1
|
Zheng Q, Wang D, Lin R, Xu W. Pyroptosis, ferroptosis, and autophagy in spinal cord injury: regulatory mechanisms and therapeutic targets. Neural Regen Res 2025; 20:2787-2806. [PMID: 39101602 PMCID: PMC11826477 DOI: 10.4103/nrr.nrr-d-24-00112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 04/24/2024] [Accepted: 06/07/2024] [Indexed: 08/06/2024] Open
Abstract
Regulated cell death is a form of cell death that is actively controlled by biomolecules. Several studies have shown that regulated cell death plays a key role after spinal cord injury. Pyroptosis and ferroptosis are newly discovered types of regulated cell deaths that have been shown to exacerbate inflammation and lead to cell death in damaged spinal cords. Autophagy, a complex form of cell death that is interconnected with various regulated cell death mechanisms, has garnered significant attention in the study of spinal cord injury. This injury triggers not only cell death but also cellular survival responses. Multiple signaling pathways play pivotal roles in influencing the processes of both deterioration and repair in spinal cord injury by regulating pyroptosis, ferroptosis, and autophagy. Therefore, this review aims to comprehensively examine the mechanisms underlying regulated cell deaths, the signaling pathways that modulate these mechanisms, and the potential therapeutic targets for spinal cord injury. Our analysis suggests that targeting the common regulatory signaling pathways of different regulated cell deaths could be a promising strategy to promote cell survival and enhance the repair of spinal cord injury. Moreover, a holistic approach that incorporates multiple regulated cell deaths and their regulatory pathways presents a promising multi-target therapeutic strategy for the management of spinal cord injury.
Collapse
Affiliation(s)
- Qingcong Zheng
- Department of Spinal Surgery, the First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian Province, China
| | - Du Wang
- Arthritis Clinical and Research Center, Peking University People’s Hospital, Beijing, China
| | - Rongjie Lin
- Department of Orthopedic Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
| | - Weihong Xu
- Department of Spinal Surgery, the First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian Province, China
| |
Collapse
|
2
|
Liu C, Wang X, Xu S, Liu M, Cao X. Regulation of autophagy: Insights into O-GlcNAc modification mechanisms. Life Sci 2025; 369:123547. [PMID: 40058573 DOI: 10.1016/j.lfs.2025.123547] [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: 12/28/2024] [Revised: 02/27/2025] [Accepted: 03/06/2025] [Indexed: 03/30/2025]
Abstract
Autophagy is a "self-eating" biological process that degrades cytoplasmic contents to ensure cellular homeostasis. Its response to stimuli occurs in two stages: Within a few to several hours of exposure to a stress condition, autophagic flow rapidly increases, which is mediated by post-translational modification (PTM). Subsequently, the transcriptional program is activated and mediates the persistent autophagic response. O-linked β-N-acetylglucosamine (O-GlcNAc) modification is an inducible and dynamically cycling PTM; mounting evidence suggests that O-GlcNAc modification participates in the total autophagic process, including autophagy initiation, autophagosome formation, autophagosome-lysosome fusion, and transcriptional process. In this review, we summarize the current knowledge on the emerging role of O-GlcNAc modification in regulating autophagy-associated proteins and explain the different regulatory effects on autophagy exerted by O-GlcNAc modification.
Collapse
Affiliation(s)
- Chengzhi Liu
- Beijing Ophthalmology & Visual Science Key Lab, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China; The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Xinyu Wang
- Beijing Ophthalmology & Visual Science Key Lab, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Shengnan Xu
- College of Basic Medicine, Dalian Medical University, Dalian 116044, China
| | - Mingyue Liu
- Beijing Ophthalmology & Visual Science Key Lab, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Xusheng Cao
- Beijing Ophthalmology & Visual Science Key Lab, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China.
| |
Collapse
|
3
|
Shams El Dine RS, Youseef HT, Awaad AK, Hammoury SI, Mohamed EI. The potentially therapeutic effects of ascorbic acid in different cell line in attempt to reduce the risk of radiation therapy. Sci Rep 2025; 15:15077. [PMID: 40301490 PMCID: PMC12041364 DOI: 10.1038/s41598-025-96697-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 03/31/2025] [Indexed: 05/01/2025] Open
Abstract
Leukemia is the most common type of serious, life-threatening cancer that requires the immediate initiation of therapy. Ascorbic acid (AsA), commonly known as Vitamin C, has been gaining attention due to its antioxidant activity as a potential treatment for human malignancies. In this study, the THP-1 monocytic cell line was treated with two doses of AsA: a low dose (L-AsA, 2.5 µg/mL) and a high dose (H-AsA, 5 µg/mL), while the K562 lymphocytic cell line was treated with two doses of AsA: a low dose (L-AsA, 4 µg/mL) and a high dose (H-AsA, 8 µg/mL). After a 24-h incubation period, all cells were exposed to different doses of X-radiation (2, 4, 8 Gy). The viability of THP-1 and K562 treated by AsA was assessed using the MTT assay. Additionally, we evaluated apoptosis, autophagy, proliferation, cell cycle progression, hypoxia-inducible factor (HIF-1), malondialdehyde (MDA), and total antioxidant capacity (TAC). Our study demonstrated that AsA, in combination with X-radiation, induced significant apoptosis and notably reduced Ki67 levels in human leukemia THP-1 cells. Furthermore, X-radiation caused DNA damage, leading to cell cycle arrest at the G0/G1 phase in THP-1 cells. Moreover, AsA significantly reduced HIF-1 levels, which are essential for the survival of tumor cells in hypoxic conditions. We also found that the administration of AsA in combination with X-radiation had a synergistic and dose-dependent effect on THP-1 and K562 cells. Notably, the combination of L-AsA with 2 Gy X-radiation showed a more pronounced effect than 8 Gy X-radiation alone. These results suggest that AsA has promising anti-proliferative, pro-apoptotic, and autophagic effects on leukemic cells. Furthermore, the dose of X-radiation may be reduced when combined with AsA in an effort to minimize its potential side effects.
Collapse
Affiliation(s)
- Rasha S Shams El Dine
- Medical Biophysics Department, Medical Research Institute, Alexandria University, Alexandria, Egypt.
- Biochemistry and Molecular Biology Department, Alexandria University, Alexandria, Egypt.
- Radiotherapy Department, Ayadi Al-Mostakbal Oncology Hospital, Alexandria, Egypt.
| | - Heba T Youseef
- Medical Biophysics Department, Medical Research Institute, Alexandria University, Alexandria, Egypt
| | - Ashraf K Awaad
- Biochemistry and Molecular Biology Department, Alexandria University, Alexandria, Egypt
| | - Sabahh I Hammoury
- Radiotherapy Department, Ayadi Al-Mostakbal Oncology Hospital, Alexandria, Egypt
| | - Ehab I Mohamed
- Medical Biophysics Department, Medical Research Institute, Alexandria University, Alexandria, Egypt
| |
Collapse
|
4
|
Dashti A, Hosseini HM, Mirhosseini SA. Epsilon toxin induces cytotoxicity by mediating autophagy and apoptosis via the PI3K/AKT/mTOR signaling pathway in A549 cells. Mol Biol Rep 2025; 52:403. [PMID: 40252125 DOI: 10.1007/s11033-025-10439-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 03/14/2025] [Indexed: 04/21/2025]
Abstract
BACKGROUND Epsilon toxin, which is synthesized by Clostridium perfringens, is a type of pore-forming protein that is associated with the development of enterotoxemia in ruminants. As toxins are agents of bioterrorism, exposure to toxin aerosols causes endothelial cell damage and cytotoxicity in human lung cells. However, little information is available regarding the cytotoxicity and mechanisms associated with lung cancer cell lines. The aim of the present study was to explore the cytotoxic effects of epsilon toxin on the human lung cell line A549 and its involvement in the PI3K/AKT/mTOR signaling pathway to clarify the underlying molecular mechanism involved. METHODS AND RESULTS A549 cells were treated with epsilon toxin, and cytotoxicity was assessed via MTT and LDH assays. Flow cytometry evaluated ROS levels, cell cycle arrest, and apoptosis, while Hoechst 33,258 staining confirmed apoptotic morphology. qRT‒PCR and Western blotting measured apoptosis-, autophagy-, and PI3K/AKT/mTOR-related markers. Epsilon toxin reduced cell viability and increased membrane leakage in a concentration-dependent manner, accompanied by ROS overproduction. It upregulated autophagy markers (beclin-1, LC3 II/I, p62) and suppressed PI3K/AKT/mTOR signaling. Cell cycle arrest at the sub-G1 phase and apoptosis were induced via p53 activation, Bax/Bcl-2 imbalance, and caspase-3 cleavage, as confirmed by annexin V/PI and Hoechst 33,258 staining. CONCLUSIONS Epsilon toxin triggers cytotoxicity in A549 cells by activating apoptosis and autophagy through PI3K/AKT/mTOR pathway inhibition. These findings elucidate molecular mechanisms underlying epsilon toxin's action in lung cancer cells, highlighting its dual role in programmed cell death and potential therapeutic relevance.
Collapse
Affiliation(s)
- Ayat Dashti
- Applied Microbiology Research Center, Biomedicine Technologies Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Hamideh Mahmoodzadeh Hosseini
- Applied Microbiology Research Center, Biomedicine Technologies Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Seyed Ali Mirhosseini
- Applied Microbiology Research Center, Biomedicine Technologies Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
5
|
Zou Y, Zhang X, Chen XY, Ma XF, Feng XY, Sun Y, Ma T, Ma QH, Zhao XD, Xu DE. Contactin -Associated protein1 Regulates Autophagy by Modulating the PI3K/AKT/mTOR Signaling Pathway and ATG4B Levels in Vitro and in Vivo. Mol Neurobiol 2025; 62:2764-2780. [PMID: 39164481 PMCID: PMC11790771 DOI: 10.1007/s12035-024-04425-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 08/06/2024] [Indexed: 08/22/2024]
Abstract
Contactin-associated protein1 (Caspr1) plays an important role in the formation and stability of myelinated axons. In Caspr1 mutant mice, autophagy-related structures accumulate in neurons, causing axonal degeneration; however, the mechanism by which Caspr1 regulates autophagy remains unknown. To illustrate the mechanism of Caspr1 in autophagy process, we demonstrated that Caspr1 knockout in primary neurons from mice along with human cell lines, HEK-293 and HeLa, induced autophagy by downregulating the PI3K/AKT/mTOR signaling pathway to promote the conversion of microtubule-associated protein light chain 3 I (LC3-I) to LC3-II. In contrast, Caspr1 overexpression in cells contributed to the upregulation of this signaling pathway. We also demonstrated that Caspr1 knockout led to increased LC3-I protein expression in mice. In addition, Caspr1 could inhibit the expression of autophagy-related 4B cysteine peptidase (ATG4B) protein by directly binding to ATG4B in overexpressed Caspr1 cells. Intriguingly, we found an accumulation of ATG4B in the Golgi apparatuses of cells overexpressing Caspr1; therefore, we speculate that Caspr1 may restrict ATG4 secretion from the Golgi apparatus to the cytoplasm. Collectively, our results indicate that Caspr1 may regulate autophagy by modulating the PI3K/AKT/mTOR signaling pathway and the levels of ATG4 protein, both in vitro and in vivo. Thus, Caspr1 can be a potential therapeutic target in axonal damage and demyelinating diseases.
Collapse
Affiliation(s)
- Yan Zou
- Department of Neurosurgery, Jiangnan University Medical Center, the Wuxi No.2 People Hospital, Wuxi, 214002, Jiangsu, China
| | - Xiao Zhang
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214002, Jiangsu, China
- Department of Neurology, Jiangnan University Medical Center, the Wuxi No.2 People Hospital, Wuxi, 214002, Jiangsu, China
| | - Xin-Yi Chen
- Department of Neurology, Jiangnan University Medical Center, the Wuxi No.2 People Hospital, Wuxi, 214002, Jiangsu, China
| | - Xiao-Fang Ma
- Hong Shan Hospital, Wuxi, 214000, Jiangsu, China
| | - Xiao-Yan Feng
- Department of Neurology, Jiangnan University Medical Center, the Wuxi No.2 People Hospital, Wuxi, 214002, Jiangsu, China
| | - Yang Sun
- Department of Neurology, Jiangnan University Medical Center, the Wuxi No.2 People Hospital, Wuxi, 214002, Jiangsu, China
| | - Tao Ma
- Department of Neurology, Jiangnan University Medical Center, the Wuxi No.2 People Hospital, Wuxi, 214002, Jiangsu, China
| | - Quan-Hong Ma
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Institute of Neuroscience, Soochow University, Suzhou, 215004, Jiangsu, China
| | - Xu-Dong Zhao
- Department of Neurosurgery, Jiangnan University Medical Center, the Wuxi No.2 People Hospital, Wuxi, 214002, Jiangsu, China.
- Wuxi Neurosurgical Institute, Wuxi, 214122, Jiangsu, China.
| | - De-En Xu
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214002, Jiangsu, China.
