1
|
Zhang X, Wang Y, Wang J, Zhang Y, Li R, Wang X, Ge X, Ye Q, Ji J, Fei D, Wang Q. Impaired stemness in aging periodontal ligament stem cells is mediated by the progerin/endoplasmic reticulum stress/p53 axis. J Adv Res 2024:S2090-1232(24)00484-3. [PMID: 39490613 DOI: 10.1016/j.jare.2024.10.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 10/05/2024] [Accepted: 10/23/2024] [Indexed: 11/05/2024] Open
Abstract
INTRODUCTION Decreased periodontal ligament stem cells (PDLSCs) stemness is a key factor in age-related alveolar bone loss. Endoplasmic reticulum (ER) stress is closely related to age-related diseases and the mesenchymal stem cell (MSC) stemness. However, the role of ER stress in regulating the stemness of senescent PDLSCs and its potential mechanism remain unclear. OBJECTIVES To investigate the detailed effect and mechanism of ER stress on impaired stemness in old periodontal ligament stem cells (OPDLSCs). METHODS The level of ER stress of Young PDLSCs (YPDLSCs) and OPDLSCs were detected, and ER stress was regulated to observe its effect on PDLSCs stemness. The expression levels of ER stress sensors (protein kinase R-like ER kinase (PERK), activating transcription factor 6 (ATF6), inositol requiring enzyme 1 (IRE1)) were upregulated in YPDLSCs and downregulated in OPDLSCs by transfection experiments to verify the detailed unfolded protein response (UPR) pathway. Mechanismly, the regulatory effect of UPR pathway on p53/p21 pathway was explored. Further study was performed to investigated the important role of progerin accumulation during aging process on ER stress, UPR and p53/p21 pathway. RESULTS Decreased stemness and ER stress activation were found in OPDLSCs. ER stress activation resulted in decreased stemness of YPDLSCs, while ER stress inhibition rescued compromised stemness of OPDLSCs. Mechanismly, ATF6 pathway regulated the OPDLSC stemness via the p53/p21 signaling as confirmed by transfection assay. Further study showed that progerin was accumulated in PDLSCs and progerin overexpression could resulted in ER stress activation, activating the ATF6/p53/p21 axis, leading to decreased stemness of aging PDLSCs. CONCLUSIONS Progerin accumulation during the aging process can lead to ER stress activation, which can suppress OPDLSC stemness via the ATF6/p53/p21 axis.
Collapse
Affiliation(s)
- Xige Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Department of Periodontology, School of Stomatology, The Fourth Military Medical University
| | - Yazheng Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Department of Periodontology, School of Stomatology, The Fourth Military Medical University
| | - Jinjin Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Department of Periodontology, School of Stomatology, The Fourth Military Medical University
| | - Yang Zhang
- Department of Stomatology, the Air Force Hospital from Eastern Theater, Nanjing, 210001, China
| | - Rui Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Department of Periodontology, School of Stomatology, The Fourth Military Medical University
| | - Xiaoyu Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Department of Periodontology, School of Stomatology, The Fourth Military Medical University
| | - Xiaotong Ge
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Department of Periodontology, School of Stomatology, The Fourth Military Medical University
| | - Qingyuan Ye
- State Key Laboratory of Military Stomatology& National Clinical Research Center for Oral Diseases& Shaanxi Clinical Research Center for Oral Diseases, Digital Dentistry Center, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Jiyun Ji
- Department of Stomatology, Hainan Hospital of Chinese PLA General Hospital, Sanya, 572000, Hainan, China
| | - Dongdong Fei
- Department of Stomatology, the Seventh Medical Center of PLA General Hospital, Beijing, 100700, China.
| | - Qintao Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Department of Periodontology, School of Stomatology, The Fourth Military Medical University.
| |
Collapse
|
2
|
Pissas G, Tziastoudi M, Divani M, Poulianiti C, Konsta MAP, Lykotsetas E, Liakopoulos V, Stefanidis I, Eleftheriadis T. Malate dehydrogenase-2 inhibition shields renal tubular epithelial cells from anoxia-reoxygenation injury by reducing reactive oxygen species. J Biochem Mol Toxicol 2024; 38:e23854. [PMID: 39287333 DOI: 10.1002/jbt.23854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 08/29/2024] [Accepted: 09/06/2024] [Indexed: 09/19/2024]
Abstract
Ischemia-reperfusion (I-R) injury is the most common cause of acute kidney injury. In experiments involving primary human renal proximal tubular epithelial cells (RPTECs) exposed to anoxia-reoxygenation, we explored the hypothesis that mitochondrial malate dehydrogenase-2 (MDH-2) inhibition redirects malate metabolism from the mitochondria to the cytoplasm, towards the malate-pyruvate cycle and reversed malate-aspartate shuttle. Colorimetry, fluorometry, and western blotting showed that MDH2 inhibition accelerates the malate-pyruvate cycle enhancing cytoplasmic NADPH, thereby regenerating the potent antioxidant reduced glutathione. It also reversed the malate-aspartate shuttle and potentially diminished mitochondrial reactive oxygen species (ROS) production by transferring electrons, in the form of NADH, from the mitochondria to the cytoplasm. The excessive ROS production induced by anoxia-reoxygenation led to DNA damage and protein modification, triggering DNA damage and unfolded protein response, ultimately resulting in apoptosis and senescence. Additionally, ROS induced lipid peroxidation, which may contribute to the process of ferroptosis. Inhibiting MDH-2 proved effective in mitigating ROS overproduction during anoxia-reoxygenation, thereby rescuing RPTECs from death or senescence. Thus, targeting MDH-2 holds promise as a pharmaceutical strategy against I-R injury.
