised: 09/16/2024] [Indexed: 11/01/2024] Open
Abstract
Abnormal autophagy and the transforming growth factor-β (TGFβ)-SMAD3/7 signaling pathway play an important role in the development of intrauterine adhesions (IUAs); however, the exact underlying mechanisms remain unclear. In this study, we used IUA patient tissue and SMAD7 conditional knockout mice to detect whether SMAD7 effected IUA via regulation of autophagy and the TGFβ-SMAD3 signaling pathway. We applied a combination of techniques for the detection of p-SMAD3, SMAD7, autophagy and fibrosis-related proteins, autophagic flux, and analysis of the SMAD3 binding site. Endometrial tissue of patients with IUA exhibited lower expression levels of SMAD7. In endometrial stromal cells, silencing of SMAD7 inhibited autophagic flux, whereas overexpressed SMAD7 promoted autophagic flux. This SMAD7-mediated autophagic flux regulates the stromal-myofibroblast transition, and these phenotypes were regulated by the TGFβ-SMAD3 signaling pathway. SMAD3 directly binds to the 3'-untranslated region of transcription factor EB (TFEB) and inhibits its transcription. SMAD7 promoted autophagic flux by inhibiting SMAD3, thereby promoting the expression of TFEB. In SMAD7 conditional knockout mice, the endometria showed a fibrotic phenotype. Simultaneously, autophagic flux was inhibited. On administering the autophagy activator rapamycin, this endometrial fibrosis phenotype was partially reversed. The loss of SMAD7 promotes endometrial fibrosis by inhibiting autophagic flux via the TGFβ-SMAD3 pathway. Therefore, this study reveals a potential therapeutic target for IUA.
Collapse
Affiliation(s)
- Min Yong
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, P.R. China
| | - Honggui Zhou
- Department of Obstetrics and Gynecology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, P.R. China
| | - Yuhua Zeng
- Department of Obstetrics and Gynecology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, P.R. China
| | - Yuqin Yao
- Department of Obstetrics and Gynecology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, P.R. China
| | - Hongtao Zhu
- Department of Obstetrics and Gynecology, Second Affiliated Hospital, Chongqing Medical University, Chongqing, P.R. China
| | - Jianguo Hu
- Department of Obstetrics and Gynecology, Second Affiliated Hospital, Chongqing Medical University, Chongqing, P.R. China
| |
Collapse
|
15
|
Chen W, Xu Z, Jiang J, Chen H, Shi R. Identification of LPCAT1 as a key biomarker for Crohn's disease based on bioinformatics and machine learnings and experimental verification. Gene 2024; 920:148519. [PMID: 38703867 DOI: 10.1016/j.gene.2024.148519] [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: 12/20/2023] [Revised: 04/26/2024] [Accepted: 04/30/2024] [Indexed: 05/06/2024]
Abstract
Epithelial-mesenchymal transition (EMT) plays a crucial role in regulating inflammatory responses and fibrosis formation. This study aims to explore the molecular mechanisms of EMT-related genes in Crohn's disease (CD) through bioinformatics methods and identify potential key biomarkers. In our research, we identified differentially expressed genes (DEGs) related to EMT based on the GSE52746 dataset and the gene set in the GeneCards database. Key genes were identified through Lasso-cox and Random Forest and validated using the external dataset GSE10616. Immune infiltration analysis showed that Lysophosphatidylcholine acyltransferase 1 (LPCAT1) was positively correlated with Neutrophils and Macrophages M1. The Gene Set Enrichment Analysis (GSEA) results for LPCAT1 showed associations with celladhesionmolecules and ECM receptor interaction. Additionally, a lncRNA-miRNA-mRNA ceRNA network was constructed. Finally, we validated that knocking down LPCAT1 could inhibit the release of inflammatory factors, EMT, and the elevation of fibrosis indices as well as the activation of NF-κB signaling pathway in LPS-induced HT-29 cells. LPCAT1 plays an important role in the occurrence and development of CD and may become a new biomarker.
Collapse
Affiliation(s)
- Wei Chen
- Department of Gastroenterology, Zhongda Hospital, Southeast University, Nanjing, 210009, China
| | - Zeyan Xu
- Department of Gastroenterology, Zhongda Hospital, Southeast University, Nanjing, 210009, China
| | - Jingjing Jiang
- Department of Gastroenterology, Zhongda Hospital, Southeast University, Nanjing, 210009, China
| | - Hong Chen
- Department of Gastroenterology, Zhongda Hospital, Southeast University, Nanjing, 210009, China.
| | - Ruihua Shi
- Department of Gastroenterology, Zhongda Hospital, Southeast University, Nanjing, 210009, China.
| |
Collapse
|
16
|
Cui X, Zhou Z, Tu H, Wu J, Zhou J, Yi Q, Liu O, Dai X. Mitophagy in fibrotic diseases: molecular mechanisms and therapeutic applications. Front Physiol 2024; 15:1430230. [PMID: 39183973 PMCID: PMC11341310 DOI: 10.3389/fphys.2024.1430230] [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: 05/09/2024] [Accepted: 07/17/2024] [Indexed: 08/27/2024] Open
Abstract
Mitophagy is a highly precise process of selective autophagy, primarily aimed at eliminating excess or damaged mitochondria to maintain the stability of both mitochondrial and cellular homeostasis. In recent years, with in-depth research into the association between mitophagy and fibrotic diseases, it has been discovered that this process may interact with crucial cellular biological processes such as oxidative stress, inflammatory responses, cellular dynamics regulation, and energy metabolism, thereby influencing the occurrence and progression of fibrotic diseases. Consequently, modulating mitophagy holds promise as a therapeutic approach for fibrosis. Currently, various methods have been identified to regulate mitophagy to prevent fibrosis, categorized into three types: natural drug therapy, biological therapy, and physical therapy. This review comprehensively summarizes the current understanding of the mechanisms of mitophagy, delves into its biological roles in fibrotic diseases, and introduces mitophagy modulators effective in fibrosis, aiming to provide new targets and theoretical basis for the investigation of fibrosis-related mechanisms and disease prevention.
Collapse
Affiliation(s)
- Xinyan Cui
- Hunan Key Laboratory of Oral Health Research, Hunan Clinical Research Center of Oral Major Diseases, Oral Health and Academician Workstation for Oral-maxilofacial, Regenerative Medicine and Xiangya Stomatological Hospital, Xiangya School of Stomatology, Central South University, Changsha, Hunan, China
| | - Zekun Zhou
- Hunan Key Laboratory of Oral Health Research, Hunan Clinical Research Center of Oral Major Diseases, Oral Health and Academician Workstation for Oral-maxilofacial, Regenerative Medicine and Xiangya Stomatological Hospital, Xiangya School of Stomatology, Central South University, Changsha, Hunan, China
| | - Hua Tu
- Hunan Key Laboratory of Oral Health Research, Hunan Clinical Research Center of Oral Major Diseases, Oral Health and Academician Workstation for Oral-maxilofacial, Regenerative Medicine and Xiangya Stomatological Hospital, Xiangya School of Stomatology, Central South University, Changsha, Hunan, China
| | - Jianjun Wu
- Hunan Key Laboratory of Oral Health Research, Hunan Clinical Research Center of Oral Major Diseases, Oral Health and Academician Workstation for Oral-maxilofacial, Regenerative Medicine and Xiangya Stomatological Hospital, Xiangya School of Stomatology, Central South University, Changsha, Hunan, China
| | - Jian Zhou
- Salivary Gland Disease Center and Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Beijing Laboratory of Oral Health and Beijing Stomatological Hospital, Capital Medical University, Beijing, China
- Department of VIP Dental Service, School of Stomatology, Capital Medical University, Beijing, China
- Laboratory for Oral and General Health Integration and Translation, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Qiao Yi
- Hunan Key Laboratory of Oral Health Research, Hunan Clinical Research Center of Oral Major Diseases, Oral Health and Academician Workstation for Oral-maxilofacial, Regenerative Medicine and Xiangya Stomatological Hospital, Xiangya School of Stomatology, Central South University, Changsha, Hunan, China
| | - Ousheng Liu
- Hunan Key Laboratory of Oral Health Research, Hunan Clinical Research Center of Oral Major Diseases, Oral Health and Academician Workstation for Oral-maxilofacial, Regenerative Medicine and Xiangya Stomatological Hospital, Xiangya School of Stomatology, Central South University, Changsha, Hunan, China
| | - Xiaohan Dai
- Hunan Key Laboratory of Oral Health Research, Hunan Clinical Research Center of Oral Major Diseases, Oral Health and Academician Workstation for Oral-maxilofacial, Regenerative Medicine and Xiangya Stomatological Hospital, Xiangya School of Stomatology, Central South University, Changsha, Hunan, China
| |
Collapse
|
17
|
Xue W, Wang J, Hou Y, Wu D, Wang H, Jia Q, Jiang Q, Wang Y, Song C, Wang Y, Zhu Z, Tian L. Lung decellularized matrix-derived 3D spheroids: Exploring silicosis through the impact of the Nrf2/Bax pathway on myofibroblast dynamics. Heliyon 2024; 10:e33585. [PMID: 39040273 PMCID: PMC11261893 DOI: 10.1016/j.heliyon.2024.e33585] [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: 03/21/2024] [Revised: 06/23/2024] [Accepted: 06/24/2024] [Indexed: 07/24/2024] Open
Abstract
Silicosis is an occupational respiratory disease caused by long-term inhalation of high concentrations of free silica particles. Studies suggest that oxidative stress is a crucial initiator of silicosis fibrosis, and previous studies have linked the antioxidative stress transcription factor known as Nrf2 to fibrosis antagonism. Myofibroblasts play a pivotal role in tissue damage repair due to oxidative stress. Unlike physiological repair, myofibroblasts in fibrosis exhibit an apoptosis-resistant phenotype, continuously synthesising and secreting significant amounts of collagen and other extracellular matrices, which could be a direct cause of silicosis fibrosis. However, the relationship and mechanism of action between oxidative stress and myofibroblast apoptosis resistance remain unclear. In this study, a new 3D cell culture model using mice lung decellularised matrix particles and fibroblasts was developed, simulating the changes in myofibroblasts during the development of silicotic nodules. Western Blot results indicate that silica stimulation leads to increased collagen deposition and decreased apoptosis-related protein Bax and oxidative stress-related protein Nrf2 in the 3D spheroid model. Immunofluorescence experiments reveal co-localisation in their expression. In Nrf2 overexpressing spheroids, Bax exhibits significant upregulation. In the Nrf2 knockout spheroids, Bax is also significantly downregulated; after intervention with Bax inhibitors, a significant downregulation of Bax-induced apoptosis was also detected in the Nrf2-overexpressed spheroids. In contrast, Bax-induced apoptosis showed a significant upregulation trend in Nrf2-overexpressed spheroids after intervention with Bax agonists. The results demonstrate that the spheroid model can mimic the development process of silicotic nodules, and silica stimulation leads to an apoptosis-resistant phenotype in myofibroblasts in the model, acting through the Nrf2/Bax pathway. This research establishes a new methodology for silicosis study, identifies therapeutic targets for silicosis, and opens new avenues for studying the mechanisms of silicosis fibrosis.