- Department of Neurology, Jiangnan University Medical Center, the Wuxi No.2 People Hospital, Wuxi, 214002, Jiangsu, China.
| |
Collapse
|
6
|
Gil J, Kim D, Choi S, Bae ON. Cadmium-induced iron dysregulation contributes to functional impairment in brain endothelial cells via the ferroptosis pathway. Toxicol Appl Pharmacol 2025; 495:117233. [PMID: 39842614 DOI: 10.1016/j.taap.2025.117233] [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: 10/04/2024] [Revised: 01/12/2025] [Accepted: 01/15/2025] [Indexed: 01/24/2025]
Abstract
Cadmium (Cd2+) is a heavy metal that is a major hazardous environmental contaminant, ubiquitously present in the environment. Cd2+ exposure has been closely associated with an increased prevalence and severity of neurological and cardiovascular diseases (CVD). The blood-brain barrier (BBB) plays a crucial role in protecting the brain from external environmental factors. Mitochondria play an important role in maintaining the barrier function of brain endothelial cells by regulating energy metabolism and redox homeostasis. In this study, we aimed to assess the cytotoxic effects of Cd2+ on the integrity and function of brain endothelial cells. After 24 h of exposure, Cd2+ reduced cell survival, tight junction protein expression, and trans-endothelial electrical resistance (TEER) in bEnd.3 cells suggest a potential BBB integrity disruption by Cd2+ exposure. To clarify the underlying mechanism, we further investigated the role of mitochondria in iron overload-mediated cell death following Cd2+ exposure. Cd2+ induced a substantial reduction in mitochondrial basal respiration and ATP production in brain endothelial cells, suggesting mitochondrial dysfunction. In addition, Cd2+ exposure led to impaired autophagy, elevated iron levels, and increased lipid peroxidation, indicating the initiation of ferroptosis, a form of cell death triggered by iron. In summary, our research suggests that Cd2+ exposure can disrupt BBB function by causing mitochondrial dysfunction and disrupting iron homeostasis.
Collapse
Affiliation(s)
- Junkyung Gil
- College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University ERICA Campus, Ansan, South Korea.
| | - Donghyun Kim
- College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University ERICA Campus, Ansan, South Korea.
| | - Sungbin Choi
- College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University ERICA Campus, Ansan, South Korea.
| | - Ok-Nam Bae
- College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University ERICA Campus, Ansan, South Korea.
| |
Collapse
|
7
|
Zeng C, Guo M, Xiao K, Li C. Autophagy mediated by ROS-AKT-FoxO pathway is required for intestinal regeneration in echinoderms. Cell Commun Signal 2025; 23:8. [PMID: 39762855 PMCID: PMC11705696 DOI: 10.1186/s12964-024-01993-0] [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: 05/09/2024] [Accepted: 12/11/2024] [Indexed: 01/11/2025] Open
Abstract
Autophagy is essential for maintaining material balance and energy circulation and plays a critical role as a regulatory mechanism in tissue regeneration. However, current studies primarily describe this phenotype, with limited exploration of its molecular mechanisms. In this study, we provided the first evidence that autophagy is required for intestinal regeneration in Apostichopus japonicus and identified a previously unrecognized regulatory mechanism involved in this process. We observed that autophagy activation was significantly associated with enhanced regeneration, and its upregulation was shown to be regulated by reactive oxygen species (ROS) bursts. Mechanistically, ROS induced the dephosphorylation of Forkhead box protein O (FoxO) through AjAKT dephosphorylation. The dephosphorylated AjFoxO translocated to the nucleus, where it bound to the promoters of AjLC3 and AjATG4, inducing their transcription. This study highlights the ROS-AjAKT-AjFoxO-AjATG4/AjLC3 pathway as a novel regulatory mechanism underlying autophagy-mediated intestinal regeneration in echinoderms, providing a reference for studying regenerative processes and cytological mechanisms in economically important echinoderms.
Collapse
Affiliation(s)
- Chuili Zeng
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, China
| | - Ming Guo
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, China
| | - Ke Xiao
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, China
| | - Chenghua Li
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, China.
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266071, PR China.
- Ningbo University, Zhejiang Province, Ningbo, 315211, P. R. China.
| |
Collapse
|
8
|
Li D, Geng D, Wang M. Advances in natural products modulating autophagy influenced by cellular stress conditions and their anticancer roles in the treatment of ovarian cancer. FASEB J 2024; 38:e70075. [PMID: 39382031 DOI: 10.1096/fj.202401409r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 08/20/2024] [Accepted: 09/13/2024] [Indexed: 10/10/2024]
Abstract
Autophagy is a conservative catabolic process that typically serves a cell-protective function. Under stress conditions, when the cellular environment becomes unstable, autophagy is activated as an adaptive response for self-protection. Autophagy delivers damaged cellular components to lysosomes for degradation and recycling, thereby providing essential nutrients for cell survival. However, this function of promoting cell survival under stress conditions often leads to malignant progression and chemotherapy resistance in cancer. Consequently, autophagy is considered a potential target for cancer therapy. Herein, we aim to review how natural products act as key modulators of autophagy by regulating cellular stress conditions. We revisit various stressors, including starvation, hypoxia, endoplasmic reticulum stress, and oxidative stress, and their regulatory relationship with autophagy, focusing on recent advances in ovarian cancer research. Additionally, we explore how polyphenolic compounds, flavonoids, alkaloids, terpenoids, and other natural products modulate autophagy mediated by stress responses, affecting the malignant biological behavior of cancer. Furthermore, we discuss their roles in ovarian cancer therapy. This review emphasizes the importance of natural products as valuable resources in cancer therapeutics, highlighting the need for further exploration of their potential in regulating autophagy. Moreover, it provides novel insights and potential therapeutic strategies in ovarian cancer by utilizing natural products to modulate autophagy.
Collapse
Affiliation(s)
- Dongxiao Li
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Danbo Geng
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Min Wang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
9
|
Manabe S, Haga Y, Tsujino H, Ikuno Y, Asahara H, Higashisaka K, Tsutsumi Y. Treatment of polyethylene microplastics degraded by ultraviolet light irradiation causes lysosome-deregulated cell death. Sci Rep 2024; 14:24008. [PMID: 39402130 PMCID: PMC11473831 DOI: 10.1038/s41598-024-74800-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 09/30/2024] [Indexed: 10/17/2024] Open
Abstract
BACKGROUND Microplastics (MPs), plastic particles < 5 mm in size, are prevalent in the environment, and human exposure to them is inevitable. To assess the potential risk of MPs on human health, it is essential to consider the physicochemical properties of environmental MPs, including polymer types, size, shape, and surface chemical modifications. Notably, environmental MPs undergo degradation due to external factors such as ultraviolet (UV) rays and waves, leading to changes in their surface characteristics. However, limited knowledge exists regarding the health effects of MPs, with a specific focus on their surface degradation. This study concentrates on cytotoxic MPs with surface degradation through UV irradiation, aiming to identify the mechanisms underlying their cell toxicity. RESULTS Polyethylene (PE) and surface-degraded PE achieved through UV light irradiation were employed as model MPs in this study. We explored the impact of PE and degraded PE on cell death in murine macrophage cell line RAW264.7 cells and human monocyte cell line THP-1 cells. Flow cytometric analysis revealed that degraded PE induced programmed cell death without activating caspase 3, while non-degraded PE did not trigger programmed cell death. These findings suggest that degraded PE might induce programmed cell death through mechanisms other than caspase-driven apoptosis. To understand the mechanisms of cell death, we investigated how cells responded to degraded PE-induced cellular stress. Immunofluorescence and western blotting analyses demonstrated that degraded PE induced autophagosome formation and increased p62 expression, indicating inhibited autophagy flux after exposure to degraded PE. Furthermore, degraded PE exposure led to a decrease in acidic lysosomes, indicating lysosomal dysregulation. These results imply that degraded PE induces lysosomal dysfunction, subsequently causing autophagy dysregulation and cell death. CONCLUSIONS This study unveils that UV-induced degradation of PE results in cell death attributed to lysosomal dysfunction. The findings presented herein provide novel insights into the effects of surface-degraded MPs on biological responses.
Collapse
Affiliation(s)
- Sota Manabe
- School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yuya Haga
- Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Hirofumi Tsujino
- Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan
- Museum Links, Osaka University, 1-13 Machikaneyama, Toyonaka, Osaka, 560-0043, Japan
| | - Yudai Ikuno
- Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Haruyasu Asahara
- Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan
- Institute for Open and Transdisciplinary Research Initiatives, Osaka University, 1-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Kazuma Higashisaka
- Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan
- Institute for Advanced Co-Creation Studies, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yasuo Tsutsumi
- Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan.
- Institute for Open and Transdisciplinary Research Initiatives, Osaka University, 1-1 Yamadaoka, Suita, Osaka, 565-0871, Japan.
- Global Center for Medical Engineering and Informatics, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
10
|
Bhutta ZA, Go RE, Choi KC. Effect of punicalagin on the autophagic cell death in triple-negative breast cancer cells. Toxicol Res 2024; 40:585-598. [PMID: 39345747 PMCID: PMC11436590 DOI: 10.1007/s43188-024-00246-z] [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: 01/25/2024] [Revised: 04/28/2024] [Accepted: 05/01/2024] [Indexed: 10/01/2024] Open
Abstract
Triple-negative breast cancer (TNBC) is a highly heterogeneous disease defined by the absence of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER-2), resulting in poor clinical outcomes and high mortality. The present study was aimed to evaluate the efficacy of Punicalagin (PCG), a polyphenol obtained from the Punica granatum, against TNBC. We evaluated the therapeutic potential of PCG in TNBC (MDA-MB-231, BT-20) and ER + (MCF-7) breast cancer cells. A dose-dependent inhibition of MDA-MB-231 cell proliferation was observed with PCG (12.5-100 μM). However, only 50 and 100 μM doses of PCG inhibited the growth of BT-20 and MCF-7 cells. PCG significantly increased mitochondrial ROS in TNBC cells and induced autophagy across all cell lines, as evidenced by an increase in autophagic vacuoles and a decrease in the ratio of LC3-II/LC3-I. PCG suppressed PI3K/Akt and activated phosphorylated c-Jun N-terminal kinase (p-JNK) signaling. Based on these findings, it can be concluded that PCG is capable of significantly inhibiting the proliferation of TNBC cells through the suppression of the PI3K/Akt pathway as well as the initiation of the JNK pathway. PCG could thus be potentially useful as a therapeutic agent for the treatment of TNBC. Graphical abstract
Collapse
Affiliation(s)
- Zeeshan Ahmad Bhutta
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 28644 Republic of Korea
| | - Ryeo-Eun Go
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 28644 Republic of Korea
| | - Kyung-Chul Choi
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 28644 Republic of Korea
| |
Collapse
|
11
|
Torres P, Rico-Rios S, Ceron-Codorniu M, Santacreu-Vilaseca M, Seoane-Miraz D, Jad Y, Ayala V, Mariño G, Beltran M, Miralles MP, Andrés-Benito P, Fernandez-Irigoyen J, Santamaria E, López-Otín C, Soler RM, Povedano M, Ferrer I, Pamplona R, Wood MJA, Varela MA, Portero-Otin M. TDP-43 regulates LC3ylation in neural tissue through ATG4B cryptic splicing inhibition. Acta Neuropathol 2024; 148:45. [PMID: 39305312 PMCID: PMC11416411 DOI: 10.1007/s00401-024-02780-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 08/01/2024] [Accepted: 08/02/2024] [Indexed: 09/25/2024]
Abstract
Amyotrophic lateral sclerosis (ALS) is an adult-onset motor neuron disease with a mean survival time of three years. The 97% of the cases have TDP-43 nuclear depletion and cytoplasmic aggregation in motor neurons. TDP-43 prevents non-conserved cryptic exon splicing in certain genes, maintaining transcript stability, including ATG4B, which is crucial for autophagosome maturation and Microtubule-associated proteins 1A/1B light chain 3B (LC3B) homeostasis. In ALS mice (G93A), Atg4b depletion worsens survival rates and autophagy function. For the first time, we observed an elevation of LC3ylation in the CNS of both ALS patients and atg4b-/- mouse spinal cords. Furthermore, LC3ylation modulates the distribution of ATG3 across membrane compartments. Antisense oligonucleotides (ASOs) targeting cryptic exon restore ATG4B mRNA in TARDBP knockdown cells. We further developed multi-target ASOs targeting TDP-43 binding sequences for a broader effect. Importantly, our ASO based in peptide-PMO conjugates show brain distribution post-IV administration, offering a non-invasive ASO-based treatment avenue for neurodegenerative diseases.