Collapse
Affiliation(s)
- Georgios Pissas
- Department of Nephrology, Faculty of Medicine, University of Thessaly, Larissa, Greece
| | - Maria Tziastoudi
- Department of Nephrology, Faculty of Medicine, University of Thessaly, Larissa, Greece
| | - Maria Divani
- Department of Nephrology, Faculty of Medicine, University of Thessaly, Larissa, Greece
| | - Christina Poulianiti
- Department of Nephrology, Faculty of Medicine, University of Thessaly, Larissa, Greece
| | | | - Evangelos Lykotsetas
- Department of Nephrology, Faculty of Medicine, University of Thessaly, Larissa, Greece
| | - Vasilios Liakopoulos
- Department of Nephrology, Faculty of Medicine, University of Thessaly, Larissa, Greece
| | - Ioannis Stefanidis
- Department of Nephrology, Faculty of Medicine, University of Thessaly, Larissa, Greece
| | | |
Collapse
|
3
|
Li Q, Wang P, Gong Y, Xu M, Wang M, Luan R, Liu J, Li X, Shao Y. α-Klotho prevents diabetic retinopathy by reversing the senescence of macrophages. Cell Commun Signal 2024; 22:449. [PMID: 39327553 PMCID: PMC11426092 DOI: 10.1186/s12964-024-01838-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: 07/09/2024] [Accepted: 09/18/2024] [Indexed: 09/28/2024] Open
Abstract
BACKGROUND Diabetic retinopathy (DR) is a common microvascular complication of diabetes mellitus (DM) and a significant cause of acquired blindness in the working-age population worldwide. Aging is considered as an important risk factor for DR development. Macrophages in aged mice bear typical M2 marker proteins but simultaneously express a pro-inflammatory factor profile. This may explain why the level of intraocular inflammation does not decrease during proliferative diabetic retinopathy (PDR) despite the occurrence of neovascularization and fibrosis (M2 activation). α-Klotho (KL) was originally discovered as a soluble anti-aging factor, which is mainly expressed in kidney tubular epithelium, choroid plexus in the brain and secreted in the blood. However, the role of KL in DR pathophysiology has not been previously reported. METHODS Type 1 (streptozotocin [STZ]-induced) and type 2 (a high-fat diet along with a low dose of STZ) diabetic mouse models were established and injected with or without KL adenovirus via the tail vein for 12 weeks. Vldlr-/- mice were injected intravitreally with or without soluble KL protein from P8 to P15. The retinal structure and function were analyzed by electroretinogram and optical coherence tomography. The neovascular lesions were analyzed by retinal flat mount and RPE flat mount. The senescence markers, macrophage morphology, and KL expression levels were detected by immunofluorescence staining. A cell model was constructed using RAW264.7 cells stimulated by 4-hydroxynonenal (4HNE) and transfected with or without KL adenovirus. The senescence-associated secretory phenotypes were detected by qRT-PCR. Senescence was detected by SA-β-Gal staining. Serum, aqueous humor, and vitreous humor KL levels of proliferative diabetic retinopathy (PDR) patients were measured by enzyme-linked immunosorbent assay. Quantitative proteomics and bioinformatics were applied to predict the change of proteins and biological function after overexpression of KL in macrophages. The effects of KL on the HECTD1 binding to IRS1 were analyzed by bioinformatics, molecular docking, and Western Blot. RESULTS Serum, aqueous humor, and vitreous humor KL levels were lower in patients with PDR than in those with cataracts. KL relieved the retinal structure damage, improved retina function, and inhibited retinal senescence in diabetic mice. KL administration attenuated the neovascular lesions in VLDLR-/- mice by decreasing the secretion of VEGFA and FGF2 from macrophages. KL also protected RAW264.7 cells from 4HNE-induced senescence. Additionally, it inhibited E3 ubiquitin ligase HECTD1 expression in both diabetic mouse peripheral blood mononuclear cells and 4HNE-treated RAW264.7 cells. KL inhibited HECTD1 binding to IRS1 and reduced the ubiquitination of IRS1. CONCLUSIONS Macrophage aging is involved in DM-induced retinopathy. KL alleviates DM-induced retinal macrophage senescence by downregulating HECTD1 and decreasing IRS1 ubiquitination and degradation. Meanwhile, KL administration attenuated the neovascular lesions by altering the activation state of macrophages and decreasing the expression of VEGFA and FGF2.
Collapse
Affiliation(s)
- Qingbo Li
- Tianjin Key Laboratory of Retinal Functions and Diseases, Eye Institute & School of Optometry and Ophthalmology, Tianjin Medical University Eye Hospital, Nankai District, No. 251, Fukang Road, Tianjin, 300384, China
- Tianjin International Joint Research and Development Centre of Ophthalmology and Vision Science, Tianjin Medical University Eye Hospital, Nankai District, No. 251, Fukang Road, Tianjin, 300384, China
- Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Nankai District, No. 251, Fukang Road, Tianjin, 300384, China
| | - Peiyu Wang
- Tianjin Key Laboratory of Retinal Functions and Diseases, Eye Institute & School of Optometry and Ophthalmology, Tianjin Medical University Eye Hospital, Nankai District, No. 251, Fukang Road, Tianjin, 300384, China
- Tianjin International Joint Research and Development Centre of Ophthalmology and Vision Science, Tianjin Medical University Eye Hospital, Nankai District, No. 251, Fukang Road, Tianjin, 300384, China
- Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Nankai District, No. 251, Fukang Road, Tianjin, 300384, China
| | - Yi Gong
- Tianjin Key Laboratory of Retinal Functions and Diseases, Eye Institute & School of Optometry and Ophthalmology, Tianjin Medical University Eye Hospital, Nankai District, No. 251, Fukang Road, Tianjin, 300384, China
- Tianjin International Joint Research and Development Centre of Ophthalmology and Vision Science, Tianjin Medical University Eye Hospital, Nankai District, No. 251, Fukang Road, Tianjin, 300384, China
- Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Nankai District, No. 251, Fukang Road, Tianjin, 300384, China
| | - Manhong Xu
- Tianjin Key Laboratory of Retinal Functions and Diseases, Eye Institute & School of Optometry and Ophthalmology, Tianjin Medical University Eye Hospital, Nankai District, No. 251, Fukang Road, Tianjin, 300384, China
- Tianjin International Joint Research and Development Centre of Ophthalmology and Vision Science, Tianjin Medical University Eye Hospital, Nankai District, No. 251, Fukang Road, Tianjin, 300384, China
- Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Nankai District, No. 251, Fukang Road, Tianjin, 300384, China
| | - Manqiao Wang
- Tianjin Key Laboratory of Retinal Functions and Diseases, Eye Institute & School of Optometry and Ophthalmology, Tianjin Medical University Eye Hospital, Nankai District, No. 251, Fukang Road, Tianjin, 300384, China
- Tianjin International Joint Research and Development Centre of Ophthalmology and Vision Science, Tianjin Medical University Eye Hospital, Nankai District, No. 251, Fukang Road, Tianjin, 300384, China
- Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Nankai District, No. 251, Fukang Road, Tianjin, 300384, China
| | - Rong Luan
- Tianjin Key Laboratory of Retinal Functions and Diseases, Eye Institute & School of Optometry and Ophthalmology, Tianjin Medical University Eye Hospital, Nankai District, No. 251, Fukang Road, Tianjin, 300384, China
- Tianjin International Joint Research and Development Centre of Ophthalmology and Vision Science, Tianjin Medical University Eye Hospital, Nankai District, No. 251, Fukang Road, Tianjin, 300384, China
- Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Nankai District, No. 251, Fukang Road, Tianjin, 300384, China
| | - Juping Liu
- Tianjin Key Laboratory of Retinal Functions and Diseases, Eye Institute & School of Optometry and Ophthalmology, Tianjin Medical University Eye Hospital, Nankai District, No. 251, Fukang Road, Tianjin, 300384, China.
- Tianjin International Joint Research and Development Centre of Ophthalmology and Vision Science, Tianjin Medical University Eye Hospital, Nankai District, No. 251, Fukang Road, Tianjin, 300384, China.
- Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Nankai District, No. 251, Fukang Road, Tianjin, 300384, China.
| | - Xiaorong Li
- Tianjin Key Laboratory of Retinal Functions and Diseases, Eye Institute & School of Optometry and Ophthalmology, Tianjin Medical University Eye Hospital, Nankai District, No. 251, Fukang Road, Tianjin, 300384, China.
- Tianjin International Joint Research and Development Centre of Ophthalmology and Vision Science, Tianjin Medical University Eye Hospital, Nankai District, No. 251, Fukang Road, Tianjin, 300384, China.
- Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Nankai District, No. 251, Fukang Road, Tianjin, 300384, China.
| | - Yan Shao
- Tianjin Key Laboratory of Retinal Functions and Diseases, Eye Institute & School of Optometry and Ophthalmology, Tianjin Medical University Eye Hospital, Nankai District, No. 251, Fukang Road, Tianjin, 300384, China.
- Tianjin International Joint Research and Development Centre of Ophthalmology and Vision Science, Tianjin Medical University Eye Hospital, Nankai District, No. 251, Fukang Road, Tianjin, 300384, China.
- Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Nankai District, No. 251, Fukang Road, Tianjin, 300384, China.
- University of Tibetan Medicine, Lhasa, 850000, China.
| |
Collapse
|
4
|
Teng M, Wu TJ, Jing X, Day BW, Pritchard KA, Naylor S, Teng RJ. Temporal Dynamics of Oxidative Stress and Inflammation in Bronchopulmonary Dysplasia. Int J Mol Sci 2024; 25:10145. [PMID: 39337630 PMCID: PMC11431892 DOI: 10.3390/ijms251810145] [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: 07/30/2024] [Revised: 09/04/2024] [Accepted: 09/20/2024] [Indexed: 09/30/2024] Open
Abstract
Bronchopulmonary dysplasia (BPD) is the most common lung complication of prematurity. Despite extensive research, our understanding of its pathophysiology remains limited, as reflected by the stable prevalence of BPD. Prematurity is the primary risk factor for BPD, with oxidative stress (OS) and inflammation playing significant roles and being closely linked to premature birth. Understanding the interplay and temporal relationship between OS and inflammation is crucial for developing new treatments for BPD. Animal studies suggest that OS and inflammation can exacerbate each other. Clinical trials focusing solely on antioxidants or anti-inflammatory therapies have been unsuccessful. In contrast, vitamin A and caffeine, with antioxidant and anti-inflammatory properties, have shown some efficacy, reducing BPD by about 10%. However, more than one-third of very preterm infants still suffer from BPD. New therapeutic agents are needed. A novel tripeptide, N-acetyl-lysyltyrosylcysteine amide (KYC), is a reversible myeloperoxidase inhibitor and a systems pharmacology agent. It reduces BPD severity by inhibiting MPO, enhancing antioxidative proteins, and alleviating endoplasmic reticulum stress and cellular senescence in a hyperoxia rat model. KYC represents a promising new approach to BPD treatment.
Collapse
Affiliation(s)
- Michelle Teng
- Department of Pediatrics, Medical College of Wisconsin, Suite C410, Children Corporate Center, 999N 92nd Street, Milwaukee, WI 53226, USA; (M.T.); (T.-J.W.); (X.J.)
- Children’s Research Institute, Medical College of Wisconsin, 8701 W Watertown Plank Rd., Wauwatosa, WI 53226, USA;
| | - Tzong-Jin Wu
- Department of Pediatrics, Medical College of Wisconsin, Suite C410, Children Corporate Center, 999N 92nd Street, Milwaukee, WI 53226, USA; (M.T.); (T.-J.W.); (X.J.)
- Children’s Research Institute, Medical College of Wisconsin, 8701 W Watertown Plank Rd., Wauwatosa, WI 53226, USA;
| | - Xigang Jing
- Department of Pediatrics, Medical College of Wisconsin, Suite C410, Children Corporate Center, 999N 92nd Street, Milwaukee, WI 53226, USA; (M.T.); (T.-J.W.); (X.J.)
- Children’s Research Institute, Medical College of Wisconsin, 8701 W Watertown Plank Rd., Wauwatosa, WI 53226, USA;
| | - Billy W. Day
- ReNeuroGen LLC, 2160 San Fernando Dr., Elm Grove, WI 53122, USA; (B.W.D.); (S.N.)
| | - Kirkwood A. Pritchard
- Children’s Research Institute, Medical College of Wisconsin, 8701 W Watertown Plank Rd., Wauwatosa, WI 53226, USA;
- ReNeuroGen LLC, 2160 San Fernando Dr., Elm Grove, WI 53122, USA; (B.W.D.); (S.N.)
- Department of Surgery, Medical College of Wisconsin, 8701 Watertown Plank Rd., Milwaukee, WI 53226, USA
| | - Stephen Naylor
- ReNeuroGen LLC, 2160 San Fernando Dr., Elm Grove, WI 53122, USA; (B.W.D.); (S.N.)
| | - Ru-Jeng Teng
- Department of Pediatrics, Medical College of Wisconsin, Suite C410, Children Corporate Center, 999N 92nd Street, Milwaukee, WI 53226, USA; (M.T.); (T.-J.W.); (X.J.)
- Children’s Research Institute, Medical College of Wisconsin, 8701 W Watertown Plank Rd., Wauwatosa, WI 53226, USA;
| |
Collapse
|
5
|
Bai M, Lei J, Li F, Wang X, Fu H, Yan Z, Huang X, Zhu Y. Short-chain chlorinated paraffins may induce thymic aging in mice by activating PERK-CHOP. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 357:124438. [PMID: 38942270 DOI: 10.1016/j.envpol.2024.124438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 06/10/2024] [Accepted: 06/23/2024] [Indexed: 06/30/2024]
Abstract
Humans indirectly consume approximately 0.02 mg/kg/day of short-chained chlorinated paraffins (SCCPs) through the environment; however, the thymic senescence/damage induced by SCCPs has not been assessed. In this study, 16 female mice (4-week-old) per group were orally administered 0, 0.01, 0.1, and 1 mg/kg/day of SCCPs for 21 days, and the phenotypes and levels of superoxide dismutase (SOD), malondialdehyde (MDA), Tβ4, αβ TCR, SA-β-Gal, GRP78, PERK/CHOP, P53/P21, and CASPASE-1 of the thymus were assessed as indicators. Another group comprising 16 mice was killed at 4-week-old and these indicators were assessed. Thereafter, the thymuses cultured in vitro were exposed to 0, 14, 140, and 1400 μg/L SCCPs, respectively, and the above indicators were measured after 7-day. Based on the results, the oral administration of ≥0.01 mg/kg/day SCCPs to mice and ≥14 μg/L of SCCPs in medium caused thymic aging features, such as a decrease in the ratio of cortex to medulla, gradual blurring of the boundary between the cortex and medulla, dose-dependent oxidative stress (decreased SOD and increased MDA), and decreased levels of Tβ4 and αβ TCRs in the thymus. The oral administration of ≥1 mg/kg/day of SCCPs also impeded the growth and development of female mice and their thymuses. Exposure to the low levels of SCCPs activated PERK-CHOP in the mouse thymus, which modulated increases in SA-β-Gal, IL-1β, P53, and CASPASE-1 in vivo and in vitro. Overall, environmental levels and human blood concentrations (14.8-1400 μg/L) of SCCPs may induce mouse thymus senescence by activating PERK-CHOP in vivo and in vitro, respectively.