Collapse
Affiliation(s)
- Wenming Xue
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, China
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Jiaxin Wang
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, China
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Yao Hou
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, China
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Di Wu
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, China
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Hongwei Wang
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, China
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Qiyue Jia
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, China
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Qiyue Jiang
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, China
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Yan Wang
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, China
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Chenzhao Song
- Department of Pathology, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
| | - Yifei Wang
- Experimental Teaching Center of Public Heatlh and Preventive Medicine, School of Public Health, Capital Medical University, China
| | - Zhonghui Zhu
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, China
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Lin Tian
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, China
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| |
Collapse
|
18
|
Roodnat AW, Callaghan B, Doyle C, Vallabh NA, Atkinson SD, Willoughby CE. Genome-wide RNA sequencing of ocular fibroblasts from glaucomatous and normal eyes: Implications for glaucoma management. PLoS One 2024; 19:e0307227. [PMID: 38990974 PMCID: PMC11239048 DOI: 10.1371/journal.pone.0307227] [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: 03/28/2024] [Accepted: 06/28/2024] [Indexed: 07/13/2024] Open
Abstract
Primary open angle glaucoma is a leading cause of visual impairment and blindness which is commonly treated with drugs or laser but may require surgery. Tenon's ocular fibroblasts are involved in wound-healing after glaucoma filtration surgery and may compromise a favourable outcome of glaucoma surgery by contributing to fibrosis. To investigate changes in gene expression and key pathways contributing to the glaucomatous state we performed genome-wide RNA sequencing. Human Tenon's ocular fibroblasts were cultured from normal and glaucomatous human donors undergoing eye surgery (n = 12). mRNA was extracted and RNA-Seq performed on the Illumina platform. Differentially expressed genes were identified using a bioinformatics pipeline consisting of FastQC, STAR, FeatureCounts and edgeR. Changes in biological functions and pathways were determined using Enrichr and clustered using Cytoscape. A total of 5817 genes were differentially expressed between Tenon's ocular fibroblasts from normal versus glaucomatous eyes. Enrichment analysis showed 787 significantly different biological functions and pathways which were clustered into 176 clusters. Tenon's ocular fibroblasts from glaucomatous eyes showed signs of fibrosis with fibroblast to myofibroblast transdifferentiation and associated changes in mitochondrial fission, remodeling of the extracellular matrix, proliferation, unfolded protein response, inflammation and apoptosis which may relate to the pathogenesis of glaucoma or the detrimental effects of topical glaucoma therapies. Altered gene expression in glaucomatous Tenon's ocular fibroblasts may contribute to an unfavourable outcome of glaucoma filtration surgery. This work presents a genome-wide transcriptome of glaucomatous versus normal Tenon's ocular fibroblasts which may identify genes or pathways of therapeutic value to improve surgical outcomes.
Collapse
Affiliation(s)
- Anton W. Roodnat
- Centre for Genomic Medicine, Biomedical Sciences Research Institute, Ulster University, Coleraine, Northern Ireland, United Kingdom
| | - Breedge Callaghan
- Centre for Genomic Medicine, Biomedical Sciences Research Institute, Ulster University, Coleraine, Northern Ireland, United Kingdom
| | - Chelsey Doyle
- Centre for Genomic Medicine, Biomedical Sciences Research Institute, Ulster University, Coleraine, Northern Ireland, United Kingdom
| | - Neeru A. Vallabh
- Department of Eye and Vision Science, Insitute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
- St. Paul’s Eye Unit, Liverpool University Hospital NHS Foundation Trust, Liverpool, United Kingdom
| | - Sarah D. Atkinson
- Centre for Genomic Medicine, Biomedical Sciences Research Institute, Ulster University, Coleraine, Northern Ireland, United Kingdom
| | - Colin E. Willoughby
- Centre for Genomic Medicine, Biomedical Sciences Research Institute, Ulster University, Coleraine, Northern Ireland, United Kingdom
| |
Collapse
|
19
|
Bruschi M, Granata S, Candiano G, Petretto A, Bartolucci M, Kajana X, Spinelli S, Verlato A, Provenzano M, Zaza G. Proteomic Changes Induced by the Immunosuppressant Everolimus in Human Podocytes. Int J Mol Sci 2024; 25:7336. [PMID: 39000447 PMCID: PMC11242170 DOI: 10.3390/ijms25137336] [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: 06/18/2024] [Revised: 07/01/2024] [Accepted: 07/02/2024] [Indexed: 07/16/2024] Open
Abstract
mTOR inhibitors (mTOR-Is) may induce proteinuria in kidney transplant recipients through podocyte damage. However, the mechanism has only been partially defined. Total cell lysates and supernatants of immortalized human podocytes treated with different doses of everolimus (EVE) (10, 100, 200, and 500 nM) for 24 h were subjected to mass spectrometry-based proteomics. Support vector machine and partial least squares discriminant analysis were used for data analysis. The results were validated in urine samples from 28 kidney transplant recipients receiving EVE as part of their immunosuppressive therapy. We identified more than 7000 differentially expressed proteins involved in several pathways, including kinases, cell cycle regulation, epithelial-mesenchymal transition, and protein synthesis, according to gene ontology. Among these, after statistical analysis, 65 showed an expression level significantly and directly correlated with EVE dosage. Polo-Like Kinase 1 (PLK1) content was increased, whereas osteopontin (SPP1) content was reduced in podocytes and supernatants in a dose-dependent manner and significantly correlated with EVE dose (p < 0.0001, FDR < 5%). Similar results were obtained in the urine of kidney transplant patients. This study analyzed the impact of different doses of mTOR-Is on podocytes, helping to understand not only the biological basis of their therapeutic effects but also the possible mechanisms underlying proteinuria.
Collapse
Affiliation(s)
- Maurizio Bruschi
- Laboratory of Molecular Nephrology, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy; (M.B.); (G.C.); (X.K.); (S.S.)
- Department of Experimental Medicine (DIMES), University of Genoa, 16132 Genoa, Italy
| | - Simona Granata
- Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy;
| | - Giovanni Candiano
- Laboratory of Molecular Nephrology, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy; (M.B.); (G.C.); (X.K.); (S.S.)
| | - Andrea Petretto
- Proteomics and Clinical Metabolomics Unit at the Core Facilities, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy; (A.P.); (M.B.)
| | - Martina Bartolucci
- Proteomics and Clinical Metabolomics Unit at the Core Facilities, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy; (A.P.); (M.B.)
| | - Xhuliana Kajana
- Laboratory of Molecular Nephrology, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy; (M.B.); (G.C.); (X.K.); (S.S.)
| | - Sonia Spinelli
- Laboratory of Molecular Nephrology, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy; (M.B.); (G.C.); (X.K.); (S.S.)
| | - Alberto Verlato
- Renal Unit, Department of Medicine, University Hospital of Verona, 37124 Verona, Italy;
| | - Michele Provenzano
- Nephrology, Dialysis and Transplantation Unit, Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy;
| | - Gianluigi Zaza
- Nephrology, Dialysis and Transplantation Unit, Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy;
| |
Collapse
|
20
|
Esrefoglu M. Harnessing autophagy: A potential breakthrough in digestive disease treatment. World J Gastroenterol 2024; 30:3036-3043. [PMID: 38983959 PMCID: PMC11230060 DOI: 10.3748/wjg.v30.i24.3036] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 04/30/2024] [Accepted: 06/04/2024] [Indexed: 06/25/2024] Open
Abstract
Autophagy, a conserved cellular degradation process, is crucial for various cellular processes such as immune responses, inflammation, metabolic and oxidative stress adaptation, cell proliferation, development, and tissue repair and remodeling. Dysregulation of autophagy is suspected in numerous diseases, including cancer, neurodegenerative diseases, digestive disorders, metabolic syndromes, and infectious and inflammatory diseases. If autophagy is disrupted, for example, this can have serious consequences and lead to chronic inflammation and tissue damage, as occurs in diseases such as Chron's disease and ulcerative colitis. On the other hand, the influence of autophagy on the development and progression of cancer is not clear. Autophagy can both suppress and promote the progression and metastasis of cancer at various stages. From inflammatory bowel diseases to gastrointestinal cancer, researchers are discovering the intricate role of autophagy in maintaining gut health and its potential as a therapeutic target. Researchers should carefully consider the nature and progression of diseases such as cancer when trying to determine whether inhibiting or stimulating autophagy is likely to be beneficial. Multidisciplinary approaches that combine cutting-edge research with clinical expertise are key to unlocking the full therapeutic potential of autophagy in digestive diseases.
Collapse
Affiliation(s)
- Mukaddes Esrefoglu
- Department of Histology and Embryology, Bezmialem Vakif University Medical Faculty, Istanbul 34093, Türkiye
| |
Collapse
|
21
|
Dang J, Huang S, Li S, Liu J, Chen Z, Wang L, Wang J, Chen H, Xu S. Effects of the Biomimetic Microstructure in Electrospun Fiber Sutures and Mechanical Tension on Tissue Repair. ACS APPLIED MATERIALS & INTERFACES 2024; 16:29087-29097. [PMID: 38788159 DOI: 10.1021/acsami.4c01478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2024]
Abstract
Electrospun microfibers, designed to emulate the extracellular matrix (ECM), play a crucial role in regulating the cellular microenvironment for tissue repair. Understanding their mechanical influence and inherent biological interactions at the ECM interface, however, remains a complex challenge. This study delves into the role of mechanical cues in tissue repair by fabricating Col/PLCL microfibers with varying chemical compositions and alignments that mimic the structure of the ECM. Furthermore, we optimized these microfibers to create the Col/PLCL@PDO aligned suture, with a specific emphasis on mechanical tension in tissue repair. The result reveals that within fibers of identical chemical composition, fibroblast proliferation is more pronounced in aligned fibers than in unaligned ones. Moreover, cells on aligned fibers exhibit an increased aspect ratio. In vivo experiments demonstrated that as the tension increased to a certain level, cell proliferation augmented, cells assumed more elongated morphologies with distinct protrusions, and there was an elevated secretion of collagen III and tension suture, facilitating soft tissue repair. This research illuminates the structural and mechanical dynamics of electrospun fiber scaffolds; it will provide crucial insights for the advancement of precise and controllable tissue engineering materials.