Collapse
Affiliation(s)
- Pascual Torres
- Metabolic Pathophysiology Research Group, Department of Experimental Medicine, University of Lleida (UdL), Lleida Biomedical Research Institute (IRBLleida), 25198, Lleida, Spain
| | - Santiago Rico-Rios
- Metabolic Pathophysiology Research Group, Department of Experimental Medicine, University of Lleida (UdL), Lleida Biomedical Research Institute (IRBLleida), 25198, Lleida, Spain
| | - Miriam Ceron-Codorniu
- Metabolic Pathophysiology Research Group, Department of Experimental Medicine, University of Lleida (UdL), Lleida Biomedical Research Institute (IRBLleida), 25198, Lleida, Spain
| | - Marta Santacreu-Vilaseca
- Metabolic Pathophysiology Research Group, Department of Experimental Medicine, University of Lleida (UdL), Lleida Biomedical Research Institute (IRBLleida), 25198, Lleida, Spain
| | - David Seoane-Miraz
- Department of Paediatrics, Institute of Developmental and Regenerative Medicine (IDRM), University of Oxford, Roosevelt Dr, Oxford, OX3 7TY, UK
- MDUK Oxford Neuromuscular Centre, University of Oxford, Oxford, UK
| | - Yahya Jad
- Department of Paediatrics, Institute of Developmental and Regenerative Medicine (IDRM), University of Oxford, Roosevelt Dr, Oxford, OX3 7TY, UK
- MDUK Oxford Neuromuscular Centre, University of Oxford, Oxford, UK
| | - Victòria Ayala
- Metabolic Pathophysiology Research Group, Department of Experimental Medicine, University of Lleida (UdL), Lleida Biomedical Research Institute (IRBLleida), 25198, Lleida, Spain
| | - Guillermo Mariño
- Departamento de Biología Funcional, Facultad de Medicina, Universidad de Oviedo, 33006, Oviedo, Spain
- Instituto Universitario de Oncología (IUOPA), 33006, Oviedo, Spain
- Instituto de Investigación Sanitaria Del Principado de Asturias (ISPA), 33011, Oviedo, Spain
| | - Maria Beltran
- Neuronal Signaling Unit, Department of Experimental Medicine, Lleida Biomedical Research Institute (IRBLleida), University of Lleida (UdL), 25198, Lleida, Spain
| | - Maria P Miralles
- Neuronal Signaling Unit, Department of Experimental Medicine, Lleida Biomedical Research Institute (IRBLleida), University of Lleida (UdL), 25198, Lleida, Spain
| | - Pol Andrés-Benito
- Neurologic Diseases and Neurogenetics Group, Bellvitge Institute for Biomedical Research (IDIBELL), 08907, L'Hospitalet de Llobregat, Spain
- CIBERNED (Network Centre of Biomedical Research of Neurodegenerative Diseases), Institute of Health Carlos III, 08907, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Joaquin Fernandez-Irigoyen
- Clinical Neuroproteomics Unit, Proteomics Platform, Proteored-ISCIII, Navarrabiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), diSNA, 31008, Pamplona, Spain
| | - Enrique Santamaria
- Clinical Neuroproteomics Unit, Proteomics Platform, Proteored-ISCIII, Navarrabiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), diSNA, 31008, Pamplona, Spain
| | - Carlos López-Otín
- Departamento de Biología Funcional, Facultad de Medicina, Universidad de Oviedo, 33006, Oviedo, Spain
- Instituto Universitario de Oncología (IUOPA), 33006, Oviedo, Spain
- Departamento de Bioquímica y Biología Molecular, Universidad de Oviedo, 33006, Oviedo, Spain
| | - Rosa M Soler
- Neuronal Signaling Unit, Department of Experimental Medicine, Lleida Biomedical Research Institute (IRBLleida), University of Lleida (UdL), 25198, Lleida, Spain
| | - Monica Povedano
- Neurologic Diseases and Neurogenetics Group, Bellvitge Institute for Biomedical Research (IDIBELL), 08907, L'Hospitalet de Llobregat, Spain
- CIBERNED (Network Centre of Biomedical Research of Neurodegenerative Diseases), Institute of Health Carlos III, 08907, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Isidro Ferrer
- CIBERNED (Network Centre of Biomedical Research of Neurodegenerative Diseases), Institute of Health Carlos III, 08907, L'Hospitalet de Llobregat, Barcelona, Spain
- Neuropathology Group, Institute of Biomedical Research, IDIBELL, 08907, L'Hospitalet de Llobregat, Spain
- Department of Pathology and Experimental Therapeutics, University of Barcelona, 08007, Barcelona, Spain
| | - Reinald Pamplona
- Metabolic Pathophysiology Research Group, Department of Experimental Medicine, University of Lleida (UdL), Lleida Biomedical Research Institute (IRBLleida), 25198, Lleida, Spain
| | - Matthew J A Wood
- Department of Paediatrics, Institute of Developmental and Regenerative Medicine (IDRM), University of Oxford, Roosevelt Dr, Oxford, OX3 7TY, UK
- MDUK Oxford Neuromuscular Centre, University of Oxford, Oxford, UK
| | - Miguel A Varela
- Department of Paediatrics, Institute of Developmental and Regenerative Medicine (IDRM), University of Oxford, Roosevelt Dr, Oxford, OX3 7TY, UK.
- MDUK Oxford Neuromuscular Centre, University of Oxford, Oxford, UK.
| | - Manuel Portero-Otin
- Metabolic Pathophysiology Research Group, Department of Experimental Medicine, University of Lleida (UdL), Lleida Biomedical Research Institute (IRBLleida), 25198, Lleida, Spain.
| |
Collapse
|
12
|
Zhang B, Li Z, Ye G, Hu K. Biologic activity and treatment resistance to gastrointestinal cancer: the role of circular RNA in autophagy regulation. Front Oncol 2024; 14:1393670. [PMID: 39281375 PMCID: PMC11392687 DOI: 10.3389/fonc.2024.1393670] [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: 02/29/2024] [Accepted: 08/15/2024] [Indexed: 09/18/2024] Open
Abstract
Circular RNAs (circRNAs) lack the 5'-end methylated guanine cap structure and 3' polyadenylate tail structure, classifying it as a non-coding RNA. With the extensive investigation of circRNA, its role in regulating cell death has garnered significant attention in recent years, establishing it as a recognized participant in cancer's biological processes. Autophagy, an essential pathway in programmed cell death (PCD), involves the formation of autophagosomes using lysosomes to degrade cellular contents under the regulation of various autophagy-related (ATG) genes. Numerous studies have demonstrated that circRNA can modulate the biological activity of cancer cells by influencing the autophagy pathway, exhibiting a dualistic role in suppressing or promoting carcinogenesis. In this review, we comprehensively analyze how autophagy-related circRNA impacts the progression of gastrointestinal cancer (GIC). Additionally, we discuss drug resistance phenomena associated with autophagy regulation in GIC. This review offers valuable insights into exploring potential biological targets for prognosis and treatment strategies related to GIC.
Collapse
Affiliation(s)
- Bo Zhang
- Health Science Center, Ningbo University, Ningbo, China
- Department of Gastroenterology, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Zhe Li
- Department of Gastroenterology, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Guoliang Ye
- Department of Gastroenterology, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Kefeng Hu
- Department of Gastroenterology, The First Affiliated Hospital of Ningbo University, Ningbo, China
| |
Collapse
|
13
|
Szoka L, Stocki M, Isidorov V. Dammarane-Type 3,4- seco-Triterpenoid from Silver Birch ( Betula pendula Roth) Buds Induces Melanoma Cell Death by Promotion of Apoptosis and Autophagy. Molecules 2024; 29:4091. [PMID: 39274939 PMCID: PMC11397366 DOI: 10.3390/molecules29174091] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 08/21/2024] [Accepted: 08/27/2024] [Indexed: 09/16/2024] Open
Abstract
Despite unquestionable advances in therapy, melanoma is still characterized by a high mortality rate. For years, high expectations have been raised by compounds of natural origin as a component of pharmacotherapy, particularly by triterpenes found in the bark of birch trees. In this study, 3,4-seco-dammara-4(29),20(21),24(25)-trien-3-oic acid (SDT) was isolated from buds of silver birch and its mechanisms of cell death induction, including apoptosis and autophagy, were determined. Cytotoxicity of SDT was evaluated by the cell viability test and clonogenic assay, whereas induction of apoptosis and autophagy was determined by annexin V staining and Western blot. The results revealed dose- and time-dependent reductions in viability of melanoma cells. Treatment of cells for 48 h led to an increase in the percentage of annexin V-positive cells, activation of caspase-8, caspase-9, and caspase-3, and cleavage of PARP, confirming apoptosis. Simultaneously, it was found that SDT increased the level of autophagy marker LC3-II and initiator of autophagy beclin-1. Pretreatment of cells with caspase-3 inhibitor or autophagy inhibitor significantly reduced the cytotoxicity of SDT and revealed that both apoptosis and autophagy contribute to a decrease in cell viability. These findings suggest that 3,4-seco-dammaranes may become a promising group of natural compounds for searching for anti-melanoma agents.
Collapse
Affiliation(s)
- Lukasz Szoka
- Department of Medicinal Chemistry, Medical University of Bialystok, 15-222 Białystok, Poland
| | - Marcin Stocki
- Institute of Forest Sciences, Białystok University of Technology, 15-351 Białystok, Poland
| | - Valery Isidorov
- Institute of Forest Sciences, Białystok University of Technology, 15-351 Białystok, Poland
| |
Collapse
|
14
|
Wen Y, Zhan Y, Chen T, Li J, Long Q, Zheng F, Tang S, Tang X. Total Flavonoids of Aurantii Fructus Immaturus Regulate miR-5100 to Improve Constipation by Targeting Fzd2 to Alleviate Calcium Balance and Autophagy in Interstitial Cells of Cajal. Mol Neurobiol 2024; 61:5882-5900. [PMID: 38244148 DOI: 10.1007/s12035-024-03958-3] [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: 10/23/2023] [Accepted: 01/12/2024] [Indexed: 01/22/2024]
Abstract
Aurantii Fructus Immaturus total flavonoids (AFIF) is the main effective fraction extracted from AFI, which has a good effect on promoting gastrointestinal motility. This study aimed to investigate AFIF which regulates miR-5100 to improve constipation symptoms in mice by targeting Frizzled-2 (Fzd2) to alleviate interstitial cells of Cajal (ICCs) calcium ion balance and autophagy apoptosis. The constipated mouse model was induced by an antibiotic suspension, and then treated with AFIF. RNA-seq sequencing, luciferase assay, immunofluorescence staining, transmission electron microscopy, ELISA, flow cytometry, quantitative polymerase chain reaction (PCR), and Western blot were applied in this study. The results showed that AFIF improved constipation symptoms in antibiotic-induced constipated mice, and decreased the autophagy-related protein Beclin1 levels and the LC3-II/I ratio in ICCs. miR-5100 and its target gene Fzd2 were screened as key miRNAs and regulator associated with autophagy. Downregulation of miR-5100 caused increased expression of Fzd2, decreased proliferation activity of ICCs, increased apoptotic cells, and enhanced calcium ion release and autophagy signals. After AFIF treatment, miR-5100 expression was upregulated and Fzd2 was downregulated, while autophagy-related protein levels and calcium ion concentration decreased. Furthermore, AFIF increased the levels of SP, 5-HT, and VIP, and increased the expression of PGP9.5, Sy, and Cx43, which alleviated constipation by improving the integrity of the enteric nervous system network. In conclusion, AFIF could attenuate constipation symptoms by upregulating the expression of miR-5100 and targeting inhibition of Fzd2, alleviating calcium overload and autophagic death of ICCs, regulating the content of neurotransmitters, and enhancing the integrity of the enteric nervous system network.
Collapse
Affiliation(s)
- Yong Wen
- Department of Traditional Chinese Medicine, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Yu Zhan
- Anorectal Department, Affiliated Hospital of Integrative Chinese Medicine and Western Medicine of Chengdu University of TCM, Chengdu, 610042, China
- Chengdu First People's Hospital, Chengdu, 610042, China
| | - Taiyu Chen
- Department of Integrated Traditional and Western Medicine Anorectal, Affiliated Hospital of North Sichuan Medical College, No. 1 Maoyuan South Road, Nanchong, Sichuan, China
| | - Jun Li
- Department of Traditional Chinese Medicine, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Qing Long
- Department of Traditional Chinese Medicine, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Fan Zheng
- People's Hospital of Deyang City, Deyang, China
- Universiti Sains Malaysia, Pulau Pinang, Malaysia
| | - Shiyu Tang
- North Sichuan Medical College, Nanchong, 637000, China
| | - Xuegui Tang
- Department of Integrated Traditional and Western Medicine Anorectal, Affiliated Hospital of North Sichuan Medical College, No. 1 Maoyuan South Road, Nanchong, Sichuan, China.
| |
Collapse
|
15
|
Elias MG, Aputen AD, Fatima S, Mann TJ, Karan S, Mikhael M, de Souza P, Gordon CP, Scott KF, Aldrich-Wright JR. Chemotherapeutic Potential of Chlorambucil-Platinum(IV) Prodrugs against Cisplatin-Resistant Colorectal Cancer Cells. Int J Mol Sci 2024; 25:8252. [PMID: 39125821 PMCID: PMC11312340 DOI: 10.3390/ijms25158252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 07/23/2024] [Accepted: 07/25/2024] [Indexed: 08/12/2024] Open
Abstract
Chlorambucil-platinum(IV) prodrugs exhibit multi-mechanistic chemotherapeutic activity with promising anticancer potential. The platinum(II) precursors of the prodrugs have been previously found to induce changes in the microtubule cytoskeleton, specifically actin and tubulin of HT29 colon cells, while chlorambucil alkylates the DNA. These prodrugs demonstrate significant anticancer activity in 2D cell and 3D spheroid viability assays. A notable production of reactive oxygen species has been observed in HT29 cells 72 h post treatment with prodrugs of this type, while the mitochondrial membrane potential was substantially reduced. The cellular uptake of the chlorambucil-platinum(IV) prodrugs, assessed by ICP-MS, confirmed that active transport was the primary uptake mechanism, with platinum localisation identified primarily in the cytoskeletal fraction. Apoptosis and necrosis were observed at 72 h of treatment as demonstrated by Annexin V-FITC/PI assay using flow cytometry. Immunofluorescence measured via confocal microscopy showed significant changes in actin and tubulin intensity and in architecture. Western blot analysis of intrinsic and extrinsic pathway apoptotic markers, microtubule cytoskeleton markers, cell proliferation markers, as well as autophagy markers were studied post 72 h of treatment. The proteomic profile was also studied with a total of 1859 HT29 proteins quantified by mass spectroscopy, with several dysregulated proteins. Network analysis revealed dysregulation in transcription, MAPK markers, microtubule-associated proteins and mitochondrial transport dysfunction. This study confirms that chlorambucil-platinum(IV) prodrugs are candidates with promising anticancer potential that act as multi-mechanistic chemotherapeutics.