Collapse
Affiliation(s)
- Mingxin Bai
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Department of Preventive Medicine, Medical School, Hunan Normal University, Changsha, 410013, Hunan, China
| | - Jiawei Lei
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Department of Preventive Medicine, Medical School, Hunan Normal University, Changsha, 410013, Hunan, China
| | - Fan Li
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Department of Preventive Medicine, Medical School, Hunan Normal University, Changsha, 410013, Hunan, China
| | - Xuning Wang
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Department of Preventive Medicine, Medical School, Hunan Normal University, Changsha, 410013, Hunan, China
| | - Hu Fu
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Department of Preventive Medicine, Medical School, Hunan Normal University, Changsha, 410013, Hunan, China; Key Laboratory of Protein Chemistry and Fish Developmental Biology of Ministry of Education, Hunan Normal University, Changsha, 410081, China
| | - Zhengli Yan
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Department of Preventive Medicine, Medical School, Hunan Normal University, Changsha, 410013, Hunan, China; Key Laboratory of Protein Chemistry and Fish Developmental Biology of Ministry of Education, Hunan Normal University, Changsha, 410081, China
| | - Xin Huang
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Department of Preventive Medicine, Medical School, Hunan Normal University, Changsha, 410013, Hunan, China.
| | - Yongfei Zhu
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Department of Preventive Medicine, Medical School, Hunan Normal University, Changsha, 410013, Hunan, China; Key Laboratory of Protein Chemistry and Fish Developmental Biology of Ministry of Education, Hunan Normal University, Changsha, 410081, China.
| |
Collapse
|
6
|
Chilosi M, Piciucchi S, Ravaglia C, Spagnolo P, Sverzellati N, Tomassetti S, Wuyts W, Poletti V. "Alveolar stem cell exhaustion, fibrosis and bronchiolar proliferation" related entities. A narrative review. Pulmonology 2024:S2531-0437(24)00092-8. [PMID: 39277539 DOI: 10.1016/j.pulmoe.2024.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 05/11/2024] [Accepted: 05/27/2024] [Indexed: 09/17/2024] Open
Affiliation(s)
- M Chilosi
- Department of Medical Specialities/Pulmonology Ospedale GB Morgagni, Forlì I
| | - S Piciucchi
- Department of Radiology, Ospedale GB Morgagni, Forlì I.
| | - C Ravaglia
- Department of Medical Specialities/Pulmonology Ospedale GB Morgagni, Forlì (I); DIMEC, Bologna University, Forlì Campus, Forlì I, Department
| | - P Spagnolo
- Respiratory Disease Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - N Sverzellati
- Scienze Radiologiche, Department of Medicine and Surgery, University Hospital Parma, Parma, Italy
| | - S Tomassetti
- Department of Experimental and Clinical Medicine, Careggi University Hospital, Florence, Italy
| | - W Wuyts
- Pulmonology Department, UZ Leuven, Leuven, Belgium
| | - V Poletti
- Department of Medical Specialities/Pulmonology Ospedale GB Morgagni, Forlì (I); DIMEC, Bologna University, Forlì Campus, Forlì I, Department; Department of Respiratory Diseases & Allergy, Aarhus University, Aarhus, Denmark
| |
Collapse
|
7
|
Mohamed AAA, Soliman SS, Soliman ASH, Hanafy A, Jin Y. Endoplasmic reticulum stress is involved in mycotoxin zearalenone induced inflammatory response, proliferation, and apoptosis in goat endometrial stromal cells. Reprod Biol 2024; 24:100948. [PMID: 39232304 DOI: 10.1016/j.repbio.2024.100948] [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: 06/09/2024] [Revised: 08/26/2024] [Accepted: 08/27/2024] [Indexed: 09/06/2024]
Abstract
Zearalenone (ZEA) is an estrogen-like mycotoxin and is considered a secondary metabolite produced by Fusarium fungi, which are widely found in the surrounding environment. ZEA has been found to cause reproductive dysfunction in female and male animals, but the underlying mechanism remains unclear. Therefore, this study examined cell proliferation, cell apoptosis, autophagy protein expression, and some inflammatory cytokines such as IL-1β and IL-8 of goat endometrial stromal cells (ESCs) induced by different concentrations (0, 15, 30, 60, and 90 µM) of ZEA. The apoptosis rate was detected by flow cytometry. Western Blot and ELISA assay were used to identify the ER stress signaling pathway and some inflammatory cytokines. Our results revealed that ZEA induced cell proliferation and inhibited cell apoptosis at low and middle concentrations, while at high concentrations of ZEA, cell apoptosis was induced in ESCs. Additionally, ZEA induced the ER stress protein markers such as ATF6, IRE1α, EIF2α, and ATF4. LC3 as a marker of autophagy was up-regulated at all concentrations of ZEA. Moreover, IL-1β and IL-8 showed down-regulation at a low concentration of ZEA, but middle and high concentrations showed up-regulation. In the present study, Knockdown ERN1 can inhibit autophagy and the main markers of ER stress. These results suggest that the IRE1 pathway can reduce apoptosis protein markers, down activate IRE1, and unfolded protein response branches such as ATF6 and LC3 in ESCs. Additionally, IL-1β and IL-8 achieve up-regulation under knockdown IRE1, which can block ER stress markers.
Collapse
Affiliation(s)
- Amira Abdalla Abdelshafy Mohamed
- College of Veterinary Medicine, Northwest Agriculture and Forestry University, Yangling 712100, China; Department of Animal Production, Faculty of Environmental Agricultural Sciences, Arish University, Al-Arish, North-Sinai 45511, Egypt.
| | - Seham Samir Soliman
- Department of Animal Reproduction and Artificial Insemination, Veterinary Research Institute, National Research Centre (NRC), Dokki, Cairo 12622, Egypt
| | - Ahmed S H Soliman
- Department of Animal Production, Faculty of Agriculture, New Vally University, Al kharga city, New Vally, Egypt
| | - Ahmed Hanafy
- Department of Animal Production, Faculty of Agricultural, Suez Canal University, Ismalilia 41522, Egypt
| | - Yaping Jin
- College of Veterinary Medicine, Northwest Agriculture and Forestry University, Yangling 712100, China.
| |
Collapse
|
8
|
Tufail M, Huang YQ, Hu JJ, Liang J, He CY, Wan WD, Jiang CH, Wu H, Li N. Cellular Aging and Senescence in Cancer: A Holistic Review of Cellular Fate Determinants. Aging Dis 2024:AD.2024.0421. [PMID: 38913050 DOI: 10.14336/ad.2024.0421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Accepted: 05/21/2024] [Indexed: 06/25/2024] Open
Abstract
This comprehensive review navigates the complex relationship between cellular aging, senescence, and cancer, unraveling the determinants of cellular fate. Beginning with an overview of cellular aging's significance in cancer, the review explores processes, changes, and molecular pathways influencing senescence. The review explores senescence as a dual mechanism in cancer, acting as a suppressor and contributor, focusing on its impact on therapy response. This review highlights opportunities for cancer therapies that target cellular senescence. The review further examines the senescence-associated secretory phenotype and strategies to modulate cellular aging to influence tumor behavior. Additionally, the review highlights the mechanisms of senescence escape in aging and cancer cells, emphasizing their impact on cancer prognosis and resistance to therapy. The article addresses current advances, unexplored aspects, and future perspectives in understanding cellular aging and senescence in cancer.