Collapse
Affiliation(s)
- Jie Dang
- Institute for Advanced Study, Shenzhen University, Shenzhen 518060, China
| | - Shifen Huang
- Institute for Advanced Study, Shenzhen University, Shenzhen 518060, China
| | - Shengmei Li
- Institute for Advanced Study, Shenzhen University, Shenzhen 518060, China
| | - Jingyao Liu
- Institute for Advanced Study, Shenzhen University, Shenzhen 518060, China
| | - Zibo Chen
- Institute for Advanced Study, Shenzhen University, Shenzhen 518060, China
| | - Liu Wang
- Institute for Advanced Study, Shenzhen University, Shenzhen 518060, China
| | - Jie Wang
- Institute for Advanced Study, Shenzhen University, Shenzhen 518060, China
| | - Hao Chen
- Institute for Advanced Study, Shenzhen University, Shenzhen 518060, China
| | - Shanshan Xu
- Institute for Advanced Study, Shenzhen University, Shenzhen 518060, China
| |
Collapse
|
22
|
Sun J, Su F, Chen Y, Wang T, Ali W, Jin H, Xiong L, Ma Y, Liu Z, Zou H. Co-exposure to PVC microplastics and cadmium induces oxidative stress and fibrosis in duck pancreas. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 927:172395. [PMID: 38608882 DOI: 10.1016/j.scitotenv.2024.172395] [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: 12/07/2023] [Revised: 03/20/2024] [Accepted: 04/09/2024] [Indexed: 04/14/2024]
Abstract
PVC microplastics (PVC-MPs) are environmental pollutants that interact with cadmium (Cd) to exert various biological effects. Ducks belong to the waterfowl family of birds and therefore are at a higher risk of exposure to PVC-MPs and Cd than other animals. However, the effects of co-exposure of ducks to Cd and PVC-MPs are poorly understood. Here, we used Muscovy ducks to establish an in vivo model to explore the effects of co-exposure to 1 mg/L PVC-MPs and 50 mg/kg Cd on duck pancreas. After 2 months of treatment with 50 mg/kg Cd, pancreas weight decreased by 21 %, and the content of amylase and lipase increased by 25 % and 233 %. However, exposure to PVC-MPs did not significantly affect the pancreas. Moreover, co-exposure to PVC-MPs and Cd worsened the reduction of pancreas weight and disruption of pancreas function compared to exposure to either substance alone. Furthermore, our research has revealed that exposure to PVC-MPs or Cd disrupted mitochondrial structure, reduced ATP levels by 10 % and 18 %, inhibited antioxidant enzyme activity, and increased malondialdehyde levels by 153.8 % and 232.5 %. It was found that exposure to either PVC-MPs or Cd can induce inflammation and fibrosis in the duck pancreas. Notably, co-exposure to PVC-MPs and Cd exacerbated inflammation and fibrosis, with the content of IL-1, IL-6, and TNF-α increasing by 169 %, 199 %, and 98 %, compared to Cd exposure alone. The study emphasizes the significance of comprehending the potential hazards linked to exposure to these substances. In conclusion, it presents promising preliminary evidence that PVC-MPs accumulate in duck pancreas, and increase the accumulation of Cd. Co-exposure to PVC-MPs and Cd disrupts the structure and function of mitochondria and promotes the development of pancreas inflammation and fibrosis.
Collapse
Affiliation(s)
- Jian Sun
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, PR China
| | - Fangyu Su
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, PR China
| | - Yan Chen
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, PR China
| | - Tao Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, PR China
| | - Waseem Ali
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, PR China
| | - Hengqi Jin
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, PR China
| | - Ling Xiong
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Yonggang Ma
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Zongping Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China.
| | - Hui Zou
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China.
| |
Collapse
|
23
|
Xiang Z, Chen H, Xu B, Wang H, Zhang T, Guan X, Ma Z, Liang K, Shi Q. Gelatin/heparin coated bio-inspired polyurethane composite fibers to construct small-caliber artificial blood vessel grafts. Int J Biol Macromol 2024; 269:131849. [PMID: 38670202 DOI: 10.1016/j.ijbiomac.2024.131849] [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: 02/01/2024] [Revised: 04/21/2024] [Accepted: 04/23/2024] [Indexed: 04/28/2024]
Abstract
Long-term patency and ability for revascularization remain challenges for small-caliber blood vessel grafts to treat cardiovascular diseases clinically. Here, a gelatin/heparin coated bio-inspired polyurethane composite fibers-based artificial blood vessel with continuous release of NO and biopeptides to regulate vascular tissue repair and maintain long-term patency is fabricated. A biodegradable polyurethane elastomer that can catalyze S-nitrosothiols in the blood to release NO is synthesized (NPU). Then, the NPU core-shell structured nanofiber grafts with requisite mechanical properties and biopeptide release for inflammation manipulation are fabricated by electrospinning and lyophilization. Finally, the surface of tubular NPU nanofiber grafts is coated with heparin/gelatin and crosslinked with glutaraldehyde to obtain small-caliber artificial blood vessels (ABVs) with the ability of vascular revascularization. We demonstrate that artificial blood vessel grafts promote the growth of endothelial cells but inhibit the growth of smooth muscle cells by the continuous release of NO; vascular grafts can regulate inflammatory balance for vascular tissue remodel without excessive collagen deposition through the release of biological peptides. Vascular grafts prevent thrombus and vascular stenosis to obtain long-term patency. Hence, our work paves a new way to develop small-caliber artificial blood vessel grafts that can maintain long-term patency in vivo and remodel vascular tissue successfully.
Collapse
Affiliation(s)
- Zehong Xiang
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China; University of Science and Technology of China, Hefei, Anhui 230026, China; Zhuhai Institute of Advanced Technology, Chinese Academy of Sciences, Zhuhai, Guangdong 519000, China
| | - Honghong Chen
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China; University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Baofeng Xu
- Stroke Center, Department of Neurology, the First Hospital of Jilin University, Chang Chun 130021, China; Hunan Provincial Key Laboratory of the R&D of Novel Pharmaceutical Preparations, Changsha Medical University, Changsha 410219, China.
| | - Haozheng Wang
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China; University of Science and Technology of China, Hefei, Anhui 230026, China.
| | - Tianci Zhang
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China; University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Xinghua Guan
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China; University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Zhifang Ma
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
| | - Kuntang Liang
- Zhuhai Institute of Advanced Technology, Chinese Academy of Sciences, Zhuhai, Guangdong 519000, China
| | - Qiang Shi
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China; University of Science and Technology of China, Hefei, Anhui 230026, China.
| |
Collapse
|
24
|
İnan S, Barış E. The role of autophagy in odontogenesis, dental implant surgery, periapical and periodontal diseases. J Cell Mol Med 2024; 28:e18297. [PMID: 38613351 PMCID: PMC11015398 DOI: 10.1111/jcmm.18297] [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: 10/31/2023] [Revised: 03/03/2024] [Accepted: 03/25/2024] [Indexed: 04/14/2024] Open
Abstract
Autophagy is a cellular process that is evolutionarily conserved, involving the sequestration of damaged organelles and proteins into autophagic vesicles, which subsequently fuse with lysosomes for degradation. Autophagy controls the development of many diseases by influencing apoptosis, inflammation, the immune response and different cellular processes. Autophagy plays a significant role in the aetiology of disorders associated with dentistry. Autophagy controls odontogenesis. Furthermore, it is implicated in the pathophysiology of pulpitis and periapical disorders. It enhances the survival, penetration and colonization of periodontal pathogenic bacteria into the host periodontal tissues and facilitates their escape from host defences. Autophagy plays a crucial role in mitigating exaggerated inflammatory reactions within the host's system during instances of infection and inflammation. Autophagy also plays a role in the relationship between periodontal disease and systemic diseases. Autophagy promotes wound healing and may enhance implant osseointegration. This study reviews autophagy's dento-alveolar effects, focusing on its role in odontogenesis, periapical diseases, periodontal diseases and dental implant surgery, providing valuable insights for dentists on tooth development and dental applications. A thorough examination of autophagy has the potential to discover novel and efficacious treatment targets within the field of dentistry.
Collapse
Affiliation(s)
- Sevinç İnan
- Department of Oral Pathology, Faculty of DentistryGazi UniversityAnkaraTurkey
| | - Emre Barış
- Department of Oral Pathology, Faculty of DentistryGazi UniversityAnkaraTurkey
| |
Collapse
|
25
|
Gan H, Ma Q, Hao W, Yang N, Chen ZS, Deng L, Chen J. Targeting autophagy to counteract neuroinflammation: A novel antidepressant strategy. Pharmacol Res 2024; 202:107112. [PMID: 38403256 DOI: 10.1016/j.phrs.2024.107112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 02/01/2024] [Accepted: 02/19/2024] [Indexed: 02/27/2024]
Abstract
Depression is a common disease that affects physical and mental health and imposes a considerable burden on afflicted individuals and their families worldwide. Depression is associated with a high rate of disability and suicide. It causes a severe decline in productivity and quality of life. Unfortunately, the pathophysiological mechanisms underlying depression have not been fully elucidated, and the risk of its treatment is still presented. Studies have shown that the expression of autophagic markers in the brain and peripheral inflammatory mediators are dysregulated in depression. Autophagy-related genes regulate the level of autophagy and change the inflammatory response in depression. Depression is related to several aspects of immunity. The regulation of the immune system and inflammation by autophagy may lead to the development or deterioration of mental disorders. This review highlights the role of autophagy and neuroinflammation in the pathophysiology of depression, sumaries the autophagy-targeting small moleculars, and discusses a novel therapeutic strategy based on anti-inflammatory mechanisms that target autophagy to treat the disease.
Collapse
Affiliation(s)
- Hua Gan
- Guangzhou Key Laboratory of Formula-Pattern Research Center, School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China
| | - Qingyu Ma
- Guangzhou Key Laboratory of Formula-Pattern Research Center, School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China
| | - Wenzhi Hao
- Guangzhou Key Laboratory of Formula-Pattern Research Center, School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China
| | - Nating Yang
- Guangzhou Key Laboratory of Formula-Pattern Research Center, School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA.
| | - Lijuan Deng
- Guangzhou Key Laboratory of Formula-Pattern Research Center, School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China.
| | - Jiaxu Chen
- Guangzhou Key Laboratory of Formula-Pattern Research Center, School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China; School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
26
|
Zhou Y, Zheng Z, Wu S, Zhu J. Ubiquitin-conjugating enzyme E2 for regulating autophagy in diabetic cardiomyopathy: A mini-review. J Diabetes 2024; 16:e13511. [PMID: 38052719 PMCID: PMC10925883 DOI: 10.1111/1753-0407.13511] [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: 09/30/2023] [Accepted: 11/18/2023] [Indexed: 12/07/2023] Open
Abstract
The prevalence of diabetic cardiomyopathy (DCM) increases year by year with the increase in the prevalence of diabetes mellitus (DM), which is one of the most serious cardiovascular complications of DM and a major cause of death in diabetic patients. Although the pathological molecular features of DCM have not been fully elucidated, increasing evidence suggests that impaired autophagy in cardiomyocytes plays a nonnegligible role in the development of DCM. It has been shown that SUMOylation [SUMO = small ubiquitin-like modifier], a post-translational modification of proteins, and its associated ubiquitin-proteasome system mediates protein quality control in the heart and plays an important role in the proteotoxic environment of the heart. Specifically, the expression of ubiquitin-conjugating enzyme E2 (Ubc9), the only SUMO-E2 enzyme, exerts a positive regulatory effect on autophagy in cardiomyocytes with potential cardioprotective effects. This review focuses on the role that autophagy plays in DCM and the potential for Ubc9-regulated autophagy pathways to ameliorate DCM, highlighting the potential of Ubc9 as an interventional target in DCM and providing new insights into the pathogenesis of the disease.