Collapse
Affiliation(s)
- Maria George Elias
- School of Science, Western Sydney University, Sydney, NSW 2751, Australia; (M.G.E.); (A.D.A.); (S.K.); (C.P.G.)
- Medical Oncology, Ingham Institute for Applied Medical Research, Liverpool Hospital, Liverpool, NSW 2170, Australia; (S.F.); (T.J.M.); (K.F.S.)
| | - Angelico D. Aputen
- School of Science, Western Sydney University, Sydney, NSW 2751, Australia; (M.G.E.); (A.D.A.); (S.K.); (C.P.G.)
| | - Shadma Fatima
- Medical Oncology, Ingham Institute for Applied Medical Research, Liverpool Hospital, Liverpool, NSW 2170, Australia; (S.F.); (T.J.M.); (K.F.S.)
- School of Medicine, Western Sydney University, Sydney, NSW 2751, Australia
| | - Timothy J. Mann
- Medical Oncology, Ingham Institute for Applied Medical Research, Liverpool Hospital, Liverpool, NSW 2170, Australia; (S.F.); (T.J.M.); (K.F.S.)
- School of Medicine, Western Sydney University, Sydney, NSW 2751, Australia
| | - Shawan Karan
- School of Science, Western Sydney University, Sydney, NSW 2751, Australia; (M.G.E.); (A.D.A.); (S.K.); (C.P.G.)
| | - Meena Mikhael
- Mass Spectrometry Facility, Western Sydney University, Sydney, NSW 2751, Australia;
| | - Paul de Souza
- Nepean Clinical School, Faculty of Medicine and Health, University of Sydney, Kingswood, NSW 2747, Australia;
| | - Christopher P. Gordon
- School of Science, Western Sydney University, Sydney, NSW 2751, Australia; (M.G.E.); (A.D.A.); (S.K.); (C.P.G.)
| | - Kieran F. Scott
- Medical Oncology, Ingham Institute for Applied Medical Research, Liverpool Hospital, Liverpool, NSW 2170, Australia; (S.F.); (T.J.M.); (K.F.S.)
- School of Medicine, Western Sydney University, Sydney, NSW 2751, Australia
| | - Janice R. Aldrich-Wright
- School of Science, Western Sydney University, Sydney, NSW 2751, Australia; (M.G.E.); (A.D.A.); (S.K.); (C.P.G.)
- Medical Oncology, Ingham Institute for Applied Medical Research, Liverpool Hospital, Liverpool, NSW 2170, Australia; (S.F.); (T.J.M.); (K.F.S.)
| |
Collapse
|
16
|
Nucera F, Di Stefano A, Ricciardolo FLM, Gnemmi I, Pizzimenti C, Monaco F, Tuccari G, Caramori G, Ieni A. Role of ATG4 Autophagy-Related Protein Family in the Lower Airways of Patients with Stable COPD. Int J Mol Sci 2024; 25:8182. [PMID: 39125750 PMCID: PMC11311497 DOI: 10.3390/ijms25158182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 07/18/2024] [Accepted: 07/25/2024] [Indexed: 08/12/2024] Open
Abstract
Autophagy is a complex physiological pathway mediating homeostasis and survival of cells degrading damaged organelles and regulating their recycling. Physiologic autophagy can maintain normal lung function, decrease lung cellular senescence, and inhibit myofibroblast differentiation. It is well known that autophagy is activated in several chronic inflammatory diseases; however, its role in the pathogenesis of chronic obstructive pulmonary disease (COPD) and the expression of autophagy-related genes (ATGs) in lower airways of COPD patients is still controversial. The expression and localization of all ATG proteins that represented key components of the autophagic machinery modulating elongation, closure, and maturation of autophagosome membranes were retrospectively measured in peripheral lungs of patients with stable COPD (n = 10), control smokers with normal lung function (n = 10), and control nonsmoking subjects (n = 8) using immunohistochemical analysis. These results show an increased expression of ATG4 protein in alveolar septa and bronchiolar epithelium of stable COPD patients compared to smokers with normal lung function and non-smoker subjects. In particular, the genes in the ATG4 protein family (including ATG4A, ATG4B, ATG4C, and ATG4D) that have a key role in the modulation of the physiological autophagic machinery are the most important ATGs increased in the compartment of lower airways of stable COPD patients, suggesting that the alteration shown in COPD patients can be also correlated to impaired modulation of autophagic machinery modulating elongation, closure, and maturation of autophagosomes membranes. Statistical analysis was performed by the Kruskal-Wallis test and the Mann-Whitney U test for comparison between groups. A statistically significant increased expression of ATG4A (p = 0.0047), ATG4D (p = 0.018), and ATG5 (p = 0.019) was documented in the bronchiolar epithelium as well in alveolar lining for ATG4A (p = 0.0036), ATG4B (p = 0.0054), ATG4C (p = 0.0064), ATG4D (p = 0.0084), ATG5 (p = 0.0088), and ATG7 (p = 0.018) in patients with stable COPD compared to control groups. The ATG4 isoforms may be considered as additional potential targets for the development of new drugs in COPD.
Collapse
Affiliation(s)
- Francesco Nucera
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences, Section of Pneumology, University of Messina, 98125 Messina, Italy;
| | - Antonino Di Stefano
- Istituti Clinici Scientifici Maugeri, IRCCS, Respiratory Rehabilitation Unit of Gattico-Veruno, Section of Pneumology, Laboratory of Cytoimmunopathology in Cardio Respiratory System, 28013 Gattico-Veruno, Italy; (A.D.S.); (I.G.)
| | - Fabio Luigi Massimo Ricciardolo
- Department of Clinical and Biological Sciences, Severe Asthma, Rare Lung Disease and Respiratory Pathophysiology Unit, San Luigi Gonzaga University Hospital, University of Turin, 10043 Orbassano, Italy;
| | - Isabella Gnemmi
- Istituti Clinici Scientifici Maugeri, IRCCS, Respiratory Rehabilitation Unit of Gattico-Veruno, Section of Pneumology, Laboratory of Cytoimmunopathology in Cardio Respiratory System, 28013 Gattico-Veruno, Italy; (A.D.S.); (I.G.)
| | - Cristina Pizzimenti
- Department of Human Pathology in Adult and Developmental Age ‘Gaetano Barresi’, Section of Pathology, University of Messina, 98125 Messina, Italy; (C.P.); (G.T.)
| | - Francesco Monaco
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences, Section of Toracic Surgery, University of Messina, 98125 Messina, Italy;
| | - Giovanni Tuccari
- Department of Human Pathology in Adult and Developmental Age ‘Gaetano Barresi’, Section of Pathology, University of Messina, 98125 Messina, Italy; (C.P.); (G.T.)
| | - Gaetano Caramori
- Department of Medicine and Surgery, Sections of Pneumology, University of Parma, 43126 Parma, Italy;
| | - Antonio Ieni
- Department of Human Pathology in Adult and Developmental Age ‘Gaetano Barresi’, Section of Pathology, University of Messina, 98125 Messina, Italy; (C.P.); (G.T.)
| |
Collapse
|
17
|
Zhou Z, Huang S, Fan S, Li X, Wang C, Yu W, Du D, Zhang Y, Chen K, Fu W, Luo C. Structure-Based Design and Discovery of a Potent and Cell-Active LC3A/B Covalent Inhibitor. J Med Chem 2024; 67:12184-12204. [PMID: 39010658 DOI: 10.1021/acs.jmedchem.4c00898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/17/2024]
Abstract
Autophagy is a highly conserved cellular homeostasis maintenance mechanism in eukaryotes. Microtubule-associated protein light chain 3 (LC3) plays a crucial role in autophagy. It has multiple pairs of protein-protein interactions (PPIs) with other proteins, and these PPIs have an effect on the regulation of autophagosome formation and the recruitment of autophagic substrates. In our previous work, a small molecule covalent inhibitor DC-LC3in-D5 which could inhibit LC3A/B PPIs was identified, but a detailed study of structure-activity relationships (SARs) was lacking. Herein, a new molecule LC3in-C42 was discovered utilizing the hybridization of advantageous fragments, whose potency (IC50 = 7.6 nM) had been greatly improved compared with that of DC-LC3in-D5. LC3in-C42 inhibits autophagy at the cellular level and its efficacy far exceeds that of DC-LC3in-D5. Thus far, LC3in-C42 stands as the most potent LC3A/B small molecule inhibitor. LC3in-C42 could serve as a powerful tool for LC3A/B protein and autophagy research.
Collapse
Affiliation(s)
- Zhenfei Zhou
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai 201203, China
- Drug Discovery and Design Center and The Center for Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Guangdong 528400, China
| | - Siqi Huang
- Drug Discovery and Design Center and The Center for Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shijie Fan
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Guangdong 528400, China
| | - Xueyuan Li
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Guangdong 528400, China
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Chengyu Wang
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Guangdong 528400, China
| | - Wanlin Yu
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Guangdong 528400, China
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Daohai Du
- Drug Discovery and Design Center and The Center for Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yuanyuan Zhang
- Drug Discovery and Design Center and The Center for Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Kaixian Chen
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai 201203, China
- Drug Discovery and Design Center and The Center for Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wei Fu
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Cheng Luo
- Drug Discovery and Design Center and The Center for Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Guangdong 528400, China
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| |
Collapse
|
18
|
Elias MG, Fatima S, Mann TJ, Karan S, Mikhael M, de Souza P, Gordon CP, Scott KF, Aldrich-Wright JR. Anticancer Effect of Pt IIPHEN SS, Pt II5ME SS, Pt II56ME SS and Their Platinum(IV)-Dihydroxy Derivatives against Triple-Negative Breast Cancer and Cisplatin-Resistant Colorectal Cancer. Cancers (Basel) 2024; 16:2544. [PMID: 39061185 PMCID: PMC11274883 DOI: 10.3390/cancers16142544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/05/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024] Open
Abstract
Development of resistance to cisplatin, oxaliplatin and carboplatin remains a challenge for their use as chemotherapies, particularly in breast and colorectal cancer. Here, we compare the anticancer effect of novel complexes [Pt(1,10-phenanthroline)(1S,2S-diaminocyclohexane)](NO3)2 (PtIIPHENSS), [Pt(5-methyl-1,10-phenanthroline)(1S,2S-diaminocyclohexane)](NO3)2 (PtII5MESS) and [Pt(5,6-dimethyl-1,10-phenanthroline)(1S,2S-diaminocyclohexane)](NO3)2 (PtII56MESS) and their platinum(IV)-dihydroxy derivatives with cisplatin. Complexes are greater than 11-fold more potent than cisplatin in both 2D and 3D cell line cultures with increased selectivity for cancer cells over genetically stable cells. ICP-MS studies showed cellular uptake occurred through an active transport mechanism with considerably altered platinum concentrations found in the cytoskeleton across all complexes after 24 h. Significant reactive oxygen species generation was observed, with reduced mitochondrial membrane potential at 72 h of treatment. Late apoptosis/necrosis was shown by Annexin V-FITC/PI flow cytometry assay, accompanied by increased sub-G0/G1 cells compared with untreated cells. An increase in S and G2+M cells was seen with all complexes. Treatment resulted in significant changes in actin and tubulin staining. Intrinsic and extrinsic apoptosis markers, MAPK/ERK and PI3K/AKT activation markers, together with autophagy markers showed significant activation of these pathways by Western blot. The proteomic profile investigated post-72 h of treatment identified 1597 MDA-MB-231 and 1859 HT29 proteins quantified by mass spectroscopy, with several differentially expressed proteins relative to no treatment. GO enrichment analysis revealed a statistically significant enrichment of RNA/DNA-associated proteins in both the cell lines and specific additional processes for individual drugs. This study shows that these novel agents function as multi-mechanistic chemotherapeutics, offering promising anticancer potential, and thereby supporting further research into their application as cancer therapeutics.
Collapse
Affiliation(s)
- Maria George Elias
- School of Science, Western Sydney University, Sydney, NSW 2751, Australia; (M.G.E.); (S.K.); (M.M.); (C.P.G.)
- Medical Oncology, Ingham Institute for Applied Medical Research, Liverpool, NSW 2170, Australia; (S.F.); (T.J.M.)
| | - Shadma Fatima
- Medical Oncology, Ingham Institute for Applied Medical Research, Liverpool, NSW 2170, Australia; (S.F.); (T.J.M.)
- School of Medicine, Western Sydney University, Sydney, NSW 2751, Australia
| | - Timothy J. Mann
- Medical Oncology, Ingham Institute for Applied Medical Research, Liverpool, NSW 2170, Australia; (S.F.); (T.J.M.)