Collapse
Affiliation(s)
- Muhammad Tufail
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China
| | - Yu-Qi Huang
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China
| | - Jia-Ju Hu
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China
| | - Jie Liang
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China
| | - Cai-Yun He
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China
| | - Wen-Dong Wan
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China
| | - Can-Hua Jiang
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China
- Institute of Oral Precancerous Lesions, Central South University, Changsha, China
- Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Hong Wu
- State Key Laboratory of Powder Metallurgy, Central South University, Changsha, 410083, China
| | - Ning Li
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China
- Institute of Oral Precancerous Lesions, Central South University, Changsha, China
- Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
9
|
Deshetty UM, Chatterjee N, Buch S, Periyasamy P. HIV-1 Tat-Mediated Human Müller Glial Cell Senescence Involves Endoplasmic Reticulum Stress and Dysregulated Autophagy. Viruses 2024; 16:903. [PMID: 38932195 PMCID: PMC11209317 DOI: 10.3390/v16060903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 05/25/2024] [Accepted: 05/27/2024] [Indexed: 06/28/2024] Open
Abstract
Antiretroviral treatments have notably extended the lives of individuals with HIV and reduced the occurrence of comorbidities, including ocular manifestations. The involvement of endoplasmic reticulum (ER) stress in HIV-1 pathogenesis raises questions about its correlation with cellular senescence or its role in initiating senescent traits. This study investigated how ER stress and dysregulated autophagy impact cellular senescence triggered by HIV-1 Tat in the MIO-M1 cell line (human Müller glial cells). Cells exposed to HIV-1 Tat exhibited increased vimentin expression combined with markers of ER stress (BiP, p-eIF2α), autophagy (LC3, Beclin-1, p62), and the senescence marker p21 compared to control cells. Western blotting and staining techniques like SA-β-gal were employed to examine these markers. Additionally, treatments with ER stress inhibitor 4-PBA before HIV-1 Tat exposure led to a decreased expression of ER stress, senescence, and autophagy markers. Conversely, pre-treatment with the autophagy inhibitor 3-MA resulted in reduced autophagy and senescence markers but did not alter ER stress markers compared to control cells. The findings suggest a link between ER stress, dysregulated autophagy, and the initiation of a senescence phenotype in MIO-M1 cells induced by HIV-1 Tat exposure.
Collapse
Affiliation(s)
- Uma Maheswari Deshetty
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880, USA;
| | - Nivedita Chatterjee
- Vision Research Foundation, Sankara Netralaya, 18, College Road, Chennai 600006, India;
| | - Shilpa Buch
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880, USA;
| | - Palsamy Periyasamy
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880, USA;
| |
Collapse
|
10
|
Hemagirri M, Chen Y, Gopinath SCB, Sahreen S, Adnan M, Sasidharan S. Crosstalk between protein misfolding and endoplasmic reticulum stress during ageing and their role in age-related disorders. Biochimie 2024; 221:159-181. [PMID: 37918463 DOI: 10.1016/j.biochi.2023.10.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 10/25/2023] [Accepted: 10/30/2023] [Indexed: 11/04/2023]
Abstract
Maintaining the proteome is crucial to retaining cell functionality and response to multiple intrinsic and extrinsic stressors. Protein misfolding increased the endoplasmic reticulum (ER) stress and activated the adaptive unfolded protein response (UPR) to restore cell homeostasis. Apoptosis occurs when ER stress is prolonged or the adaptive response fails. In healthy young cells, the ratio of protein folding machinery to quantities of misfolded proteins is balanced under normal circumstances. However, the age-related deterioration of the complex systems for handling protein misfolding is accompanied by ageing-related disruption of protein homeostasis, which results in the build-up of misfolded and aggregated proteins. This ultimately results in decreased cell viability and forms the basis of common age-related diseases called protein misfolding diseases. Proteins or protein fragments convert from their ordinarily soluble forms to insoluble fibrils or plaques in many of these disorders, which build up in various organs such as the liver, brain, or spleen. Alzheimer's, Parkinson's, type II diabetes, and cancer are diseases in this group commonly manifest in later life. Thus, protein misfolding and its prevention by chaperones and different degradation paths are becoming understood from molecular perspectives. Proteodynamics information will likely affect future interventional techniques to combat cellular stress and support healthy ageing by avoiding and treating protein conformational disorders. This review provides an overview of the diverse proteostasis machinery, protein misfolding, and ER stress involvement, which activates the UPR sensors. Here, we will discuss the crosstalk between protein misfolding and ER stress and their role in developing age-related diseases.
Collapse
Affiliation(s)
- Manisekaran Hemagirri
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, USM, 11800, Pulau Pinang, Malaysia
| | - Yeng Chen
- Department of Oral & Craniofacial Sciences, Faculty of Dentistry, University of Malaya, Kuala Lumpur, 50603, Malaysia
| | - Subash C B Gopinath
- Faculty of Chemical Engineering and Technology, Universiti Malaysia Perlis, Arau, 02600, Malaysia
| | - Sumaira Sahreen
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, USM, 11800, Pulau Pinang, Malaysia
| | - Mohd Adnan
- Department of Biology, College of Science, University of Ha'il, Ha'il, P. O. Box 2440, Saudi Arabia.
| | - Sreenivasan Sasidharan
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, USM, 11800, Pulau Pinang, Malaysia.
| |
Collapse
|
11
|
Harada M. Cellular senescence in the pathogenesis of ovarian dysfunction. J Obstet Gynaecol Res 2024; 50:800-808. [PMID: 38412992 DOI: 10.1111/jog.15918] [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: 02/01/2024] [Accepted: 02/17/2024] [Indexed: 02/29/2024]
Abstract
The follicular microenvironment is crucial for normal ovarian function, and intra-ovarian factors, in coordination with gonadotropins, contribute to its regulation. Recent research has revealed that the accumulation of senescent cells worsens the adverse environment of various tissues and plays critical roles in chronological aging and various pathological conditions. Cellular senescence involves cell-cycle arrest, a senescence-associated secretory phenotype (SASP), macromolecular damage, and dysmetabolism. In this review, I summarize the latest knowledge regarding the role of cellular senescence in pathological conditions in the ovary, in the context of reproduction. Specifically, cellular senescence is known to impair follicular and oocyte health in cisplatin- and cyclophosphamide-induced primary ovarian insufficiency and to contribute to the pathogenesis of polycystic ovary syndrome (PCOS). In addition, cellular senescence is induced during the decline in ovarian reserve that is associated with chronological aging, endometriosis, psychological stress, and obesity, but it remains unclear whether it plays a causative role in these conditions. Finally, I discuss the potential for use of cellular senescence as a novel therapeutic target. The modification of SASP using a senomorphic and/or the elimination of senescent cells using a senolytic represent promising therapeutic strategies. Further elucidation of the role of cellular senescence in the effects of various insults on ovarian reserve, including chronological aging, as well as in pathogenesis of ovarian pathologies, including PCOS, may facilitate a new era of reproductive medicine.