Collapse
Affiliation(s)
- Yueran Zhou
- Institute of Clinical Electrocardiology, First Affiliated Hospital of Shantou University Medical CollegeShantouChina
| | - Zequn Zheng
- Institute of Clinical Electrocardiology, First Affiliated Hospital of Shantou University Medical CollegeShantouChina
| | - Shenglin Wu
- Institute of Clinical Electrocardiology, First Affiliated Hospital of Shantou University Medical CollegeShantouChina
| | - Jinxiu Zhu
- Institute of Clinical Electrocardiology, First Affiliated Hospital of Shantou University Medical CollegeShantouChina
- Longgang Maternity and Child Institute of Shantou University Medical College (Longgang District Maternity & Child Healthcare Hospital of Shenzhen City)ShenzhenChina
| |
Collapse
|
27
|
Wu Y, Li L, Ning Z, Li C, Yin Y, Chen K, Li L, Xu F, Gao J. Autophagy-modulating biomaterials: multifunctional weapons to promote tissue regeneration. Cell Commun Signal 2024; 22:124. [PMID: 38360732 PMCID: PMC10868121 DOI: 10.1186/s12964-023-01346-3] [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: 08/24/2023] [Accepted: 09/29/2023] [Indexed: 02/17/2024] Open
Abstract
Autophagy is a self-renewal mechanism that maintains homeostasis and can promote tissue regeneration by regulating inflammation, reducing oxidative stress and promoting cell differentiation. The interaction between biomaterials and tissue cells significantly affects biomaterial-tissue integration and tissue regeneration. In recent years, it has been found that biomaterials can affect various processes related to tissue regeneration by regulating autophagy. The utilization of biomaterials in a controlled environment has become a prominent approach for enhancing the tissue regeneration capabilities. This involves the regulation of autophagy in diverse cell types implicated in tissue regeneration, encompassing the modulation of inflammatory responses, oxidative stress, cell differentiation, proliferation, migration, apoptosis, and extracellular matrix formation. In addition, biomaterials possess the potential to serve as carriers for drug delivery, enabling the regulation of autophagy by either activating or inhibiting its processes. This review summarizes the relationship between autophagy and tissue regeneration and discusses the role of biomaterial-based autophagy in tissue regeneration. In addition, recent advanced technologies used to design autophagy-modulating biomaterials are summarized, and rational design of biomaterials for providing controlled autophagy regulation via modification of the chemistry and surface of biomaterials and incorporation of cells and molecules is discussed. A better understanding of biomaterial-based autophagy and tissue regeneration, as well as the underlying molecular mechanisms, may lead to new possibilities for promoting tissue regeneration. Video Abstract.
Collapse
Affiliation(s)
- Yan Wu
- Heilongjiang Key Laboratory of Tissue Damage and Repair, Mudanjiang Medical University, Mudanjiang, 157000, China
| | - Luxin Li
- Heilongjiang Key Laboratory of Tissue Damage and Repair, Mudanjiang Medical University, Mudanjiang, 157000, China
| | - Zuojun Ning
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Changrong Li
- Heilongjiang Key Laboratory of Tissue Damage and Repair, Mudanjiang Medical University, Mudanjiang, 157000, China
| | - Yongkui Yin
- Heilongjiang Key Laboratory of Tissue Damage and Repair, Mudanjiang Medical University, Mudanjiang, 157000, China
| | - Kaiyuan Chen
- Heilongjiang Key Laboratory of Tissue Damage and Repair, Mudanjiang Medical University, Mudanjiang, 157000, China
| | - Lu Li
- Department of plastic surgery, Naval Specialty Medical Center of PLA, Shanghai, 200052, China.
| | - Fei Xu
- Department of plastic surgery, Naval Specialty Medical Center of PLA, Shanghai, 200052, China.
| | - Jie Gao
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China.
| |
Collapse
|
28
|
Chen Z, Liang H, Yan X, Liang Q, Bai Z, Xie T, Dai J, Zhao X, Xiao Y. Astragalus polysaccharide promotes autophagy and alleviates diabetic nephropathy by targeting the lncRNA Gm41268/PRLR pathway. Ren Fail 2023; 45:2284211. [PMID: 37994436 PMCID: PMC11001349 DOI: 10.1080/0886022x.2023.2284211] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 11/11/2023] [Indexed: 11/24/2023] Open
Abstract
BACKGROUND Astragalus polysaccharide (APS) is a major bioactive component of the Chinese herb astragalus, with well-established protective effects on the kidney. However, the effect of APS on diabetic nephropathy (DN) is unclear. METHODS Long non-coding RNA (lncRNA) expression profiles in kidney samples from control, db/db, and APS-treated db/db mice were evaluated using RNA high-throughput sequencing techniques. Additionally, rat renal tubular epithelial (NRK-52E) cells were cultured in high glucose (HG) media. We inhibited the expression of Gm41268 and prolactin receptor (PRLR) by transfecting NRK-52E cells with Gm41268-targeting antisense oligonucleotides and PRLR siRNA. RESULTS We found that APS treatment reduced 24-h urinary protein levels and fasting blood glucose and improved glucose intolerance and pathological renal damage in db/db mice. Furthermore, APS treatment enhanced autophagy and alleviated fibrosis in the db/db mice. We identified a novel lncRNA, Gm41268, which was differentially expressed in the three groups, and the cis-regulatory target gene PRLR. APS treatment induced autophagy by reducing p62 and p-mammalian target of rapamycin (mTOR) protein levels and increasing the LC3 II/I ratio. Furthermore, APS alleviated fibrosis by downregulating fibronectin (FN), transforming growth factor-β (TGF-β), and collagen IV levels. In addition, APS reversed the HG-induced overexpression of Gm41268 and PRLR. Reduction of Gm41268 decreased PRLR expression, restored autophagy, and ameliorated renal fibrosis in vitro. Inhibition of PRLR could enhance the protective effect of APS. CONCLUSIONS In summary, we demonstrated that the therapeutic effect of APS on DN is mediated via the Gm41268/PRLR pathway. This information contributes to the exploration of bioactive constituents in Chinese herbs as potential treatments for DN.
Collapse
Affiliation(s)
- Zedong Chen
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Huiyu Liang
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Xianxin Yan
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Qiuer Liang
- Affiliated Dongguan People’s Hospital, Southern Medical University (Dongguan People’s Hospital), Guangzhou, China
| | - Zhenyu Bai
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Ting Xie
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Jiaojiao Dai
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Xiaoshan Zhao
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Ya Xiao
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| |
Collapse
|
29
|
Zhang Q, Wang M, Deng X, Zhao D, Zhao F, Xiao J, Ma J, Pan X. Shikonin promotes hypertrophic scar repair by autophagy of hypertrophic scar-derived fibroblasts. Acta Cir Bras 2023; 38:e384623. [PMID: 37878984 PMCID: PMC10592587 DOI: 10.1590/acb384623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 08/14/2023] [Indexed: 10/27/2023] Open
Abstract
PURPOSE To investigate the Shikonin (SHI) induce autophagy of hypertrophic scar-derived fibroblasts (HSFs) and the mechanism of which in repairing hypertrophic scar. METHODS This study showed that SHI induced autophagy from HSFs and repaired skin scars through the AMPK/mTOR pathway. Alamar Blue and Sirius red were used to identify cell activity and collagen. Electron microscopy, label-free quantitative proteomic analysis, fluorescence and other methods were used to identify autophagy. The differences in the expression of autophagy and AMPK/mTOR pathway-related proteins after SHI treatment were quantitatively analyzed by Western blots. A quantitative real-time polymerase chain reaction assay was used to detect the expression of LC3, AMPK and ULK after adding chloroquine (CQ) autophagy inhibitor. RESULTS After treatment with SHI for 24 hours, it was found that the viability of HSFs was significantly reduced, the protein expression of LC3-II/LC3-I and Beclin1 increased, while the protein expression of P62 decreased. The expression of phosphorylated AMPK increased and expression of phosphorylated mTOR decreased. After the use of CQ, the cell autophagy caused by SHI was blocked. The key genes LC3 and P62 were then reexamined by immunohistochemistry using a porcine full-thickness burn hypertrophic scar model, and the results verified that SHI could induce autophagy in vivo. CONCLUSIONS These findings suggested that SHI promoted autophagy of HSFs cells, and the potential mechanism may be related to the AMPK/mTOR signal pathway, which provided new insights for the treatment of hypertrophic scars.
Collapse
Affiliation(s)
- Qing Zhang
- Ningxia Medical University General Hospital - Tissue Organ Bank & Tissue Engineering Centre - c (Ningxia) - China
- Ningxia Medical University - Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education - School of Basic Medicine - Yinchuan (Ningxia) - China
| | - Maomao Wang
- Ningxia Medical University - Clinical Medical School - Yinchuan (Ningxia) - China
| | - Xingwang Deng
- The First People's Hospital - Department of Burns and Plastic Surgery - Shizuishan - China
| | - Dan Zhao
- Ningxia Medical University General Hospital - Tissue Organ Bank & Tissue Engineering Centre - c (Ningxia) - China
| | - Fang Zhao
- Ningxia Medical University General Hospital - Tissue Organ Bank & Tissue Engineering Centre - c (Ningxia) - China
| | - Jinli Xiao
- Ningxia Medical University - Clinical Medical School - Yinchuan (Ningxia) - China
| | - Jiaxiang Ma
- Ningxia Medical University General Hospital - Tissue Organ Bank & Tissue Engineering Centre - c (Ningxia) - China
| | - Xiaoliang Pan
- Ningxia Medical University General Hospital - Tissue Organ Bank & Tissue Engineering Centre - c (Ningxia) - China
| |
Collapse
|
30
|
Titus AS, Sung EA, Zablocki D, Sadoshima J. Mitophagy for cardioprotection. Basic Res Cardiol 2023; 118:42. [PMID: 37798455 PMCID: PMC10556134 DOI: 10.1007/s00395-023-01009-x] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 09/13/2023] [Accepted: 09/14/2023] [Indexed: 10/07/2023]
Abstract
Mitochondrial function is maintained by several strictly coordinated mechanisms, collectively termed mitochondrial quality control mechanisms, including fusion and fission, degradation, and biogenesis. As the primary source of energy in cardiomyocytes, mitochondria are the central organelle for maintaining cardiac function. Since adult cardiomyocytes in humans rarely divide, the number of dysfunctional mitochondria cannot easily be diluted through cell division. Thus, efficient degradation of dysfunctional mitochondria is crucial to maintaining cellular function. Mitophagy, a mitochondria specific form of autophagy, is a major mechanism by which damaged or unnecessary mitochondria are targeted and eliminated. Mitophagy is active in cardiomyocytes at baseline and in response to stress, and plays an essential role in maintaining the quality of mitochondria in cardiomyocytes. Mitophagy is mediated through multiple mechanisms in the heart, and each of these mechanisms can partially compensate for the loss of another mechanism. However, insufficient levels of mitophagy eventually lead to mitochondrial dysfunction and the development of heart failure. In this review, we discuss the molecular mechanisms of mitophagy in the heart and the role of mitophagy in cardiac pathophysiology, with the focus on recent findings in the field.