- School of Medicine, Western Sydney University, Sydney, NSW 2751, Australia
| | - Shawan Karan
- School of Science, Western Sydney University, Sydney, NSW 2751, Australia; (M.G.E.); (S.K.); (M.M.); (C.P.G.)
| | - Meena Mikhael
- School of Science, Western Sydney University, Sydney, NSW 2751, Australia; (M.G.E.); (S.K.); (M.M.); (C.P.G.)
| | - Paul de Souza
- Nepean Clinical School, Faculty of Medicine and Health, University of Sydney, Kingswood, NSW 2747, Australia;
| | - Christopher P. Gordon
- School of Science, Western Sydney University, Sydney, NSW 2751, Australia; (M.G.E.); (S.K.); (M.M.); (C.P.G.)
| | - Kieran F. Scott
- Medical Oncology, Ingham Institute for Applied Medical Research, Liverpool, NSW 2170, Australia; (S.F.); (T.J.M.)
- School of Medicine, Western Sydney University, Sydney, NSW 2751, Australia
| | - Janice R. Aldrich-Wright
- School of Science, Western Sydney University, Sydney, NSW 2751, Australia; (M.G.E.); (S.K.); (M.M.); (C.P.G.)
- School of Medicine, Western Sydney University, Sydney, NSW 2751, Australia
| |
Collapse
|
19
|
Qian Z, Xiong W, Mao X, Li J. Macrophage Perspectives in Liver Diseases: Programmed Death, Related Biomarkers, and Targeted Therapy. Biomolecules 2024; 14:700. [PMID: 38927103 PMCID: PMC11202214 DOI: 10.3390/biom14060700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 06/06/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024] Open
Abstract
Macrophages, as important immune cells of the organism, are involved in maintaining intrahepatic microenvironmental homeostasis and can undergo rapid phenotypic changes in the injured or recovering liver. In recent years, the crucial role of macrophage-programmed cell death in the development and regression of liver diseases has become a research hotspot. Moreover, macrophage-targeted therapeutic strategies are emerging in both preclinical and clinical studies. Given the macrophages' vital role in complex organismal environments, there is tremendous academic interest in developing novel therapeutic strategies that target these cells. This review provides an overview of the characteristics and interactions between macrophage polarization, programmed cell death, related biomarkers, and macrophage-targeted therapies. It aims to deepen the understanding of macrophage immunomodulation and molecular mechanisms and to provide a basis for the treatment of macrophage-associated liver diseases.
Collapse
Affiliation(s)
- Zibing Qian
- The First Clinical Medical College of Lanzhou University, Lanzhou 730000, China; (Z.Q.); (W.X.)
| | - Wanyuan Xiong
- The First Clinical Medical College of Lanzhou University, Lanzhou 730000, China; (Z.Q.); (W.X.)
| | - Xiaorong Mao
- The First Clinical Medical College of Lanzhou University, Lanzhou 730000, China; (Z.Q.); (W.X.)
- Department of Infectious Disease, The First Hospital of Lanzhou University, Lanzhou 730000, China
| | - Junfeng Li
- The First Clinical Medical College of Lanzhou University, Lanzhou 730000, China; (Z.Q.); (W.X.)
- Institute of Infectious Diseases, The First Hospital of Lanzhou University, Lanzhou 730000, China
- Department of Hepatology, The First Hospital of Lanzhou University, Lanzhou 730000, China
| |
Collapse
|
20
|
Levine J, Lobyntseva A, Shazman S, Hakim F, Gozes I. Longitudinal Genotype-Phenotype (Vineland Questionnaire) Characterization of 15 ADNP Syndrome Cases Highlights Mutated Protein Length and Structural Characteristics Correlation with Communicative Abilities Accentuated in Males. J Mol Neurosci 2024; 74:15. [PMID: 38282129 DOI: 10.1007/s12031-024-02189-4] [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] [Accepted: 01/08/2024] [Indexed: 01/30/2024]
Abstract
Activity-dependent neuroprotective protein (ADNP) is essential for neurodevelopment and de novo mutations in ADNP cause the ADNP syndrome. From brain pathologies point of view, tauopathy has been demonstrated at a young age, implying stunted development coupled with early/accelerated neurodegeneration. Given potential genotype-phenotype differences and age-dependency, we have assessed here a cohort of 15 individuals (1-27-year-old), using 1-3 longitudinal parent (caretaker) interview/s (Vineland 3 questionnaire) over several years. Our results indicated developmental delays, or even developmental arrests, coupled with potential spurts of development at early ages. Severe outcomes correlated with the truncating high impact mutation, in other words, the remaining mutated protein length as well as with the tested individual age, corroborating the hypothesis of developmental delays coupled with accelerated aging. A significant correlation was noted between mutated protein length and communication, implying a high impact of ADNP on communicative skills. Additionally, correlations were discovered between the two previously described epi-genetic signatures in ADNP emphasizing aberrant acquisition of motor behaviors, with truncating mutations around the nuclear localization signal being mostly affected. Finally, all individuals seem to acquire an age equivalent of 1-6 years, requiring disease modification treatment, such as the ADNP-derived drug candidate, NAP (davunetide), which has recently shown efficacy in women suffering from the neurodegenerative disorder, progressive supranuclear palsy (PSP), a late-onset tauopathy.
Collapse
Affiliation(s)
- Jospeh Levine
- The Elton Laboratory for Molecular Neuroendocrinology, Department of Human Molecular Genetics and Biochemistry, Faculty of Medicine, Sagol School of Neuroscience and Adams Super Center for Brain Studies, Tel Aviv University, Tel Aviv, 6997801, Israel
- Psychiatric Division, Ben Gurion University of the Negev, Beersheba, Israel
| | - Alexandra Lobyntseva
- The Elton Laboratory for Molecular Neuroendocrinology, Department of Human Molecular Genetics and Biochemistry, Faculty of Medicine, Sagol School of Neuroscience and Adams Super Center for Brain Studies, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Shula Shazman
- Department of Mathematics and Computer Science, The Open University of Israel, Ra'anana, Israel
| | | | - Illana Gozes
- The Elton Laboratory for Molecular Neuroendocrinology, Department of Human Molecular Genetics and Biochemistry, Faculty of Medicine, Sagol School of Neuroscience and Adams Super Center for Brain Studies, Tel Aviv University, Tel Aviv, 6997801, Israel.
| |
Collapse
|
21
|
Khalil MI, Ali MM, Holail J, Houssein M. Growth or death? Control of cell destiny by mTOR and autophagy pathways. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2023; 185:39-55. [PMID: 37944568 DOI: 10.1016/j.pbiomolbio.2023.10.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 10/08/2023] [Accepted: 10/23/2023] [Indexed: 11/12/2023]
Abstract
One of the central regulators of cell growth, proliferation, and metabolism is the mammalian target of rapamycin, mTOR, which exists in two structurally and functionally different complexes: mTORC1 and mTORC2; unlike m TORC2, mTORC1 is activated in response to the sufficiency of nutrients and is inhibited by rapamycin. mTOR complexes have critical roles not only in protein synthesis, gene transcription regulation, proliferation, tumor metabolism, but also in the regulation of the programmed cell death mechanisms such as autophagy and apoptosis. Autophagy is a conserved catabolic mechanism in which damaged molecules are recycled in response to nutrient starvation. Emerging evidence indicates that the mTOR signaling pathway is frequently activated in tumors. In addition, dysregulation of autophagy was associated with the development of a variety of human diseases, such as cancer and aging. Since mTOR can inhibit the induction of the autophagic process from the early stages of autophagosome formation to the late stage of lysosome degradation, the use of mTOR inhibitors to regulate autophagy could be considered a potential therapeutic option. The present review sheds light on the mTOR and autophagy signaling pathways and the mechanisms of regulation of mTOR-autophagy.
Collapse
Affiliation(s)
- Mahmoud I Khalil
- Department of Biological Sciences, Faculty of Science, Beirut Arab University, Beirut, 11072809, Lebanon; Molecular Biology Unit, Department of Zoology, Faculty of Science, Alexandria University, Alexandria, 21511, Egypt.
| | - Mohamad M Ali
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala University, SE-751 23, Uppsala, Sweden.
| | - Jasmine Holail
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, Riyadh, Saudi Arabia; Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom.
| | - Marwa Houssein
- Department of Biological Sciences, Faculty of Science, Beirut Arab University, Beirut, 11072809, Lebanon.
| |
Collapse
|
22
|
Ahamed A, Hasan M, Samanta A, Alam SSM, Jamil Z, Ali S, Hoque M. Prospective pharmacological potential of cryptotanshinone in cancer therapy. PHARMACOLOGICAL RESEARCH - MODERN CHINESE MEDICINE 2023; 9:100308. [DOI: 10.1016/j.prmcm.2023.100308] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2025]
|
23
|
Wang S, Nie J, Xu K, Liu Y, Tong W, Li A, Zuo W, Liu Z, Yang F. YY1 is regulated by ALKBH5-mediated m6A modification and promotes autophagy and cancer progression through targeting ATG4B. Aging (Albany NY) 2023; 15:9590-9613. [PMID: 37724907 PMCID: PMC10564435 DOI: 10.18632/aging.205037] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 08/29/2023] [Indexed: 09/21/2023]
Abstract
YY1 affects tumorigenesis and metastasis in multiple ways. However, the function of YY1 and the potential mechanisms through which it operates in gastric cancer (GC) progression by regulating autophagy remains poorly understood. This study aimed to assess the essential transcription factors (TFs) involved in autophagy regulation in GC. Western blot, RFP-GFP-LC3 double fluorescence and transmission electron microscopy (TEM) assays were used to probe autophagy activity in GC cells. Methylated RNA immunoprecipitation (MeRIP) was utilized to evaluate the ALKBH5-regulated m6A levels of YY1. Gain- and loss-of-function assays were employed in the scrutiny of the biological effects of the ALKBH5/YY1/ATG4B axis on cancer cell proliferation and invasion abilities in vitro. Per the findings, YY1 was identified as a crucial transcriptional activator of cancer autophagy-related genes and promoted the proliferation and aggressiveness of cancer cells associated with enhanced ATG4B-mediated autophagy. However, ectopic ALKBH5 expression abolished the YY1-induced effect via m6A modification. Importantly, YTHDF1 facilitated the mRNA stability of YY1 through m6A recognition. Collectively, this study found that YY1 was regulated by ALKBH5 and YTHDF1-mediated m6A modification and served as an autophagy-dependent tumor driver to accelerate cancer progression through ATG4B transactivation, providing an exploitable therapeutic target for GC.
Collapse
Affiliation(s)
- Shijiang Wang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang 330006, People’s Republic of China
- Medical Innovation Center, The First Affiliated Hospital of Nanchang University, Nanchang 330006, People’s Republic of China
- Institute of Spine and Spinal Cord, The First Affiliated Hospital of Nanchang University, Nanchang 330006, People’s Republic of China
| | - Jiangbo Nie
- Department of Orthopedic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang 330006, People’s Republic of China
- Medical Innovation Center, The First Affiliated Hospital of Nanchang University, Nanchang 330006, People’s Republic of China
- Institute of Spine and Spinal Cord, The First Affiliated Hospital of Nanchang University, Nanchang 330006, People’s Republic of China
| | - Kaiying Xu
- Medical Innovation Center, The First Affiliated Hospital of Nanchang University, Nanchang 330006, People’s Republic of China
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang 330006, People’s Republic of China
| | - Yangyang Liu
- Medical Innovation Center, The First Affiliated Hospital of Nanchang University, Nanchang 330006, People’s Republic of China
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang 330006, People’s Republic of China
| | - Weilai Tong
- Department of Orthopedic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang 330006, People’s Republic of China
- Medical Innovation Center, The First Affiliated Hospital of Nanchang University, Nanchang 330006, People’s Republic of China
- Institute of Spine and Spinal Cord, The First Affiliated Hospital of Nanchang University, Nanchang 330006, People’s Republic of China
| | - Anan Li
- Department of Orthopedic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang 330006, People’s Republic of China
- Medical Innovation Center, The First Affiliated Hospital of Nanchang University, Nanchang 330006, People’s Republic of China
- Institute of Spine and Spinal Cord, The First Affiliated Hospital of Nanchang University, Nanchang 330006, People’s Republic of China
| | - Wei Zuo
- Department of Orthopedic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang 330006, People’s Republic of China
- Medical Innovation Center, The First Affiliated Hospital of Nanchang University, Nanchang 330006, People’s Republic of China
- Institute of Spine and Spinal Cord, The First Affiliated Hospital of Nanchang University, Nanchang 330006, People’s Republic of China
| | - Zhili Liu
- Department of Orthopedic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang 330006, People’s Republic of China
- Medical Innovation Center, The First Affiliated Hospital of Nanchang University, Nanchang 330006, People’s Republic of China
- Institute of Spine and Spinal Cord, The First Affiliated Hospital of Nanchang University, Nanchang 330006, People’s Republic of China
| | - Feng Yang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang 330006, People’s Republic of China
- Medical Innovation Center, The First Affiliated Hospital of Nanchang University, Nanchang 330006, People’s Republic of China
- Institute of Spine and Spinal Cord, The First Affiliated Hospital of Nanchang University, Nanchang 330006, People’s Republic of China
- Postdoctoral Innovation Practice Base, The First Affiliated Hospital of Nanchang University, Nanchang 330006, People’s Republic of China
| |
Collapse
|
24
|
Roda E, De Luca F, Priori EC, Ratto D, Pinelli S, Corradini E, Mozzoni P, Poli D, Mazzini G, Bottone MG, Gatti AM, Marti M, Locatelli CA, Rossi P, Bottai D. The Designer Drug αPHP Affected Cell Proliferation and Triggered Deathly Mechanisms in Murine Neural Stem/Progenitor Cells. BIOLOGY 2023; 12:1225. [PMID: 37759624 PMCID: PMC10525791 DOI: 10.3390/biology12091225] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/08/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023]
Abstract
Increasing reports of neurological and psychiatric outcomes due to psychostimulant synthetic cathinones (SCs) have recently raised public concern. However, the understanding of neurotoxic mechanisms is still lacking, particularly for the under-investigated αPHP, one of the major MDPV derivatives. In particular, its effects on neural stem/progenitor cell cultures (NSPCs) are still unexplored. Therefore, in the current in vitro study, the effects of increasing αPHP concentrations (25-2000 μM), on cell viability/proliferation, morphology/ultrastructure, genotoxicity and cell death pathways, have been evaluated after exposure in murine NSPCs, using a battery of complementary techniques, i.e., MTT and clonogenic assay, flow cytometry, immunocytochemistry, TEM, and patch clamp. We revealed that αPHP was able to induce a dose-dependent significant decrease of the viability, proliferation and clonal capability of the NSPCs, paralleled by the resting membrane potential depolarization and apoptotic/autophagic/necroptotic pathway activation. Moreover, ultrastructural alterations were clearly observed. Overall, our current findings demonstrate that αPHP, damaging NSPCs and the morpho-functional fundamental units of adult neurogenic niches may affect neurogenesis, possibly triggering long-lasting, irreversible CNS damage. The present investigation could pave the way for a broadened understanding of SCs toxicology, needed to establish an appropriate treatment for NPS and the potential consequences for public health.