Collapse
Affiliation(s)
- Miyuki Harada
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
12
|
García-López D, Zaragoza-Ojeda M, Eguía-Aguilar P, Arenas-Huertero F. Endoplasmic Reticulum Stress in Gliomas: Exploiting a Dual-Effect Dysfunction through Chemical Pharmaceutical Compounds and Natural Derivatives for Therapeutical Uses. Int J Mol Sci 2024; 25:4078. [PMID: 38612890 PMCID: PMC11012637 DOI: 10.3390/ijms25074078] [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: 10/31/2023] [Revised: 01/09/2024] [Accepted: 01/10/2024] [Indexed: 04/14/2024] Open
Abstract
The endoplasmic reticulum maintains proteostasis, which can be disrupted by oxidative stress, nutrient deprivation, hypoxia, lack of ATP, and toxicity caused by xenobiotic compounds, all of which can result in the accumulation of misfolded proteins. These stressors activate the unfolded protein response (UPR), which aims to restore proteostasis and avoid cell death. However, endoplasmic response-associated degradation (ERAD) is sometimes triggered to degrade the misfolded and unassembled proteins instead. If stress persists, cells activate three sensors: PERK, IRE-1, and ATF6. Glioma cells can use these sensors to remain unresponsive to chemotherapeutic treatments. In such cases, the activation of ATF4 via PERK and some proteins via IRE-1 can promote several types of cell death. The search for new antitumor compounds that can successfully and directly induce an endoplasmic reticulum stress response ranges from ligands to oxygen-dependent metabolic pathways in the cell capable of activating cell death pathways. Herein, we discuss the importance of the ER stress mechanism in glioma and likely therapeutic targets within the UPR pathway, as well as chemicals, pharmaceutical compounds, and natural derivatives of potential use against gliomas.
Collapse
Affiliation(s)
- Daniel García-López
- Laboratorio de Investigación en Patología Experimental, Hospital Infantil de México Federico Gómez, Mexico City 06720, Mexico; (D.G.-L.); (M.Z.-O.); (P.E.-A.)
- Facultad de Ciencia y Tecnología, Universidad Simón Bolívar, Mexico City 03920, Mexico
| | - Montserrat Zaragoza-Ojeda
- Laboratorio de Investigación en Patología Experimental, Hospital Infantil de México Federico Gómez, Mexico City 06720, Mexico; (D.G.-L.); (M.Z.-O.); (P.E.-A.)
| | - Pilar Eguía-Aguilar
- Laboratorio de Investigación en Patología Experimental, Hospital Infantil de México Federico Gómez, Mexico City 06720, Mexico; (D.G.-L.); (M.Z.-O.); (P.E.-A.)
- Departamento de Patología Clínica y Experimental, Hospital Infantil de México Federico Gómez, Mexico City 06720, Mexico
| | - Francisco Arenas-Huertero
- Laboratorio de Investigación en Patología Experimental, Hospital Infantil de México Federico Gómez, Mexico City 06720, Mexico; (D.G.-L.); (M.Z.-O.); (P.E.-A.)
- Centro de Investigación en Biomedicina y Bioseguridad, Hospital Infantil de México Federico Gómez, Mexico City 06720, Mexico
| |
Collapse
|
13
|
Rodzinski É, Martin N, Rouget R, Pioger A, Dehennaut V, Molendi-Coste O, Dombrowicz D, Goy E, de Launoit Y, Abbadie C. [Sorting of senescent cells by flow cytometry: Specificities and pitfalls to avoid]. Med Sci (Paris) 2024; 40:275-282. [PMID: 38520103 DOI: 10.1051/medsci/2024011] [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: 03/25/2024] Open
Abstract
Cells can be reprogrammed into senescence to adapt to a variety of stresses, most often affecting the genome integrity. Senescent cells accumulate with age or upon various insults in almost all tissues, and contribute to the development of several age-associated pathologies. Studying the molecular pathways involved in senescence induction, maintenance, or escape is challenged by the heterogeneity in the level of commitment to senescence, and by the pollution of senescent cell populations by proliferating pre- or post-senescent cells. We coped with these difficulties by developing a protocol for sorting senescent cells by flow cytometry, based on three major senescence markers : the SA-β-Galactosidase activity, the size of the cells, and their granularity reflecting the accumulation of aggregates, lysosomes, and altered mitochondria. We address the issues related to sorting senescent cells, the pitfalls to avoid, and propose solutions for sorting viable cells expressing senescent markers at different extents.