Collapse
Affiliation(s)
- Allen Sam Titus
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, 185 South Orange Ave, MSB G-609, Newark, NJ, 07103, USA
| | - Eun-Ah Sung
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, 185 South Orange Ave, MSB G-609, Newark, NJ, 07103, USA
| | - Daniela Zablocki
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, 185 South Orange Ave, MSB G-609, Newark, NJ, 07103, USA
| | - Junichi Sadoshima
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, 185 South Orange Ave, MSB G-609, Newark, NJ, 07103, USA.
| |
Collapse
|
31
|
Du Y, Shang Y, Qian Y, Guo Y, Chen S, Lin X, Cao W, Tang X, Zhou A, Huang S, Zhang A, Jia Z, Zhang Y. Plk1 promotes renal tubulointerstitial fibrosis by targeting autophagy/lysosome axis. Cell Death Dis 2023; 14:571. [PMID: 37640723 PMCID: PMC10462727 DOI: 10.1038/s41419-023-06093-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 08/07/2023] [Accepted: 08/21/2023] [Indexed: 08/31/2023]
Abstract
The prevalence of chronic kidney disease (CKD) has been increasing over the past decades. However, no effective therapies are available for delaying or curing CKD. Progressive fibrosis is the major pathological feature of CKD, which leads to end-stage renal disease (ESRD). The present study showed that Polo-like kinase 1 (Plk1) was upregulated in the kidneys of CKD patients and mice subjected to unilateral ureteral obstruction (UUO) with location in proximal tubules and tubulointerstitial fibroblasts. Pharmacological inhibition, genetic silencing or knockout of Plk1 attenuated obstructive nephropathy due to suppressed fibroblast activation mediated by reduced autophagic flux. We found Plk1 plays a critical role in maintaining intralysosomal pH by regulating ATP6V1A phosphorylation, and inhibition of Plk1 impaired lysosomal function leading to blockade of autophagic flux. In addition, Plk1 also prevented partial epithelial-mesenchymal transition (pEMT) of tubular epithelial cells via autophagy pathway. In conclusion, this study demonstrated that Plk1 plays a pathogenic role in renal tubulointerstitial fibrosis by regulating autophagy/lysosome axis. Thus, targeting Plk1 could be a promising strategy for CKD treatment.
Collapse
Affiliation(s)
- Yang Du
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Guangzhou Road #72, Gulou District, 210008, Nanjing, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Hanzhong Road #140, Gulou District, 210029, Nanjing, China
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Gulou District, Guangzhou Road #72, 210008, Nanjing, China
| | - Yaqiong Shang
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Guangzhou Road #72, Gulou District, 210008, Nanjing, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Hanzhong Road #140, Gulou District, 210029, Nanjing, China
| | - Yun Qian
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Guangzhou Road #72, Gulou District, 210008, Nanjing, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Hanzhong Road #140, Gulou District, 210029, Nanjing, China
| | - Yan Guo
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Guangzhou Road #72, Gulou District, 210008, Nanjing, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Hanzhong Road #140, Gulou District, 210029, Nanjing, China
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Gulou District, Guangzhou Road #72, 210008, Nanjing, China
| | - Shuang Chen
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Guangzhou Road #72, Gulou District, 210008, Nanjing, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Hanzhong Road #140, Gulou District, 210029, Nanjing, China
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Gulou District, Guangzhou Road #72, 210008, Nanjing, China
| | - Xiuli Lin
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Guangzhou Road #72, Gulou District, 210008, Nanjing, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Hanzhong Road #140, Gulou District, 210029, Nanjing, China
| | - Weidong Cao
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Guangzhou Road #72, Gulou District, 210008, Nanjing, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Hanzhong Road #140, Gulou District, 210029, Nanjing, China
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Gulou District, Guangzhou Road #72, 210008, Nanjing, China
| | - Xiaomei Tang
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Guangzhou Road #72, Gulou District, 210008, Nanjing, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Hanzhong Road #140, Gulou District, 210029, Nanjing, China
| | - Anning Zhou
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Guangzhou Road #72, Gulou District, 210008, Nanjing, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Hanzhong Road #140, Gulou District, 210029, Nanjing, China
| | - Songming Huang
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Guangzhou Road #72, Gulou District, 210008, Nanjing, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Hanzhong Road #140, Gulou District, 210029, Nanjing, China
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Gulou District, Guangzhou Road #72, 210008, Nanjing, China
| | - Aihua Zhang
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Guangzhou Road #72, Gulou District, 210008, Nanjing, China.
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Hanzhong Road #140, Gulou District, 210029, Nanjing, China.
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Gulou District, Guangzhou Road #72, 210008, Nanjing, China.
| | - Zhanjun Jia
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Guangzhou Road #72, Gulou District, 210008, Nanjing, China.
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Hanzhong Road #140, Gulou District, 210029, Nanjing, China.
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Gulou District, Guangzhou Road #72, 210008, Nanjing, China.
| | - Yue Zhang
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Guangzhou Road #72, Gulou District, 210008, Nanjing, China.
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Hanzhong Road #140, Gulou District, 210029, Nanjing, China.
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Gulou District, Guangzhou Road #72, 210008, Nanjing, China.
| |
Collapse
|
32
|
Lopez-Soler RI, Nikouee A, Kim M, Khan S, Sivaraman L, Ding X, Zang QS. Beclin-1 dependent autophagy improves renal outcomes following Unilateral Ureteral Obstruction (UUO) injury. Front Immunol 2023; 14:1104652. [PMID: 36875088 PMCID: PMC9978333 DOI: 10.3389/fimmu.2023.1104652] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 01/31/2023] [Indexed: 02/18/2023] Open
Abstract
Background Interstitial Fibrosis and Tubular Atrophy (IFTA) is the most common cause of long-term graft failure following renal transplant. One of the hallmarks of IFTA is the development of interstitial fibrosis and loss of normal renal architecture. In this study, we evaluated the role of autophagy initiation factor Beclin-1 in protecting against post-renal injury fibrosis. Methods Adult male wild type (WT) C57BL/6 mice were subjected to Unilateral Ureteral Obstruction (UUO), and kidney tissue samples were harvested at 72-hour, 1- and 3-week post-injury. The UUO-injured and uninjured kidney samples were examined histologically for fibrosis, autophagy flux, inflammation as well activation of the Integrated Stress Response (ISR). We compared WT mice with mice carrying a forced expression of constitutively active mutant form of Beclin-1, Becn1F121A/F121A . Results In all experiments, UUO injury induces a progressive development of fibrosis and inflammation. These pathological signs were diminished in Becn1F121A/F121A mice. In WT animals, UUO caused a strong blockage of autophagy flux, indicated by continuously increases in LC3II accompanied by an over 3-fold accumulation of p62 1-week post injury. However, increases in LC3II and unaffected p62 level by UUO were observed in Becn1F121A/F121A mice, suggesting an alleviation of disrupted autophagy. Beclin-1 F121A mutation causes a significant decrease in phosphorylation of inflammatory STING signal and limited production of IL6 and IFNγ, but had little effect on TNF-α, in response to UUO. Furthermore, activation of ISR signal cascade was detected in UUO-injured in kidneys, namely the phosphorylation signals of elF2S1 and PERK in addition to the stimulated expression of ISR effector ATF4. However, Becn1F121A/F121A mice did not reveal signs of elF2S1 and PERK activation under the same condition and had a dramatically reduced ATF level at 3-week post injury. Conclusions The results suggest that UUO causes a insufficient, maladaptive renal autophagy, which triggered downstream activation of inflammatory STING pathway, production of cytokines, and pathological activation of ISR, eventually leading to the development of fibrosis. Enhancing autophagy via Beclin-1 improved renal outcomes with diminished fibrosis, via underlying mechanisms of differential regulation of inflammatory mediators and control of maladaptive ISR.
Collapse
Affiliation(s)
- Reynold I. Lopez-Soler
- Section of Renal Transplantation, Edward Hines Jr. VA Hospital, Hines, IL, United States
- Department of Surgery, Division of Intra-Abdominal Transplantation, Loyola University Chicago Stritch School of Medicine, Maywood, IL, United States
| | - Azadeh Nikouee
- Department of Surgery, Burn & Shock Trauma Research Institute; Loyola University Chicago Stritch School of Medicine, Maywood, IL, United States
| | - Matthew Kim
- Department of Surgery, Burn & Shock Trauma Research Institute; Loyola University Chicago Stritch School of Medicine, Maywood, IL, United States
| | - Saman Khan
- Department of Surgery, Burn & Shock Trauma Research Institute; Loyola University Chicago Stritch School of Medicine, Maywood, IL, United States
| | - Lakshmi Sivaraman
- Department of Biology, Loyola University Chicago, Chicago, IL, United States
| | - Xiangzhong Ding
- Department of Pathology, Loyola University Chicago Stritch School of Medicine, Maywood, IL, United States
| | - Qun Sophia Zang
- Department of Surgery, Burn & Shock Trauma Research Institute; Loyola University Chicago Stritch School of Medicine, Maywood, IL, United States
| |
Collapse
|
33
|
Zhang J, Li L, Yu J, Zhang F, Shi J, LI M, Liu J, Li H, Gao J, Wu Y. Autophagy-Modulated Biomaterial: A Robust Weapon for Modulating the Wound Environment to Promote Skin Wound Healing. Int J Nanomedicine 2023; 18:2567-2588. [PMID: 37213350 PMCID: PMC10198186 DOI: 10.2147/ijn.s398107] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 03/28/2023] [Indexed: 05/23/2023] Open
Abstract
Autophagy, a self-renewal mechanism, can help to maintain the stability of the intracellular environment of organisms. Autophagy can also regulate several cellular functions and is strongly related to the onset and progression of several diseases. Wound healing is a biological process that is coregulated by different types of cells. However, it is troublesome owing to prolonged treatment duration and poor recovery. In recent years, biomaterials have been reported to influence the skin wound healing process by finely regulating autophagy. Biomaterials that regulate autophagy in various cells involved in skin wound healing to regulate the differentiation, proliferation and migration of cells, inflammatory responses, oxidative stress and formation of the extracellular matrix (ECM) have emerged as a key method for improving the tissue regeneration ability of biomaterials. During the inflammatory phase, autophagy enhances the clearance of pathogens from the wound site and leads to macrophage polarization from the M1 to the M2 phenotype, thus preventing enhanced inflammation that can lead to further tissue damage. Autophagy plays important roles in facilitating the formation of extracellular matrix (ECM) during the proliferative phase, removing excess intracellular ROS, and promoting the proliferation and differentiation of endothelial cells, fibroblasts, and keratinocytes. This review summarizes the close association between autophagy and skin wound healing and discusses the role of biomaterial-based autophagy in tissue regeneration. The applications of recent biomaterials designed to target autophagy are highlighted, including polymeric materials, cellular materials, metal nanomaterials, and carbon-based materials. A better understanding of biomaterial-regulated autophagy and skin regeneration and the underlying molecular mechanisms may open new possibilities for promoting skin regeneration. Moreover, this can lay the foundation for the development of more effective therapeutic approaches and novel biomaterials for clinical applications.