Collapse
Affiliation(s)
- Elisa Roda
- Laboratory of Clinical & Experimental Toxicology, Pavia Poison Centre, National Toxicology Information Centre, Toxicology Unit, Istituti Clinici Scientifici Maugeri IRCCS, 27100 Pavia, Italy (C.A.L.)
| | - Fabrizio De Luca
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, 27100 Pavia, Italy; (F.D.L.); (P.R.)
| | - Erica Cecilia Priori
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, 27100 Pavia, Italy; (F.D.L.); (P.R.)
| | - Daniela Ratto
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, 27100 Pavia, Italy; (F.D.L.); (P.R.)
| | - Silvana Pinelli
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Emilia Corradini
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Paola Mozzoni
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Diana Poli
- INAIL Research, Department of Occupational and Environmental Medicine, Epidemiology and Hygiene, Via Fontana Candida, 1, 00078 Monte Porzio Catone, Italy
| | - Giuliano Mazzini
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, 27100 Pavia, Italy; (F.D.L.); (P.R.)
- Institute of Molecular Genetics—CNR (National Research Council), 27100 Pavia, Italy
| | - Maria Grazia Bottone
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, 27100 Pavia, Italy; (F.D.L.); (P.R.)
| | - Anna Maria Gatti
- Laboratory of Clinical & Experimental Toxicology, Pavia Poison Centre, National Toxicology Information Centre, Toxicology Unit, Istituti Clinici Scientifici Maugeri IRCCS, 27100 Pavia, Italy (C.A.L.)
| | - Matteo Marti
- Department of Translational Medicine, Section of Legal Medicine, LTTA Center and University Center of Gender Medicine, University of Ferrara, 44121 Ferrara, Italy;
- Collaborative Centre for the Italian National Early Warning System, Department of Anti-Drug Policies, Presidency of the Council of Ministers, 44121 Ferrara, Italy
| | - Carlo Alessandro Locatelli
- Laboratory of Clinical & Experimental Toxicology, Pavia Poison Centre, National Toxicology Information Centre, Toxicology Unit, Istituti Clinici Scientifici Maugeri IRCCS, 27100 Pavia, Italy (C.A.L.)
| | - Paola Rossi
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, 27100 Pavia, Italy; (F.D.L.); (P.R.)
| | - Daniele Bottai
- Department of Pharmaceutical Sciences, Section of Pharmacology and Biosciences, University of Milan, Via Balzaretti 9, 20133 Milan, Italy;
| |
Collapse
|
25
|
Mareninova OA, Gretler SR, Lee GE, Pimienta M, Qin Y, Elperin JM, Ni J, Razga Z, Gukovskaya AS, Gukovsky I. Ethanol inhibits pancreatic acinar cell autophagy through upregulation of ATG4B, mediating pathological responses of alcoholic pancreatitis. Am J Physiol Gastrointest Liver Physiol 2023; 325:G265-G278. [PMID: 37431575 PMCID: PMC10511161 DOI: 10.1152/ajpgi.00053.2023] [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: 03/17/2023] [Revised: 06/23/2023] [Accepted: 07/01/2023] [Indexed: 07/12/2023]
Abstract
Excessive alcohol intake is a major risk factor for pancreatitis, sensitizing the exocrine pancreas to stressors by mechanisms that remain obscure. Impaired autophagy drives nonalcoholic pancreatitis, but the effects of ethanol (EtOH) and alcoholic pancreatitis on autophagy are poorly understood. Here, we find that ethanol reduces autophagosome formation in pancreatic acinar cells, both in a mouse model of alcoholic pancreatitis induced by a combination of EtOH diet and cerulein (a CCK ortholog) and in EtOH+CCK-treated acinar cells (ex vivo model). Ethanol treatments decreased pancreatic level of LC3-II, a key mediator of autophagosome formation. This was caused by ethanol-induced upregulation of ATG4B, a cysteine protease that, cell dependently, regulates the balance between cytosolic LC3-I and membrane-bound LC3-II. We show that ATG4B negatively regulates LC3-II in acinar cells subjected to EtOH treatments. Ethanol raised ATG4B level by inhibiting its degradation, enhanced ATG4B enzymatic activity, and strengthened its interaction with LC3-II. We also found an increase in ATG4B and impaired autophagy in a dissimilar, nonsecretagogue model of alcoholic pancreatitis induced by EtOH plus palmitoleic acid. Adenoviral ATG4B overexpression in acinar cells greatly reduced LC3-II and inhibited autophagy. Furthermore, it aggravated trypsinogen activation and necrosis, mimicking key responses of ex vivo alcoholic pancreatitis. Conversely, shRNA Atg4B knockdown enhanced autophagosome formation and alleviated ethanol-induced acinar cell damage. The results reveal a novel mechanism, whereby ethanol inhibits autophagosome formation and thus sensitizes pancreatitis, and a key role of ATG4B in ethanol's effects on autophagy. Enhancing pancreatic autophagy, particularly by downregulating ATG4B, could be beneficial in mitigating the severity of alcoholic pancreatitis.NEW & NOTEWORTHY Ethanol sensitizes mice and humans to pancreatitis, but the underlying mechanisms remain obscure. Autophagy is important for maintaining pancreatic acinar cell homeostasis, and its impairment drives pancreatitis. This study reveals a novel mechanism, whereby ethanol inhibits autophagosome formation through upregulating ATG4B, a key cysteine protease. ATG4B upregulation inhibits autophagy in acinar cells and aggravates pathological responses of experimental alcoholic pancreatitis. Enhancing pancreatic autophagy, particularly by down-regulating ATG4B, could be beneficial for treatment of alcoholic pancreatitis.
Collapse
Affiliation(s)
- Olga A Mareninova
- David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California, United States
- Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California, United States
- Southern California Research Center for Alcoholic Liver and Pancreatic Diseases and Cirrhosis, Los Angeles, California, United States
| | - Sophie R Gretler
- David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California, United States
- Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California, United States
- Southern California Research Center for Alcoholic Liver and Pancreatic Diseases and Cirrhosis, Los Angeles, California, United States
| | - Grace E Lee
- David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California, United States
| | - Michael Pimienta
- David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California, United States
| | - Yueqiu Qin
- David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California, United States
- Division of Gastroenterology and Hepatology, Youjiang Medical University for Nationalities, Baise, China
| | - Jason M Elperin
- David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California, United States
| | - Jinliang Ni
- David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California, United States
- First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Zsolt Razga
- Institute of Pathology, University of Szeged, Szeged, Hungary
| | - Anna S Gukovskaya
- David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California, United States
- Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California, United States
- Southern California Research Center for Alcoholic Liver and Pancreatic Diseases and Cirrhosis, Los Angeles, California, United States
| | - Ilya Gukovsky
- David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California, United States
- Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California, United States
- Southern California Research Center for Alcoholic Liver and Pancreatic Diseases and Cirrhosis, Los Angeles, California, United States
| |
Collapse
|
26
|
Li C, Zhang Y, Zhao X, Li L, Kong X. Autophagy regulation of virus infection in aquatic animals. REVIEWS IN AQUACULTURE 2023; 15:1405-1420. [DOI: 10.1111/raq.12785] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 01/04/2023] [Indexed: 01/04/2025]
Abstract
AbstractAutophagy is a conserved intracellular degradation process that is required to maintain host homeostasis and cope with invading pathogens. Over the past few decades, studies on mammals have greatly increased our understanding of the relationship between autophagy and virus infection. Autophagy may convey the invader to lysosomes to degrade or activate the host immune response against virus replication. However, many viruses have developed some strategies that evade the degradative nature of autophagy or hijack this pathway for their gain. It follows that autophagy during viral infection is a double‐edged sword. In contrast to mammals, the review on autophagy modulated by the aquatic animal virus is limited. Here, after a brief description of the main information about autophagy, we highlight current progress on the interplays between autophagy and virus infection in aquatic animals, including the phenomenon of autophagy upon virus infection, the effect of modulating autophagy on virus replication, and the crosstalk between autophagy and immune response during virus infection. This review will help us better understand the pathogenic mechanism of aquatic animal viruses and develop proper antiviral countermeasures aimed at modulating autophagy.
Collapse
Affiliation(s)
- Chen Li
- Engineering Lab of Henan Province for Aquatic Animal Disease Control College of Fisheries, Henan Normal University Xinxiang Henan Province PR China
| | - Yunli Zhang
- Engineering Lab of Henan Province for Aquatic Animal Disease Control College of Fisheries, Henan Normal University Xinxiang Henan Province PR China
| | - Xianliang Zhao
- Engineering Lab of Henan Province for Aquatic Animal Disease Control College of Fisheries, Henan Normal University Xinxiang Henan Province PR China
| | - Li Li
- Engineering Lab of Henan Province for Aquatic Animal Disease Control College of Fisheries, Henan Normal University Xinxiang Henan Province PR China
| | - Xianghui Kong
- Engineering Lab of Henan Province for Aquatic Animal Disease Control College of Fisheries, Henan Normal University Xinxiang Henan Province PR China
| |
Collapse
|
27
|
Li X, Sung P, Zhang D, Yan L. Curcumin in vitro Neuroprotective Effects Are Mediated by p62/keap-1/Nrf2 and PI3K/AKT Signaling Pathway and Autophagy Inhibition. Physiol Res 2023; 72:497-510. [PMID: 37795892 PMCID: PMC10634561 DOI: 10.33549/physiolres.935054] [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: 12/15/2022] [Accepted: 05/11/2023] [Indexed: 01/05/2024] Open
Abstract
Oxidative stress and autophagy are potential mechanisms associated with cerebral ischemia/reperfusion injury (IRI) and is usually linked to inflammatory responses and apoptosis. Curcumin has recently been demonstrated to exhibit anti-inflammatory, anti-oxidant, anti-apoptotic and autophagy regulation properties. However, mechanism of curcumin on IRI-induced oxidative stress and autophagy remains not well understood. We evaluated the protective effects and potential mechanisms of curcumin on cerebral microvascular endothelial cells (bEnd.3) and neuronal cells (HT22) against oxygen glucose deprivation/reoxygenation (OGD/R) in vitro models that mimic in vivo cerebral IRI. The cell counting kit-8 (CCK-8) and lactate dehydrogenase (LDH) activity assays revealed that curcumin attenuated the OGD/R-induced injury in a dose-specific manner. OGD/R induced elevated levels of inflammatory cytokines TNF-alpha, IL-6 as well as IL-1beta, and these effects were notably reduced by curcumin. OGD/R-mediated apoptosis was suppressed by curcumin via upregulating B-cell lymphoma-2 (Bcl-2) and downregulating Bcl-associated X (Bax), cleaved-caspase3 and TUNEL apoptosis marker. Additionally, curcumin increased superoxide dismutase (SOD) and glutathione (GSH), but suppressed malondialdehyde (MDA) and reactive oxygen species (ROS) content. Curcumin inhibited the levels of autophagic biomarkers such as LC3 II/LC3 I and Beclin1. Particularly, curcumin induced p62 accumulation and its interactions with keap1 and promoted NF-E2-related factor 2 (Nrf2) translocation to nucleus, accompanied by increased NADPH quinone dehydrogenase (Nqo1) and heme oxygenase 1 (HO-1). Treatment of curcumin increased phosphorylation-phosphatidylinositol 3 kinase (p-PI3K) and p-protein kinase B (p-AKT). The autophagy inhibitor 3-methyladenine (3-MA) activated the keap-1/Nrf2 and PI3K/AKT pathways. This study highlights the neuroprotective effects of curcumin on cerebral IRI.