Collapse
Affiliation(s)
- Élodie Rodzinski
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277 - CANTHER (Cancer Heterogeneity Plasticity and Resistance to Therapies), F-59000 Lille, France
| | - Nathalie Martin
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277 - CANTHER (Cancer Heterogeneity Plasticity and Resistance to Therapies), F-59000 Lille, France
| | - Raphael Rouget
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277 - CANTHER (Cancer Heterogeneity Plasticity and Resistance to Therapies), F-59000 Lille, France
| | - Adrien Pioger
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277 - CANTHER (Cancer Heterogeneity Plasticity and Resistance to Therapies), F-59000 Lille, France
| | - Vanessa Dehennaut
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277 - CANTHER (Cancer Heterogeneity Plasticity and Resistance to Therapies), F-59000 Lille, France
| | - Olivier Molendi-Coste
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, US41 - UAR 2014 - PLBS, F-59000 Lille, France
| | - David Dombrowicz
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011 - EGID, F-59000 Lille, France
| | - Erwan Goy
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277 - CANTHER (Cancer Heterogeneity Plasticity and Resistance to Therapies), F-59000 Lille, France
| | - Yvan de Launoit
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277 - CANTHER (Cancer Heterogeneity Plasticity and Resistance to Therapies), F-59000 Lille, France
| | - Corinne Abbadie
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277 - CANTHER (Cancer Heterogeneity Plasticity and Resistance to Therapies), F-59000 Lille, France
| |
Collapse
|
14
|
Hellani F, Leleu I, Saidi N, Martin N, Lecoeur C, Werkmeister E, Koffi D, Trottein F, Yapo-Etté H, Das B, Abbadie C, Pied S. Role of astrocyte senescence regulated by the non- canonical autophagy in the neuroinflammation associated to cerebral malaria. Brain Behav Immun 2024; 117:20-35. [PMID: 38157948 DOI: 10.1016/j.bbi.2023.12.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 12/12/2023] [Accepted: 12/21/2023] [Indexed: 01/03/2024] Open
Abstract
BACKGROUND Cerebral malaria (CM) is a fatal neuroinflammatory syndrome caused (in humans) by the protozoa Plasmodium (P.) falciparum. Glial cell activation is one of the mechanisms that contributes to neuroinflammation in CM. RESULT By studying a mouse model of CM (caused by P. berghei ANKA), we describe that the induction of autophagy promoted p21-dependent senescence in astrocytes and that CXCL-10 was part of the senescence-associated secretory phenotype. Furthermore, p21 expression was observed in post-mortem brain and peripheral blood samples from patients with CM. Lastly, we found that the depletion of senescent astrocytes with senolytic drugs abrogated inflammation and protected mice from CM. CONCLUSION Our data provide evidence for a novel mechanism through which astrocytes could be involved in the neuropathophysiology of CM. p21 gene expression in blood cell and an elevated plasma CXCL-10 concentration could be valuable biomarkers of CM in humans. In the end, we believe senolytic drugs shall open up new avenues to develop newer treatment options.
Collapse
Affiliation(s)
- Fatima Hellani
- Univ. Lille, CNRS UMR 9017-INSERM U1019, Center for Infection and Immunity of Lille-CIIL, Institut Pasteur de Lille F-59019 Lille, France
| | - Inès Leleu
- Univ. Lille, CNRS UMR 9017-INSERM U1019, Center for Infection and Immunity of Lille-CIIL, Institut Pasteur de Lille F-59019 Lille, France
| | - Nasreddine Saidi
- Univ. Lille, CNRS UMR 9017-INSERM U1019, Center for Infection and Immunity of Lille-CIIL, Institut Pasteur de Lille F-59019 Lille, France
| | - Nathalie Martin
- Univ. Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity, Plasticity and Resistance to Therapies F-59000 Lille, France
| | - Cécile Lecoeur
- Univ. Lille, CNRS UMR 9017-INSERM U1019, Center for Infection and Immunity of Lille-CIIL, Institut Pasteur de Lille F-59019 Lille, France
| | - Elisabeth Werkmeister
- Univ. Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, US 41 - UMS 2014 - PLBS F-59000 Lille, France
| | - David Koffi
- Parasitology and Mycology Department, Institut Pasteur de Côte d'Ivoire, Ivory Coast
| | - François Trottein
- Univ. Lille, CNRS UMR 9017-INSERM U1019, Center for Infection and Immunity of Lille-CIIL, Institut Pasteur de Lille F-59019 Lille, France
| | - Hélène Yapo-Etté
- Institute of Forensic Medicine-Faculty of Health, University Félix Houphouët-Boigny of Abidjan, Ivory Coast
| | - Bidyut Das
- SCB Medical College, Cuttack, Orissa, India
| | - Corinne Abbadie
- Univ. Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity, Plasticity and Resistance to Therapies F-59000 Lille, France
| | - Sylviane Pied
- Univ. Lille, CNRS UMR 9017-INSERM U1019, Center for Infection and Immunity of Lille-CIIL, Institut Pasteur de Lille F-59019 Lille, France.
| |
Collapse
|
15
|
Zhao X, Liu D, Zhao Y, Wang Z, Wang Y, Chen Z, Ning S, Wang G, Meng L, Yao J, Tian X. HRD1-induced TMEM2 ubiquitination promotes ER stress-mediated apoptosis through a non-canonical pathway in intestinal ischemia/reperfusion. Cell Death Dis 2024; 15:154. [PMID: 38378757 PMCID: PMC10879504 DOI: 10.1038/s41419-024-06504-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 01/24/2024] [Accepted: 01/26/2024] [Indexed: 02/22/2024]
Abstract
Intestinal ischemia/reperfusion (I/R) injury is a typical pathological course in the clinic with a high morbidity rate. Recent research has pointed out the critical role of ubiquitination during the occurrence and development of intestinal I/R by precisely mediating protein quality control and function. Here, we conducted an integrated multiomic analysis to identify critical ubiquitination-associated molecules in intestinal I/R and identified endoplasmic reticulum-located HRD1 as a candidate molecule. During intestinal I/R, excessive ER stress plays a central role by causing apoptotic pathway activation. In particular, we found that ER stress-mediated apoptosis was mitigated by HRD1 knockdown in intestinal I/R mice. Mechanistically, TMEM2 was identified as a new substrate of HRD1 in intestinal I/R by mass spectrometry analysis, which has a crucial role in attenuating apoptosis and promoting non-canonical ER stress resistance. A strong negative correlation was found between the protein levels of HRD1 and TMEM2 in human intestinal ischemia samples. Specifically, HRD1 interacted with the lysine 42 residue of TMEM2 and reduced its stabilization by K48-linked polyubiquitination. Furthermore, KEGG pathway analysis revealed that TMEM2 regulated ER stress-mediated apoptosis in association with the PI3k/Akt signaling pathway rather than canonical ER stress pathways. In summary, HRD1 regulates ER stress-mediated apoptosis through a non-canonical pathway by ubiquitinating TMEM2 and inhibiting PI3k/Akt activation during intestinal I/R. The current study shows that HRD1 is an intestinal I/R critical regulator and that targeting the HRD1/TMEM2 axis may be a promising therapeutic approach.
Collapse
Affiliation(s)
- Xuzi Zhao
- Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, 116023, Dalian, China
| | - Deshun Liu
- Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, 116023, Dalian, China
| | - Yan Zhao
- Department of Pharmacology, Dalian Medical University, 116044, Dalian, China
| | - Zhecheng Wang
- Department of Pharmacology, Dalian Medical University, 116044, Dalian, China
| | - Yue Wang
- Department of Pharmacology, Dalian Medical University, 116044, Dalian, China
| | - Zhao Chen
- Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, 116023, Dalian, China
| | - Shili Ning
- Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, 116023, Dalian, China
| | - Guangzhi Wang
- Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, 116023, Dalian, China
| | - Lu Meng
- Department of Pharmacology, Dalian Medical University, 116044, Dalian, China
| | - Jihong Yao
- Department of Pharmacology, Dalian Medical University, 116044, Dalian, China.
| | - Xiaofeng Tian
- Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, 116023, Dalian, China.