Collapse
Affiliation(s)
- Jin Zhang
- College of Life Science, Mudanjiang Medical University, Mudanjiang, People’s Republic of China
| | - Luxin Li
- College of Life Science, Mudanjiang Medical University, Mudanjiang, People’s Republic of China
| | - Jing Yu
- Department of Endocrinology, Hongqi Hospital Affiliated to Mudanjiang Medical University, Mudanjiang, 157011, People’s Republic of China
| | - Fan Zhang
- College of Life Science, Mudanjiang Medical University, Mudanjiang, People’s Republic of China
| | - Jiayi Shi
- College of Life Science, Mudanjiang Medical University, Mudanjiang, People’s Republic of China
| | - Meiyun LI
- College of Life Science, Mudanjiang Medical University, Mudanjiang, People’s Republic of China
| | - Jianyong Liu
- Department of Vascular Surgery, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Haitao Li
- Department of Vascular Surgery, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Jie Gao
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, People’s Republic of China
- Jie Gao, Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, People’s Republic of China, Tel/Fax +86 21-31166666, Email
| | - Yan Wu
- College of Life Science, Mudanjiang Medical University, Mudanjiang, People’s Republic of China
- Correspondence: Yan Wu, College of Life Science, Mudanjiang Medical University, Mudanjiang, Heilongjiang, 157001, People’s Republic of China, Tel/Fax +86-453-6984647, Email
| |
Collapse
|
34
|
Yue YL, Zhang MY, Liu JY, Fang LJ, Qu YQ. The role of autophagy in idiopathic pulmonary fibrosis: from mechanisms to therapies. Ther Adv Respir Dis 2022; 16:17534666221140972. [PMID: 36468453 PMCID: PMC9726854 DOI: 10.1177/17534666221140972] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is an interstitial pulmonary disease with an extremely poor prognosis. Autophagy is a fundamental intracellular process involved in maintaining cellular homeostasis and regulating cell survival. Autophagy deficiency has been shown to play an important role in the progression of pulmonary fibrosis. This review focused on the six steps of autophagy, as well as the interplay between autophagy and other seven pulmonary fibrosis related mechanisms, which include extracellular matrix deposition, myofibroblast differentiation, epithelial-mesenchymal transition, pulmonary epithelial cell dysfunction, apoptosis, TGF-β1 pathway, and the renin-angiotensin system. In addition, this review also summarized autophagy-related signaling pathways such as mTOR, MAPK, JAK2/STAT3 signaling, p65, and Keap1/Nrf2 signaling during the development of IPF. Furthermore, this review also illustrated the commonly used autophagy detection methods, the currently approved antifibrotic drugs pirfenidone and nintedanib, and several prospective compounds targeting autophagy for the treatment of IPF.
Collapse
Affiliation(s)
- Yue-Liang Yue
- Shandong Key Laboratory of Infectious Respiratory Diseases, Laboratory of Basic Medical Sciences, Department of Pulmonary and Critical Care Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Meng-Yu Zhang
- Shandong Key Laboratory of Infectious Respiratory Diseases, Laboratory of Basic Medical Sciences, Department of Pulmonary and Critical Care Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Jian-Yu Liu
- Shandong Key Laboratory of Infectious Respiratory Diseases, Laboratory of Basic Medical Sciences, Department of Pulmonary and Critical Care Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Li-Jun Fang
- Shandong Key Laboratory of Infectious Respiratory Diseases, Laboratory of Basic Medical Sciences, Department of Pulmonary and Critical Care Medicine, Qilu Hospital of Shandong University, Jinan, China
| | | |
Collapse
|
35
|
Bao L, Ye J, Liu N, Shao Y, Li W, Fan X, Zhao D, Wang H, Chen X. Resveratrol Ameliorates Fibrosis in Rheumatoid Arthritis-Associated Interstitial Lung Disease via the Autophagy-Lysosome Pathway. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27238475. [PMID: 36500562 PMCID: PMC9740423 DOI: 10.3390/molecules27238475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/15/2022] [Accepted: 11/23/2022] [Indexed: 12/11/2022]
Abstract
Interstitial lung disease associated with rheumatoid arthritis (RA-ILD) can lead to interstitial fibrosis and even lung failure as a complication of rheumatoid arthritis (RA), and there is currently no effective treatment and related basic research. Studies have found that resveratrol (Res) can improve the progression of RA by regulating autophagy, and increasing evidence supports the connection between autophagy and common interstitial lung disease (ILD). We explored changes in autophagy levels in fibrotic lungs in RA-ILD and found that the level of autophagy is enhanced in the early stage but inhibited in the late stage. However, resveratrol treatment improved the level of autophagy and reversed the inhibition of autophagy, and attenuated fibrosis. We created corresponding cell models that exhibited the same phenotypic changes as animal models; under the effect of resveratrol, the level of fibrosis changed accordingly, and the fusion process of lysosomes and autophagosomes in autophagy was liberated from the inhibition state. Resveratrol effects were reversed by the addition of the late autophagy inhibitor chloroquine. These results suggest that resveratrol attenuates pulmonary fibrosis, increases autophagic flux, and modulates the autophagy-lysosome pathway, and particularly it may work by improving the formation of autophagic lysosomes, which may be an effective treatment for induced RA-ILD.
Collapse
Affiliation(s)
- Lanxin Bao
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230601, China
| | - Jing Ye
- Department of Respiratory Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Nannan Liu
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230601, China
| | - Yubao Shao
- Microscopic Morphological Center Laboratory, Anhui Medical University, Hefei 230032, China
| | - Wenhao Li
- Department of Clinical Medicine, Anhui Medical University, Hefei 230032, China
| | - Xuefei Fan
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230601, China
| | - Dahai Zhao
- Department of Respiratory Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
- Correspondence: (D.Z.); (H.W.); (X.C.)
| | - Hongzhi Wang
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230601, China
- Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei 230031, China
- Correspondence: (D.Z.); (H.W.); (X.C.)
| | - Xiaoyu Chen
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230601, China
- Microscopic Morphological Center Laboratory, Anhui Medical University, Hefei 230032, China
- Correspondence: (D.Z.); (H.W.); (X.C.)
| |
Collapse
|
36
|
Cheng D, Li Z, Wang Y, Xiong H, Sun W, Zhou S, Liu Y, Ni C. Targeted delivery of ZNF416 siRNA-loaded liposomes attenuates experimental pulmonary fibrosis. J Transl Med 2022; 20:523. [PMID: 36371191 PMCID: PMC9652794 DOI: 10.1186/s12967-022-03740-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 10/29/2022] [Indexed: 11/15/2022] Open
Abstract
Background Pulmonary fibrosis is a chronic progressive fibrotic interstitial lung disease characterized by excessive extracellular matrix (ECM) deposition caused by activated fibroblasts. Increasing evidence shows that matrix stiffness is essential in promoting fibroblast activation and profibrotic changes. Here, we investigated the expression and function of matrix stiffness-regulated ZNF416 in pulmonary fibrotic lung fibroblasts. Methods 1 kappa (soft), 60 kappa (stiff) gel-coated coverslips, or transforming growth factor-beta 1 (TGF-β1)-cultured lung fibroblasts and the gain- or loss- of the ZNF416 function assays were performed in vitro. We also established two experimental pulmonary fibrosis mouse models by a single intratracheal instillation with 50 mg/kg silica or 6 mg/kg bleomycin (BLM). ZNF416 siRNA-loaded liposomes and TGF-β1 receptor inhibitor SB431542 were administrated in vivo. Results Our study identified that ZNF416 could regulate fibroblast differentiation, proliferation, and contraction by promoting the nuclear accumulation of p-Smad2/3. Besides, ZNF416 siRNA-loaded liposome delivery by tail-vein could passively target the fibrotic area in the lung, and co-administration of ZNF416 siRNA-loaded liposomes and SB431542 significantly protects mice against silica or BLM-induced lung injury and fibrosis. Conclusion In this study, our results indicate that mechanosensitive ZNF416 is a potential molecular target for the treatment of pulmonary fibrosis. Strategies aimed at silencing ZNF416 could be a promising approach to fight against pulmonary fibrosis. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-022-03740-w.
Collapse
|
37
|
Bertova A, Kontar S, Polozsanyi Z, Simkovic M, Rosenbergova Z, Rebros M, Sulova Z, Breier A, Imrichova D. Effects of Sulforaphane-Induced Cell Death upon Repeated Passage of Either P-Glycoprotein-Negative or P-Glycoprotein-Positive L1210 Cell Variants. Int J Mol Sci 2022; 23:ijms231810818. [PMID: 36142752 PMCID: PMC9501161 DOI: 10.3390/ijms231810818] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 09/11/2022] [Accepted: 09/13/2022] [Indexed: 11/21/2022] Open
Abstract
The expression of the membrane ABCB1 transporter in neoplastic cells is one of the most common causes of reduced sensitivity to chemotherapy. In our previous study, we investigated the effect of a single culture of ABCB1-negative (S) and ABCB1-positive variants of L1210 cells (R and T) in the presence of sulforaphane (SFN). We demonstrated that SFN induces the onset of autophagy more markedly in S cells than in R or T cells. In the current study, we focused on the effect of the repeated culture of S, R and T cells in SFN-containing media. The repeated cultures increased the onset of autophagy compared to the simple culture, mainly in S cells and to a lesser extent in R and T cells, as indicated by changes in the cellular content of 16 and 18 kDa fragments of LC3B protein or changes in the specific staining of cells with monodansylcadaverine. We conclude that SFN affects ABCB1-negative S cells more than ABCB1-positive R and T cells during repeated culturing. Changes in cell sensitivity to SFN appear to be related to the expression of genes for cell-cycle checkpoints, such as cyclins and cyclin-dependent kinases.