Collapse
Affiliation(s)
- X Li
- Department of Neurology, Tangshan Gongren Hospital, Tangshan, Hebei Province, China
| | | | | | | |
Collapse
|
28
|
杨 丹, 王 刚, 杨 丽, 段 壬, 陈 显. [Effect of ligustrazine on hypoxic-ischemic encephalopathy in neonatal rats by regulating autophagy through the PINK1/Parkin pathway]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2023; 25:751-758. [PMID: 37529959 PMCID: PMC10414166 DOI: 10.7499/j.issn.1008-8830.2302026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 05/15/2023] [Indexed: 08/03/2023]
Abstract
OBJECTIVES To study the effect of ligustrazine injection on mitophagy in neonatal rats with hypoxic-ischemic encephalopathy (HIE) and its molecular mechanism. METHODS Neonatal Sprague-Dawley rats, aged 7 days, were randomly divided into a sham-operation group with 8 rats, a model group with 12 rats, and a ligustrazine group with 12 rats. The rats in the model group and the ligustrazine group were used to establish a neonatal rat model of HIE by ligation of the left common carotid artery followed by hypoxia treatment, and blood vessels were exposed without any other treatment for the rats in the sham-operation group. The rats in the ligustrazine group were intraperitoneally injected with ligustrazine (20 mg/kg) daily after hypoxia-ischemia, and those in the sham-operation group and the model group were intraperitoneally injected with an equal volume of normal saline daily. Samples were collected after 7 days of treatment. Hematoxylin and eosin staining and Nissl staining were used to observe the pathological changes of neurons in brain tissue; immunohistochemical staining was used to observe the positive expression of PINK1 and Parkin in the hippocampus and cortex; TUNEL staining was used to measure neuronal apoptosis; Western blotting was used to measure the expression levels of the mitophagy pathway proteins PINK1 and Parkin and the autophagy-related proteins Beclin-1, microtubule-associated protein 1 light chain 3 (LC3), and ubiquitin-binding protein (P62). RESULTS Compared with the sham-operation group, the model group had a significant reduction in the number of neurons, an increase in intercellular space, loose arrangement, lipid vacuolization, and a reduction in Nissl bodies. The increased positive expression of PINK1 and Parkin, apoptosis rate of neurons, and protein expression levels of PINK1, Parkin, Beclin1 and LC3 (P<0.05) and the decreased protein expression level of P62 in the hippocampus were also observed in the model group (P<0.05). Compared with the model group, the ligustrazine group had a significant increase in the number of neurons with ordered arrangement and an increase in Nissl bodies, significant reductions in the positive expression of PINK1 and Parkin, the apoptosis rate of neurons, and the protein expression levels of PINK1, Parkin, Beclin1, and LC3 (P<0.05), and a significant increase in the protein expression level of P62 (P<0.05). CONCLUSIONS Ligustrazine can alleviate hypoxic-ischemic brain damage and inhibit neuronal apoptosis in neonatal rats to a certain extent, possibly by inhibiting PINK1/Parkin-mediated autophagy.
Collapse
|
29
|
Park SJ, Jang JW, Moon EY. Bisphenol A-induced autophagy ameliorates human B cell death through Nrf2-mediated regulation of Atg7 and Beclin1 expression by Syk activation. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 260:115061. [PMID: 37257343 DOI: 10.1016/j.ecoenv.2023.115061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 05/12/2023] [Accepted: 05/21/2023] [Indexed: 06/02/2023]
Abstract
The widely used plasticizer bisphenol A (BPA) is known as an endocrine-disrupting chemical (EDC). Many studies have shown that BPA contributes to diseases involving immune system alterations, but the underlying mechanisms have yet to be elucidated. We previously reported that BPA at concentration of 100 μM caused human B cell death in accordance with an increase in nuclear factor (erythroid-derived 2)-like 2(Nrf2) expression. Autophagy is a cellular process that degraded and recycles cytoplasmic constituents. Here, we investigated whether BPA induces autophagy through Nrf2, which is associated with regulation of B cell death using human WiL2-NS lymphoblast B cells. Then, cell viability was assessed by various assays using trypan blue, MTT or Celltiter glo luminescent substrate and DAPI. When WiL2-NS cells were treated with BPA, cell viability was decreased and LC3 autophagy cargo protein/puncta was increased. BPA-induced autophagy was confirmed by the modification of LC3 puncta formation or autophagy flux turnover with the treatment of hydroxychloroquine(HCQ), NH4Cl and PI3K inhibitors including 3-methyladenine(3-MA), LY294002 and wortmannin. BPA treatment increased the expression of autophagy-related gene(Atg)7 and Beclin1 as well as Nrf2 induced by the production of reactive oxygen species (ROS). The inhibition of autophagy with siAtg7 or siBeclin1 and Nrf2 depletion aggravated BPA-induced cell death. BPA enhanced the bound of Nrf2 to the specific region on Beclin1 and Atg7 promoter. Spleen tyrosine kinase(Syk) activity was enhanced in response to BPA treatment. Bay61-3606, Syk inhibitor, decreased LC3 and the expression of Atg7 and Beclin1, leading to the increase of BPA-induced B cell death. The results suggest that BPA-induced autophagy ameliorates human B cell death through Nrf2-mediated regulation of Atg7 and Beclin1 expression.
Collapse
Affiliation(s)
- So-Jeong Park
- Department of Integrative Bioscience and Biotechnology, Sejong University, Seoul 05006, the Republic of Korea
| | - Ju-Won Jang
- Department of Integrative Bioscience and Biotechnology, Sejong University, Seoul 05006, the Republic of Korea
| | - Eun-Yi Moon
- Department of Integrative Bioscience and Biotechnology, Sejong University, Seoul 05006, the Republic of Korea.
| |
Collapse
|
30
|
Taha M, Elazab ST, Baokbah TAS, Al-Kushi AG, Mahmoud ME, Abdelbagi O, Qusty NF, El-Shenbaby I, Babateen O, Badawy AM, Ibrahim MM. Palliative Role of Zamzam Water against Cyclosporine-Induced Nephrotoxicity through Modulating Autophagy and Apoptosis Crosstalk. TOXICS 2023; 11:377. [PMID: 37112604 PMCID: PMC10144806 DOI: 10.3390/toxics11040377] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 04/09/2023] [Accepted: 04/14/2023] [Indexed: 06/19/2023]
Abstract
Cyclosporine (CsA) is considered one of the main components of treatment protocols for organ transplantation owing to its immunosuppressive effect. However, its use is very restricted due to its nephrotoxic effect. ZW is an alkaline fluid rich in various trace elements and has a great ability to stimulate antioxidant processes. This study aimed to investigate the possible mitigating effect of ZW on CsA-induced nephrotoxicity and its underlying mechanisms. Forty rats were allocated into four groups (n = 10): a control group, ZW group, cyclosporine A group (injected subcutaneously (SC) with CsA (20 mg/kg/day)), and cyclosporine A+ Zamzam water group (administered CsA (SC) and ZW as their only drinking water (100 mL/cage/day) for 21 days). Exposure to CsA significantly (p < 0.001) increased the serum creatinine level, lipid peroxidation marker level (malondialdehyde; MDA), and the expression of apoptotic markers procaspase-8, caspase-8, caspase- 9, calpain, cytochrome c, caspas-3, P62, and mTOR in renal tissues. Meanwhile, it markedly decreased (p< 0.001) the autophagic markers (AMPK, ULK-I, ATag5, LC3, and Beclin-1), antiapoptotic Bcl-2, and antioxidant enzymes. Moreover, the administration of CsA caused histological alterations in renal tissues. ZW significantly (p < 0.001) reversed all the changes caused by CsA and conclusively achieved a positive outcome in restraining CsA-induced nephrotoxicity, as indicated by the restoration of the histological architecture, improvement of renal function, inhibition of apoptosis, and enhancement of autophagy via the AMPK/mTOR pathway.
Collapse
Affiliation(s)
- Medhat Taha
- Department of Anatomy and Embryology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt
- Department of Anatomy, Al-Qunfudah Medical College, Umm Al-Qura University, Al-Qunfudah 28814, Saudi Arabia
| | - Sara T. Elazab
- Department of Pharmacology, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Tourki A. S. Baokbah
- Department of Medical Emergency Services, College of Health Sciences-AlQunfudah, Umm Al-Qura University, Al-Qunfudah 28814, Saudi Arabia
| | - Abdullah G. Al-Kushi
- Department of Human Anatomy, Faculty of Medicine, Umm Al-Qura University, Makkah, Mecca 24382, Saudi Arabia
| | - Mohamed Ezzat Mahmoud
- Histology Department, Damietta Faculty of Medicine, Al-Azhar University, Damietta 34711, Egypt
| | - Omer Abdelbagi
- Department of Pathology, Qunfudah Faculty of Medicine, Umm-Al-Qura University Kingdom of Saudi Arabia, Makka 24382, Saudi Arabia
| | - Naeem F. Qusty
- Medical Laboratories Department, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah 24382, Saudi Arabia
| | - Ibrahim El-Shenbaby
- Clinical Pharmacology Department, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Omar Babateen
- Department of physiology, Faculty of Medicine, Umm Al-Qura University, Makkah 24382, Saudi Arabia
| | - Alaa. M. Badawy
- Department of Anatomy and Embryology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Mohie Mahmoud Ibrahim
- Department of Anatomy and Embryology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt
| |
Collapse
|
31
|
Strontium Ranelate Inhibits Osteoclastogenesis through NF-κB-Pathway-Dependent Autophagy. Bioengineering (Basel) 2023; 10:bioengineering10030365. [PMID: 36978756 PMCID: PMC10045081 DOI: 10.3390/bioengineering10030365] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 02/17/2023] [Accepted: 03/08/2023] [Indexed: 03/19/2023] Open
Abstract
Strontium ranelate (SR) is a pharmaceutical agent used for the prevention and treatment of osteoporosis and fragility fracture. However, little attention has been paid to the effect of SR on alveolar bone remodeling during orthodontic tooth movement and its underlying mechanism. Here, we investigated the influence of SR on orthodontic tooth movement and tooth resorption in Sprague–Dawley rats and the relationship between the nuclear factor–kappa B (NF-κB) pathway, autophagy, and osteoclastogenesis after the administration of SR in vitro and in vivo. In this study, it was found that SR reduced the expression of autophagy-related proteins at the pressure side of the first molars during orthodontic tooth movement. Similarly, the expression of these autophagy-related proteins and the size and number of autophagosomes were downregulated by SR in vitro. The results also showed that SR reduced the number of osteoclasts and suppressed orthodontic tooth movement and root resorption in rats, which could be partially restored using rapamycin, an autophagy inducer. Autophagy was attenuated after pre-osteoclasts were treated with Bay 11-7082, an NF-κB pathway inhibitor, while SR reduced the expression of the proteins central to the NF-κB pathway. Collectively, this study revealed that SR might suppress osteoclastogenesis through NF-κB-pathway-dependent autophagy, resulting in the inhibition of orthodontic tooth movement and root resorption in rats, which might offer a new insight into the treatment of malocclusion and bone metabolic diseases.
Collapse
|
32
|
Zheng R, Yu Y, Lv L, Zhang Y, Deng H, Li J, Zhang B. m 6A reader HNRNPA2B1 destabilization of ATG4B regulates autophagic activity, proliferation and olaparib sensitivity in breast cancer. Exp Cell Res 2023; 424:113487. [PMID: 36693492 DOI: 10.1016/j.yexcr.2023.113487] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 12/23/2022] [Accepted: 01/18/2023] [Indexed: 01/22/2023]
Abstract
N6-methyladenosine RNA (m6A) is the most extensive epigenetic modification in mRNA and influences tumor progression. However, the role of m6A regulators and specific mechanisms in breast cancer still need further study. Here, we investigated the significance of the m6A reader HNRNPA2B1 and explored its influence on autophagy and drug sensitivity in breast cancer. HNRNPA2B1 was selected by bioinformatics analysis, and its high expression level was identified in breast cancer tissues and cell lines. HNRNPA2B1 was related to poor prognosis. Downregulation of HNRNPA2B1 reduced proliferation, enhanced autophagic flux, and partially reversed de novo resistance to olaparib in breast cancer. ATG4B was determined by RIP and MeRIP assays as a downstream gene of HNRNPA2B1, by which recognized the m6A site in the 3'UTR. Overexpression of ATG4B rescued the malignancy driven by HNRNPA2B1 in breast cancer cells and increased the olaparib sensitivity. Our study revealed that the m6A reader HNRNPA2B1 mediated proliferation and autophagy in breast cancer cell lines by facilitating ATG4B mRNA decay and targeting HNRNPA2B1/m6A/ATG4B might enhance the olaparib sensitivity of breast cancer cells.