Collapse
Affiliation(s)
- Anna Bertova
- Institute of Molecular Physiology and Genetics, Centre of Biosciences, Slovak Academy of Sciences, Dúbravská cesta 9, 840 05 Bratislava, Slovakia
| | - Szilvia Kontar
- Institute of Molecular Physiology and Genetics, Centre of Biosciences, Slovak Academy of Sciences, Dúbravská cesta 9, 840 05 Bratislava, Slovakia
| | - Zoltan Polozsanyi
- Institute of Biochemistry and Microbiology, Faculty of Chemical and Food Technology, Slovak University of Technology in Bratislava, Radlinského 9, 812 37 Bratislava, Slovakia
| | - Martin Simkovic
- Institute of Biochemistry and Microbiology, Faculty of Chemical and Food Technology, Slovak University of Technology in Bratislava, Radlinského 9, 812 37 Bratislava, Slovakia
| | - Zuzana Rosenbergova
- Institute of Biotechnology, Faculty of Chemical and Food Technology, Slovak University of Technology in Bratislava, Radlinského 9, 812 37 Bratislava, Slovakia
| | - Martin Rebros
- Institute of Biotechnology, Faculty of Chemical and Food Technology, Slovak University of Technology in Bratislava, Radlinského 9, 812 37 Bratislava, Slovakia
| | - Zdena Sulova
- Institute of Molecular Physiology and Genetics, Centre of Biosciences, Slovak Academy of Sciences, Dúbravská cesta 9, 840 05 Bratislava, Slovakia
| | - Albert Breier
- Institute of Molecular Physiology and Genetics, Centre of Biosciences, Slovak Academy of Sciences, Dúbravská cesta 9, 840 05 Bratislava, Slovakia
- Institute of Biochemistry and Microbiology, Faculty of Chemical and Food Technology, Slovak University of Technology in Bratislava, Radlinského 9, 812 37 Bratislava, Slovakia
- Correspondence: (A.B.); (D.I.)
| | - Denisa Imrichova
- Institute of Molecular Physiology and Genetics, Centre of Biosciences, Slovak Academy of Sciences, Dúbravská cesta 9, 840 05 Bratislava, Slovakia
- Correspondence: (A.B.); (D.I.)
| |
Collapse
|
38
|
Ripszky Totan A, Imre MM, Parvu S, Meghea D, Radulescu R, Enasescu DSA, Moisa MR, Pituru SM. Autophagy Plays Multiple Roles in the Soft-Tissue Healing and Osseointegration in Dental Implant Surgery-A Narrative Review. MATERIALS (BASEL, SWITZERLAND) 2022; 15:6041. [PMID: 36079421 PMCID: PMC9457242 DOI: 10.3390/ma15176041] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 08/21/2022] [Accepted: 08/30/2022] [Indexed: 06/15/2023]
Abstract
Dental endo-osseous implants have become a widely used treatment for replacing missing teeth. Dental implants are placed into a surgically created osteotomy in alveolar bone, the healing of the soft tissue lesion and the osseointegration of the implant being key elements to long-term success. Autophagy is considered the major intracellular degradation system, playing important roles in various cellular processes involved in dental implant integration. The aim of this review is an exploration of autophagy roles in the main cell types involved in the healing and remodeling of soft tissue lesions and implant osseointegration, post-implant surgery. We have focused on the autophagy pathway in macrophages, endothelial cells; osteoclasts, osteoblasts; fibroblasts, myofibroblasts and keratinocytes. In macrophages, autophagy modulates innate and adaptive immune responses playing a key role in osteo-immunity. Autophagy induction in endothelial cells promotes apoptosis resistance, cell survival, and protection against oxidative stress damage. The autophagic machinery is also involved in transporting stromal vesicles containing mineralization-related factors to the extracellular matrix and regulating osteoblasts' functions. Alveolar bone remodeling is achieved by immune cells differentiation into osteoclasts; autophagy plays an important and active role in this process. Autophagy downregulation in fibroblasts induces apoptosis, leading to better wound healing by improving excessive deposition of extracellular matrix and inhibiting fibrosis progression. Autophagy seems to be a dual actor on the scene of dental implant surgery, imposing further research in order to completely reveal its positive features which may be essential for clinical efficacy.
Collapse
Affiliation(s)
- Alexandra Ripszky Totan
- Department of Biochemistry, Faculty of Dental Medicine, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Marina Melescanu Imre
- Department of Complete Denture, Faculty of Dental Medicine, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Simona Parvu
- Department of Complementary Sciences, Hygiene and Medical Ecology Discipline, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Daniela Meghea
- Department of Complete Denture, Faculty of Dental Medicine, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Radu Radulescu
- Department of Biochemistry, Faculty of Dental Medicine, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Dan Sebastian Alexandru Enasescu
- Department of Biochemistry, Faculty of Dental Medicine, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Mihai Radu Moisa
- Department of Biochemistry, Faculty of Dental Medicine, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Silviu Mirel Pituru
- Department of Professional Organization and Medical Legislation-Malpractice, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| |
Collapse
|
39
|
Shim MS, Liton PB. The physiological and pathophysiological roles of the autophagy lysosomal system in the conventional aqueous humor outflow pathway: More than cellular clean up. Prog Retin Eye Res 2022; 90:101064. [PMID: 35370083 PMCID: PMC9464695 DOI: 10.1016/j.preteyeres.2022.101064] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 03/09/2022] [Accepted: 03/25/2022] [Indexed: 10/18/2022]
Abstract
During the last few years, the autophagy lysosomal system is emerging as a central cellular pathway with roles in survival, acting as a housekeeper and stress response mechanism. Studies by our and other labs suggest that autophagy might play an essential role in maintaining aqueous humor outflow homeostasis, and that malfunction of autophagy in outflow pathway cells might predispose to ocular hypertension and glaucoma pathogenesis. In this review, we will collect the current knowledge and discuss the molecular mechanisms by which autophagy does or might regulate normal outflow pathway tissue function, and its response to different types of stressors (oxidative stress and mechanical stress). We will also discuss novel roles of autophagy and lysosomal enzymes in modulation of TGFβ signaling and ECM remodeling, and the link between dysregulated autophagy and cellular senescence. We will examine what we have learnt, using pre-clinical animal models about how dysregulated autophagy can contribute to disease and apply that to the current status of autophagy in human glaucoma. Finally, we will consider and discuss the challenges and the potential of autophagy as a therapeutic target for the treatment of ocular hypertension and glaucoma.
Collapse
Affiliation(s)
- Myoung Sup Shim
- Duke University, Department of Ophthalmology, Durham, NC, 27705, USA
| | - Paloma B Liton
- Duke University, Department of Ophthalmology, Durham, NC, 27705, USA.
| |
Collapse
|
40
|
Ren H, Zhao F, Zhang Q, Huang X, Wang Z. Autophagy and skin wound healing. BURNS & TRAUMA 2022; 10:tkac003. [PMID: 35187180 PMCID: PMC8847901 DOI: 10.1093/burnst/tkac003] [Citation(s) in RCA: 94] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 01/07/2022] [Indexed: 02/07/2023]
Abstract
Autophagy is a lysosome-dependent, self-renewal mechanism that can degrade and recycle cellular components in eukaryotic cells to maintain the stability of the intracellular environment and the cells ability to cope with unfavorable environments. Numerous studies suggest that autophagy participates in regulating various cellular functions and is closely associated with the onset and progression of various diseases. Wound healing is a complex, multistep biological process that involves multiple cell types. Refractory wounds, which include diabetic skin ulcers, can seriously endanger human health. Previous studies have confirmed that autophagy plays an essential role in various phases of wound healing. Specifically, in the inflammatory phase, autophagy has an anti-infection effect and it negatively regulates the inflammatory response, which prevents excessive inflammation from causing tissue damage. In the proliferative phase, local hypoxia in the wound can induce autophagy, which plays a role in anti-apoptosis and anti-oxidative stress and promotes cell survival. Autophagy of vascular endothelial cells promotes wound angiogenesis and that of keratinocytes promotes their differentiation, proliferation and migration, which is conducive to the completion of wound re-epithelialisation. In the remodeling phase, autophagy of fibroblasts affects the formation of hypertrophic scars. Additionally, a refractory diabetic wound may be associated with increased levels of autophagy, and the regulation of mesenchymal stem cell autophagy may improve its application to wound healing. Therefore, understanding the relationship between autophagy and skin wound healing and exploring the molecular mechanism of autophagy regulation may provide novel strategies for the clinical treatment of wound healing.
Collapse
Affiliation(s)
- Haiyue Ren
- Department of Pathology, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, Shenyang City 110004, Liaoning Province, China
| | - Feng Zhao
- Department of Stem Cells and Regenerative Medicine, Shenyang Key Laboratory of Stem Cell and Regenerative Medicine, China Medical University, Shenyang 110013, Liaoning, China
| | - Qiqi Zhang
- Department of Pathology, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, Shenyang City 110004, Liaoning Province, China
| | - Xing Huang
- Department of General Surgery, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, Shenyang City 110004, Liaoning Province, China
| | - Zhe Wang
- Department of Pathology, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, Shenyang City 110004, Liaoning Province, China
| |
Collapse
|
41
|
Baghaei K, Mazhari S, Tokhanbigli S, Parsamanesh G, Alavifard H, Schaafsma D, Ghavami S. Therapeutic potential of targeting regulatory mechanisms of hepatic stellate cell activation in liver fibrosis. Drug Discov Today 2021; 27:1044-1061. [PMID: 34952225 DOI: 10.1016/j.drudis.2021.12.012] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 11/11/2021] [Accepted: 12/17/2021] [Indexed: 11/03/2022]
Abstract
Hepatic fibrosis is a manifestation of different etiologies of liver disease with the involvement of multiple mediators in complex network interactions. Activated hepatic stellate cells (aHSCs) are the central driver of hepatic fibrosis, given their potential to induce connective tissue formation and extracellular matrix (ECM) protein accumulation. Therefore, identifying the cellular and molecular pathways involved in the activation of HSCs is crucial in gaining mechanistic and therapeutic perspectives to more effectively target the disease. In addition to a comprehensive summary of our current understanding of the role of HSCs in liver fibrosis, we also discuss here the proposed therapeutic strategies based on targeting HSCs.
Collapse
Affiliation(s)
- Kaveh Baghaei
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran 1985717413, Iran; Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran 1985717413, Iran
| | - Sogol Mazhari
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran 1985717413, Iran
| | - Samaneh Tokhanbigli
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran 1985717413, Iran
| | - Gilda Parsamanesh
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran 1985717413, Iran
| | - Helia Alavifard
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran 1985717413, Iran
| | | | - Saeid Ghavami
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada.
| |
Collapse
|
42
|
Reparation of an Inflamed Air-Liquid Interface Cultured A549 Cells with Nebulized Nanocurcumin. Pharmaceutics 2021; 13:pharmaceutics13091331. [PMID: 34575407 PMCID: PMC8466083 DOI: 10.3390/pharmaceutics13091331] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/15/2021] [Accepted: 08/20/2021] [Indexed: 12/29/2022] Open
Abstract
The anti-inflammatory, antifibrotic and antimicrobial activities of curcumin (CUR) are missed because of its low solubility in aqueous media, low bioavailability, and structural lability upon oral intake. Soft nanoparticles such as nanoliposomes are not efficient as CUR carriers, since crystalline CUR is expelled from them to physiological media. Nanostructures to efficiently trap and increase the aqueous solubility of CUR are needed to improve both oral or nebulized delivery of CUR. Here we showed that SRA1 targeted nanoarchaeosomes (nATC) [1:0.4 w:w:0.04] archaeolipids, tween 80 and CUR, 155 ± 16 nm sized of −20.7 ± 3.3 z potential, retained 0.22 mg CUR ± 0.09 per 12.9 mg lipids ± 4.0 (~600 μM CUR) in front to dilution, storage, and nebulization. Raman and fluorescence spectra and SAXS patterns were compatible with a mixture of enol and keto CUR tautomers trapped within the depths of nATC bilayer. Between 20 and 5 µg CUR/mL, nATC was endocytosed by THP1 and A549 liquid–liquid monolayers without noticeable cytotoxicity. Five micrograms of CUR/mL nATC nebulized on an inflamed air–liquid interface of A549 cells increased TEER, normalized the permeation of LY, and decreased il6, tnfα, and il8 levels. Overall, these results suggest the modified pharmacodynamics of CUR in nATC is useful for epithelia repair upon inflammatory damage, deserving further deeper exploration, particularly related to its targeting ability.