Collapse
Affiliation(s)
- Renjing Zheng
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan Province, 430022, People's Republic of China
| | - Yuanhang Yu
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan Province, 430022, People's Republic of China
| | - Lianqiu Lv
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan Province, 430022, People's Republic of China
| | - Yue Zhang
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan Province, 430022, People's Republic of China
| | - Huifang Deng
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan Province, 430022, People's Republic of China
| | - Jiyong Li
- Department of Breast and Thyroid Surgery, Huangpi People's Hospital, Jianghan University, Wuhan Province, 430300, People's Republic of China
| | - Bo Zhang
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan Province, 430022, People's Republic of China.
| |
Collapse
|
33
|
Zhang J, Tang Y, Hu Z, Xu W, Ma Y, Xu P, Xing H, Niu Q. The inhibition of TRPML1/TFEB leads to lysosomal biogenesis disorder, contributes to developmental fluoride neurotoxicity. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 250:114511. [PMID: 36608573 DOI: 10.1016/j.ecoenv.2023.114511] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 12/29/2022] [Accepted: 01/03/2023] [Indexed: 06/17/2023]
Abstract
Fluoride is capable of inducing developmental neurotoxicity; regrettably, the mechanism is obscure. We aimed to probe the role of lysosomal biogenesis disorder in developmental fluoride neurotoxicity-specifically, the regulating effect of the transient receptor potential mucolipin 1 (TRPML1)/transcription factor EB (TFEB) signaling pathway on lysosomal biogenesis. Sprague-Dawley rats were given fluoridated water freely, during pregnancy to the parental rats to 2 months after delivery to the offspring. In addition, neuroblastoma SH-SY5Y cells were treated with sodium fluoride (NaF), with or without mucolipin synthetic agonist 1 (ML-SA1) or adenovirus TFEB (Ad-TFEB) intervention. Our findings revealed that NaF impaired learning and memory as well as memory retention capacities in rat offspring, induced lysosomal biogenesis disorder, and decreased lysosomal degradation capacity, autophagosome accumulation, autophagic flux blockade, apoptosis, and pyroptosis. These changes were evidenced by the decreased expression of TRPML1, nuclear TFEB, LAMP2, CTSB, and CTSD, as well as increased expression of LC3-II, p62, cleaved PARP, NLRP3, Caspase1, and IL-1β. Furthermore, TRPML1 activation and TFEB overexpression both restored TFEB nuclear protein expression and promoted lysosomal biogenesis while enhancing lysosomal degradation capacity, recovering autophagic flux, and attenuating NaF-induced apoptosis and pyroptosis. Taken together, these results show that NaF promotes the progression of developmental fluoride neurotoxicity by inhibiting TRPML1/TFEB expression and impeding lysosomal biogenesis. Notably, the activation of TRPML1/TFEB alleviated NaF-induced developmental neurotoxicity. Therefore, TRPML1/TFEB may be promising markers of developmental fluoride neurotoxicity.
Collapse
Affiliation(s)
- Jingjing Zhang
- Department of Preventive Medicine, School of Medicine, Shihezi University, Shihezi, Xinjiang, People's Republic of China; Key Laboratory of Preventive Medicine, Shihezi University, Shihezi, Xinjiang, People's Republic of China; Key Laboratory of Xinjiang Endemic and Ethnic Diseases (Ministry of Education), School of Medicine, Shihezi University, Shihezi, Xinjiang, People's Republic of China; NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, School of Medicine, Shihezi University, People's Republic of China
| | - Yanling Tang
- Department of Preventive Medicine, School of Medicine, Shihezi University, Shihezi, Xinjiang, People's Republic of China; Key Laboratory of Preventive Medicine, Shihezi University, Shihezi, Xinjiang, People's Republic of China; Key Laboratory of Xinjiang Endemic and Ethnic Diseases (Ministry of Education), School of Medicine, Shihezi University, Shihezi, Xinjiang, People's Republic of China; NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, School of Medicine, Shihezi University, People's Republic of China
| | - Zeyu Hu
- Department of Preventive Medicine, School of Medicine, Shihezi University, Shihezi, Xinjiang, People's Republic of China; Key Laboratory of Preventive Medicine, Shihezi University, Shihezi, Xinjiang, People's Republic of China; Key Laboratory of Xinjiang Endemic and Ethnic Diseases (Ministry of Education), School of Medicine, Shihezi University, Shihezi, Xinjiang, People's Republic of China; NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, School of Medicine, Shihezi University, People's Republic of China
| | - Wanjing Xu
- Department of Preventive Medicine, School of Medicine, Shihezi University, Shihezi, Xinjiang, People's Republic of China; Key Laboratory of Preventive Medicine, Shihezi University, Shihezi, Xinjiang, People's Republic of China; Key Laboratory of Xinjiang Endemic and Ethnic Diseases (Ministry of Education), School of Medicine, Shihezi University, Shihezi, Xinjiang, People's Republic of China; NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, School of Medicine, Shihezi University, People's Republic of China
| | - Yue Ma
- Department of Preventive Medicine, School of Medicine, Shihezi University, Shihezi, Xinjiang, People's Republic of China; Key Laboratory of Preventive Medicine, Shihezi University, Shihezi, Xinjiang, People's Republic of China; Key Laboratory of Xinjiang Endemic and Ethnic Diseases (Ministry of Education), School of Medicine, Shihezi University, Shihezi, Xinjiang, People's Republic of China; NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, School of Medicine, Shihezi University, People's Republic of China
| | - Panpan Xu
- Department of Preventive Medicine, School of Medicine, Shihezi University, Shihezi, Xinjiang, People's Republic of China; Key Laboratory of Preventive Medicine, Shihezi University, Shihezi, Xinjiang, People's Republic of China; Key Laboratory of Xinjiang Endemic and Ethnic Diseases (Ministry of Education), School of Medicine, Shihezi University, Shihezi, Xinjiang, People's Republic of China; NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, School of Medicine, Shihezi University, People's Republic of China
| | - Hengrui Xing
- Department of Preventive Medicine, School of Medicine, Shihezi University, Shihezi, Xinjiang, People's Republic of China; Key Laboratory of Preventive Medicine, Shihezi University, Shihezi, Xinjiang, People's Republic of China; Key Laboratory of Xinjiang Endemic and Ethnic Diseases (Ministry of Education), School of Medicine, Shihezi University, Shihezi, Xinjiang, People's Republic of China; NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, School of Medicine, Shihezi University, People's Republic of China
| | - Qiang Niu
- Department of Preventive Medicine, School of Medicine, Shihezi University, Shihezi, Xinjiang, People's Republic of China; Key Laboratory of Preventive Medicine, Shihezi University, Shihezi, Xinjiang, People's Republic of China; Key Laboratory of Xinjiang Endemic and Ethnic Diseases (Ministry of Education), School of Medicine, Shihezi University, Shihezi, Xinjiang, People's Republic of China; NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, School of Medicine, Shihezi University, People's Republic of China.
| |
Collapse
|
34
|
Tang Y, Kay A, Jiang Z, Arkin MR. LC3B Binds to the Autophagy Protease ATG4b with High Affinity Using a Bipartite Interface. Biochemistry 2022; 61:2295-2302. [PMID: 36264309 PMCID: PMC9631991 DOI: 10.1021/acs.biochem.2c00482] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 09/23/2022] [Indexed: 11/29/2022]
Abstract
Autophagy is a catabolic cellular process in which unwanted proteins and organelles are degraded by lysosomes. It is characterized by the formation of the double-membrane autophagosome decorated with LC3B, a protein that mediates autophagosomal fusion with lysosomes. The cysteine protease ATG4b acts at two stages in the life cycle of LC3B. We set out to characterize the protein-protein interaction between LC3B and ATG4b. Through biochemical and biophysical studies, we show that the ubiquitin-like core of LC3B (residues 1-115; "LC3B-115"), which lacks the C-terminal cleavage site (between residue 120 and 121), binds to full-length ATG4b with a surprisingly tight dissociation constant (KD) in the low nanomolar range; 10-30-fold tighter than that of the substrate pro-LC3B (residues 1-125) or the product LC3B-I (residues 1-120). Consequently, LC3B-115 is a potent inhibitor of the ATG4b-mediated cleavage of pro-LC3B (IC50 = 15 nM). Binding of the LC3B-115 has no effect on the conformation of the active site of ATG4b, as judged by the turnover of a peptide substrate ("substrate-33"), derived from LC3B-I residues 116-120. Conversely, truncations of ATG4b show that binding and proteolysis of LC3B critically depend on the C-terminal tail of ATG4b, whereas proteolysis of the peptide substrate-33 does not require the C-terminal tail of ATG4b. These results support a bipartite model for LC3B-ATG4b binding in which the core of LC3B binds to ATG4b and the C-terminal tail of pro-LC3B organizes the ATG4b active site; additionally, the C-terminal tail of ATG4b contributes at least 1000-fold higher binding affinity to the LC3B-ATG4b interaction and likely wraps around the LC3B-ubiquitin core. PPIs are often described as containing an energetic "hot spot" for binding; in the case of LC3B-ATG4b, however, the substrate-enzyme complex contains multiple, energetically relevant domains that differentially affect binding affinity and catalytic efficiency.
Collapse
Affiliation(s)
- Yinyan Tang
- Department
of Pharmaceutical Chemistry and Small Molecule Discovery Center, University of California, San Francisco, California 94158, United States
| | - Amber Kay
- Department
of Pharmaceutical Chemistry and Small Molecule Discovery Center, University of California, San Francisco, California 94158, United States
| | - Ziwen Jiang
- Department
of Pharmaceutical Chemistry and Small Molecule Discovery Center, University of California, San Francisco, California 94158, United States
| | - Michelle R. Arkin
- Department
of Pharmaceutical Chemistry and Small Molecule Discovery Center, University of California, San Francisco, California 94158, United States
| |
Collapse
|
35
|
Yifei sanjie Pills Alleviate Chemotherapy-Related Fatigue by Reducing Skeletal Muscle Injury and Inhibiting Tumor Growth in Lung Cancer Mice. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:2357616. [PMID: 36045663 PMCID: PMC9423986 DOI: 10.1155/2022/2357616] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 08/02/2022] [Indexed: 11/18/2022]
Abstract
Chemotherapy-related fatigue (CRF), one of the most severe adverse effects observed in cancer patients, has been theoretically related to oxidative stress, and antioxidant treatment might be one of the most valuable therapeutic approaches. However, there are still few effective pharmacological therapies. Yifei Sanjie pills (YFSJ), a classical formula used to treat lung cancer as complementary and alternative medicine, have been proved to alleviate CRF of lung cancer patients in clinical practices. However, the underlying mechanisms have not been clarified. In this study, our data showed that YFSJ alleviated CRF presented as reversing the decline of swimming time and locomotor activity induced by cisplatin (DDP). Moreover, YFSJ significantly reduces the accidence of mitophagy and mitochondrial damage and reduces apoptosis in skeletal muscle tissues caused by DDP. It probably works by decreasing the oxidative stress, inhibiting the activation of the AMPK/mTOR pathway, decreasing protein expression levels of Beclin1 and other autophagy-related proteins, and attenuating the activation of Cytochrome c (cyto. C), Cleaved Caspase-9 (c-Casp 9), and other apoptosis-related proteins. Furthermore, YFSJ enhanced DDP sensitivity by specifically promoting oxidative stress and activating apoptosis and autophagy in the tumor tissues of mice. It was also found that YFSJ reduced the loss of body weight caused by DDP, reversed the ascent of serum concentrations of alanine aminotransferase (ALT), aminotransferase (AST), and creatinine (CREA), increased the spleen index, and prolonged the survival time of mice. Taken together, these results revealed that YFSJ could alleviate CRF by reducing mitophagy and apoptosis induced by oxidative stress in skeletal muscle; these results also displayed the effects of YFSJ on enhancing chemotherapy sensitivity, improving quality of life, and prolonging survival time in lung cancer mice received DDP chemotherapy.
Collapse
|
36
|
Butyrate Inhibits Colorectal Cancer Cell Proliferation through Autophagy Degradation of β-Catenin Regardless of APC and β-Catenin Mutational Status. Biomedicines 2022; 10:biomedicines10051131. [PMID: 35625868 PMCID: PMC9138675 DOI: 10.3390/biomedicines10051131] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 05/09/2022] [Accepted: 05/11/2022] [Indexed: 02/04/2023] Open
Abstract
Colorectal cancer (CRC) pathogenesis is mainly driven by alterations in WNT signaling, which results in altered transcriptional activity of β-Catenin. Mutations in APC (Adenomatous Polyposis Coli) are reflected in β-Catenin hyperactivation and loss of proliferation control. Certain intestinal bacteria metabolites have shown the ability to limit CRC cell proliferation and CRC pathogenesis. Here, we investigated the molecular mechanism underlying the anti-proliferative activity of butyrate, a microbiota-derived short chain fatty acid, in two CRC cell lines, namely HCT116 and SW620, which bear a mutation in β-Catenin and APC, respectively. In particular, we focused on autophagy, a lysosome-dependent degradation pathway, which was shown to control intestinal tissue homeostasis. Butyrate reduced CRC cell proliferation, as witnessed by the downregulation of proliferation markers. TCGA bioinformatic transcriptomic analysis of CTNNB1 (β-Catenin) gene correlation in CRC patients showed that β-Catenin negatively correlates with the autophagy gene ATG4D. In CRC cells, regardless of the mutational state of APC or β-Catenin genes, butyrate caused the autophagy-mediated degradation of β-Catenin; thus, preventing its transcriptional activity. Autophagy gene silencing restored β-Catenin levels, allowing it to translocate into the nucleus to promote the expression of downstream genes associated with cancer cell proliferation. CRC-affected patients show driver mutations in the WNT pathway; thus, targeting its crucial effector may be a promising therapeutic strategy in CRC treatment; for instance, by using ad hoc probiotics that stimulate autophagy.
Collapse
|