Collapse
|
43
|
Dent P, Booth L, Roberts JL, Poklepovic A, Cridebring D, Reiman EM. Inhibition of heat shock proteins increases autophagosome formation, and reduces the expression of APP, Tau, SOD1 G93A and TDP-43. Aging (Albany NY) 2021; 13:17097-17117. [PMID: 34252884 PMCID: PMC8312464 DOI: 10.18632/aging.203297] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 07/02/2021] [Indexed: 12/13/2022]
Abstract
Aberrant expression and denaturation of Tau, amyloid-beta and TDP-43 can lead to cell death and is a major component of pathologies such as Alzheimer’s Disease (AD). AD neurons exhibit a reduced ability to form autophagosomes and degrade proteins via autophagy. Using genetically manipulated colon cancer cells we determined whether drugs that directly inhibit the chaperone ATPase activity or cause chaperone degradation and endoplasmic reticulum stress signaling leading to macroautophagy could reduce the levels of these proteins. The antiviral chaperone ATPase inhibitor AR12 reduced the ATPase activities and total expression of GRP78, HSP90, and HSP70, and of Tau, Tau 301L, APP, APP692, APP715, SOD1 G93A and TDP-43. In parallel, it increased the phosphorylation of ATG13 S318 and eIF2A S51 and caused eIF2A-dependent autophagosome formation and autophagic flux. Knock down of Beclin1 or ATG5 prevented chaperone, APP and Tau degradation. Neratinib, used to treat HER2+ breast cancer, reduced chaperone levels and expression of Tau and APP via macroautophagy, and neratinib interacted with AR12 to cause further reductions in protein levels. The autophagy-regulatory protein ATG16L1 is expressed as two isoforms, T300 or A300: Africans trend to express T300 and Europeans A300. We observed higher basal expression of Tau in T300 cells when compared to isogenic A300 cells. ATG16L1 isoform expression did not alter basal levels of HSP90, HSP70 or HSP27, however, basal levels of GRP78 were reduced in A300 cells. The abilities of both AR12 and neratinib to stimulate ATG13 S318 and eIF2A S51 phosphorylation and autophagic flux was also reduced in A300 cells. Our data support further evaluation of AR12 and neratinib in neuronal cells as repurposed treatments for AD.
Collapse
Affiliation(s)
- Paul Dent
- Department of Biochemistry and Molecular Biology, Richmond, VA 23298, USA
| | - Laurence Booth
- Department of Biochemistry and Molecular Biology, Richmond, VA 23298, USA
| | - Jane L Roberts
- Department of Pharmacology and Toxicology, Richmond, VA 23298, USA
| | - Andrew Poklepovic
- Department of Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Derek Cridebring
- Translational Genomics Research Institute (TGEN), Phoenix, AZ 85004, USA
| | - Eric M Reiman
- Translational Genomics Research Institute (TGEN), Phoenix, AZ 85004, USA.,Banner Alzheimer's Institute, Phoenix, AZ 85006, USA
| |
Collapse
|
44
|
Kocak M, Ezazi Erdi S, Jorba G, Maestro I, Farrés J, Kirkin V, Martinez A, Pless O. Targeting autophagy in disease: established and new strategies. Autophagy 2021; 18:473-495. [PMID: 34241570 PMCID: PMC9037468 DOI: 10.1080/15548627.2021.1936359] [Citation(s) in RCA: 108] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Macroautophagy/autophagy is an evolutionarily conserved pathway responsible for clearing cytosolic aggregated proteins, damaged organelles or invading microorganisms. Dysfunctional autophagy leads to pathological accumulation of the cargo, which has been linked to a range of human diseases, including neurodegenerative diseases, infectious and autoimmune diseases and various forms of cancer. Cumulative work in animal models, application of genetic tools and pharmacologically active compounds, has suggested the potential therapeutic value of autophagy modulation in disease, as diverse as Huntington, Salmonella infection, or pancreatic cancer. Autophagy activation versus inhibition strategies are being explored, while the role of autophagy in pathophysiology is being studied in parallel. However, the progress of preclinical and clinical development of autophagy modulators has been greatly hampered by the paucity of selective pharmacological agents and biomarkers to dissect their precise impact on various forms of autophagy and cellular responses. Here, we summarize established and new strategies in autophagy-related drug discovery and indicate a path toward establishing a more efficient discovery of autophagy-selective pharmacological agents. With this knowledge at hand, modern concepts for therapeutic exploitation of autophagy might become more plausible. Abbreviations: ALS: amyotrophic lateral sclerosis; AMPK: AMP-activated protein kinase; ATG: autophagy-related gene; AUTAC: autophagy-targeting chimera; CNS: central nervous system; CQ: chloroquine; GABARAP: gamma-aminobutyric acid type A receptor-associated protein; HCQ: hydroxychloroquine; LYTAC: lysosome targeting chimera; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MTOR: mechanistic target of rapamycin kinase; NDD: neurodegenerative disease; PDAC: pancreatic ductal adenocarcinoma; PE: phosphatidylethanolamine; PIK3C3/VPS34: phosphatidylinositol 3-kinase catalytic subunit type 3; PtdIns3K: class III phosphatidylinositol 3-kinase; PtdIns3P: phosphatidylinositol 3-phosphate; PROTAC: proteolysis-targeting chimera; SARS-CoV-2: severe acute respiratory syndrome coronavirus 2; SQSTM1/p62: sequestosome 1; ULK1: unc-51 like autophagy activating kinase 1.
Collapse
Affiliation(s)
- Muhammed Kocak
- Cancer Research UK, Cancer Therapeutics Unit, the Institute of Cancer Research London, Sutton, UK
| | | | | | - Inés Maestro
- Centro De Investigaciones Biologicas "Margarita Salas"-CSIC, Madrid, Spain
| | | | - Vladimir Kirkin
- Cancer Research UK, Cancer Therapeutics Unit, the Institute of Cancer Research London, Sutton, UK
| | - Ana Martinez
- Centro De Investigaciones Biologicas "Margarita Salas"-CSIC, Madrid, Spain.,Centro De Investigación Biomédica En Red En Enfermedades Neurodegenerativas (CIBERNED), Instituto De Salud Carlos III, Madrid, Spain
| | - Ole Pless
- Fraunhofer ITMP ScreeningPort, Hamburg, Germany
| |
Collapse
|
45
|
Schaefer L, Dikic I. Autophagy: Instructions from the extracellular matrix. Matrix Biol 2021; 100-101:1-8. [PMID: 34217800 DOI: 10.1016/j.matbio.2021.06.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 06/27/2021] [Accepted: 06/27/2021] [Indexed: 02/07/2023]
Abstract
In recent years, extensive research has uncovered crucial regulatory roles for the extracellular matrix (ECM) in regulating autophagy. Autophagy is a ubiquitous and highly conserved catabolic process that allows the selective removal and recycling of cytosolic components via lysosomal or vacuolar degradation. Due to its pivotal role in cellular homeostasis, the impairment of autophagy is involved in the pathophysiology of numerous diseases, comprising infectious diseases, immune and neurodegenerative disorders, renal and hepatic diseases, intervertebral and cartilage disorders, as well as fibrosis and cancer. Several ECM-derived proteoglycans and proteins, including decorin, biglycan, endorepellin, endostatin, collagen VI, and plasminogen kringle 5, have been identified as strong inducers of autophagy. In contrast, laminin α2, perlecan, and lumican exert opposite function by suppressing autophagy. Importantly, by direct interaction with various receptors, which interplay with their co-receptors and adhesion molecules, the ECM is able to direct autophagy in a molecular and cell context-specific manner. Thus, vast pharmacological potential resides in translating this knowledge into the development of ECM-derived therapeutics selectively regulating autophagy.
Collapse
Affiliation(s)
- Liliana Schaefer
- Institute of Pharmacology and Toxicology, Goethe University, Frankfurt, Germany.
| | - Ivan Dikic
- Institute of Biochemistry II, School of Medicine, Goethe University, Frankfurt, Germany; Buchmann Institute for Molecular Life Sciences, Goethe University, Frankfurt, Germany.
| |
Collapse
|
46
|
Auger C, Christou N, Brunel A, Perraud A, Verdier M. Autophagy and Extracellular Vesicles in Colorectal Cancer: Interactions and Common Actors? Cancers (Basel) 2021; 13:cancers13051039. [PMID: 33801266 PMCID: PMC7958126 DOI: 10.3390/cancers13051039] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 02/23/2021] [Accepted: 02/25/2021] [Indexed: 02/07/2023] Open
Abstract
Autophagy is a homeostatic process involved in the degradation of disabled proteins and organelles using lysosomes. This mechanism requires the recruitment of specialized proteins for vesicle trafficking, that may also be involved in other types of machinery such as the biogenesis and secretion of extracellular vesicles (EVs), and particularly small EVs called exosomes. Among these proteins, Rab-GTPases may operate in both pathways, thus representing an interesting avenue for further study regarding the interaction between autophagy and extracellular vesicle machinery. Both mechanisms are involved in the development of colorectal cancer (CRC), particularly in cancer stem cell (CSC) survival and communication, although they are not specific to CRC or CSCs. This highlights the importance of studying the crosstalk between autophagy and EVs biogenesis and release.
Collapse
Affiliation(s)
- Clément Auger
- EA 3842, CAPTuR, GEIST, Faculty of Medicine, University of Limoges, 2 rue du Dr Marcland, 87025 Limoges CEDEX, France; (C.A.); (A.B.); (A.P.); (M.V.)
| | - Niki Christou
- EA 3842, CAPTuR, GEIST, Faculty of Medicine, University of Limoges, 2 rue du Dr Marcland, 87025 Limoges CEDEX, France; (C.A.); (A.B.); (A.P.); (M.V.)
- Endocrine, General and Digestive Surgery Department, Limoges University Hospital, 2 rue Martin Luther King, 87042 Limoges CEDEX, France
- Correspondence: ; Tel.: +33-36-8456-9392
| | - Aude Brunel
- EA 3842, CAPTuR, GEIST, Faculty of Medicine, University of Limoges, 2 rue du Dr Marcland, 87025 Limoges CEDEX, France; (C.A.); (A.B.); (A.P.); (M.V.)
| | - Aurélie Perraud
- EA 3842, CAPTuR, GEIST, Faculty of Medicine, University of Limoges, 2 rue du Dr Marcland, 87025 Limoges CEDEX, France; (C.A.); (A.B.); (A.P.); (M.V.)
- Endocrine, General and Digestive Surgery Department, Limoges University Hospital, 2 rue Martin Luther King, 87042 Limoges CEDEX, France
| | - Mireille Verdier
- EA 3842, CAPTuR, GEIST, Faculty of Medicine, University of Limoges, 2 rue du Dr Marcland, 87025 Limoges CEDEX, France; (C.A.); (A.B.); (A.P.); (M.V.)
| |
Collapse
|