| |
Collapse
|
16
|
Chen J, Zhang H, Yi X, Dou Q, Yang X, He Y, Chen J, Chen K. Cellular senescence of renal tubular epithelial cells in acute kidney injury. Cell Death Discov 2024; 10:62. [PMID: 38316761 PMCID: PMC10844256 DOI: 10.1038/s41420-024-01831-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 01/14/2024] [Accepted: 01/24/2024] [Indexed: 02/07/2024] Open
Abstract
Cellular senescence represents an irreversible state of cell-cycle arrest during which cells secrete senescence-associated secretory phenotypes, including inflammatory factors and chemokines. Additionally, these cells exhibit an apoptotic resistance phenotype. Cellular senescence serves a pivotal role not only in embryonic development, tissue regeneration, and tumor suppression but also in the pathogenesis of age-related degenerative diseases, malignancies, metabolic diseases, and kidney diseases. The senescence of renal tubular epithelial cells (RTEC) constitutes a critical cellular event in the progression of acute kidney injury (AKI). RTEC senescence inhibits renal regeneration and repair processes and, concurrently, promotes the transition of AKI to chronic kidney disease via the senescence-associated secretory phenotype. The mechanisms underlying cellular senescence are multifaceted and include telomere shortening or damage, DNA damage, mitochondrial autophagy deficiency, cellular metabolic disorders, endoplasmic reticulum stress, and epigenetic regulation. Strategies aimed at inhibiting RTEC senescence, targeting the clearance of senescent RTEC, or promoting the apoptosis of senescent RTEC hold promise for enhancing the renal prognosis of AKI. This review primarily focuses on the characteristics and mechanisms of RTEC senescence, and the impact of intervening RTEC senescence on the prognosis of AKI, aiming to provide a foundation for understanding the pathogenesis and providing potentially effective approaches for AKI treatment.
Collapse
Affiliation(s)
- Juan Chen
- Department of Nephrology, Daping Hospital, Army Medical University, 400042, Chongqing, China
| | - Huhai Zhang
- Department of Nephrology, Southwest Hospital, Army Medical University, 400042, Chongqing, China
| | - Xiangling Yi
- Department of Nephrology, Daping Hospital, Army Medical University, 400042, Chongqing, China
| | - Qian Dou
- Department of Nephrology, Daping Hospital, Army Medical University, 400042, Chongqing, China
| | - Xin Yang
- Department of Nephrology, Daping Hospital, Army Medical University, 400042, Chongqing, China
| | - Yani He
- Department of Nephrology, Daping Hospital, Army Medical University, 400042, Chongqing, China
| | - Jia Chen
- Department of Nephrology, Daping Hospital, Army Medical University, 400042, Chongqing, China.
| | - Kehong Chen
- Department of Nephrology, Daping Hospital, Army Medical University, 400042, Chongqing, China.
- State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, Chongqing, China.
| |
Collapse
|
17
|
Gadi S, Niture S, Hoang H, Qi Q, Hatcher C, Huang X, Haider J, Norford DC, Leung T, Levine KE, Kumar D. Deficiency of spns1 exacerbates per- and polyfluoroalkyl substances mediated hepatic toxicity and steatosis in zebrafish (Danio rerio). Toxicology 2023; 499:153641. [PMID: 37806615 DOI: 10.1016/j.tox.2023.153641] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 09/26/2023] [Accepted: 10/05/2023] [Indexed: 10/10/2023]
Abstract
Per- and polyfluoroalkyl substances (PFAS) are man-made long-lasting chemical compounds that are found in everyday household items. Today they occur in the environment as a major group of pollutants. These compounds are broadly used in commercial product preparation such as, for food packaging, nonstick coatings, and firefighting foam. In humans, PFAS can cause immune disorders, impaired fetal development, abnormal skeletal tissue development, osteoarthritis, thyroid dysfunctions, cholesterol changes, affect insulin regulation and lipid metabolism, and are also involved in the development of fatty liver disease. In the current study, we investigated the effect of low, but physiologically relevant, concentrations of perfluorooctanoic acid (PFOA), heptafluorobutyric acid (HFBA), and perfluorotetradecanoic acid (PFTA) on gene expression markers of an inflammatory response (tnfa, il-1b, il-6, rplp0, edem1, and dnajc3a), unfolded protein response (UPR) (bip, atf4a, atf6, xbp1, and ddit3), senescence (p21, pai1, smp30, mdm2, and baxa), lipogenesis (scd1, acc, srebp1, pparγ, and fasn) and autophagy (p62, atg3, atg7, rab7, lc3b, and becn1) in AB wild-type (+/+), spns1-wt sibling (+/+), (+/-) and spns1 homozygous mutant (-/-) zebrafish embryos. Exposure to PFOA and HFBA (50 and 100 nM) specifically modulated inflammatory, UPR, senescence, lipogenic, and autophagy signaling in spns1-wt (+/+), (+/-), and spns1-mutant (-/-) zebrafish embryos. Furthermore, PFOA, but not HFBA, upregulated lipogenic-related gene expression and enhanced hepatic steatosis in spns1-wt (+/+), (+/-) zebrafish embryos. Combined exposure to PFOA, HFBA, and PFTA differentially expressed inflammatory, senescence, lipogenic, and autophagy-associated gene expression in spns1-mutant (-/-) zebrafish embryos compared with spns1-wt (+/+), (+/-) and AB-wt (+/+) zebrafish embryos. In addition, chronic exposure (∼2 months) to PFOA (120-600 nM) upregulated the expression of hepatic lipogenic and steatosis biomarkers in AB-wt (+/+) zebrafish. Collectively, our data suggest that acute/chronic physiologically relevant concentrations of PFOA upregulate inflammatory, UPR, senescence, and lipogenic signaling in spns1-wt (+/+), (+/-) and spns1-mutant (-/-) zebrafish embryos as well as in two-month-old AB-wt zebrafish, by targeting autophagy and hence induces toxicity that could promote nonalcoholic fatty liver disease.
Collapse
Affiliation(s)
- Sashi Gadi
- The Julius L. Chambers Biomedical/Biotechnology Research Institute (JLC-BBRI), North Carolina Central University (NCCU), Durham, NC, USA
| | - Suryakant Niture
- The Julius L. Chambers Biomedical/Biotechnology Research Institute (JLC-BBRI), North Carolina Central University (NCCU), Durham, NC, USA; NCCU-RTI Center for Applied Research in Environmental Sciences (CARES), RTI International, Research Triangle Park, Durham, NC, USA.
| | - Hieu Hoang
- The Julius L. Chambers Biomedical/Biotechnology Research Institute (JLC-BBRI), North Carolina Central University (NCCU), Durham, NC, USA
| | - Qi Qi
- The Julius L. Chambers Biomedical/Biotechnology Research Institute (JLC-BBRI), North Carolina Central University (NCCU), Durham, NC, USA
| | - Charles Hatcher
- The Julius L. Chambers Biomedical/Biotechnology Research Institute (JLC-BBRI), North Carolina Central University (NCCU), Durham, NC, USA
| | - Xiaoyan Huang
- The NCCU, JLC-BBRI North Carolina Research Campus, Kannapolis, NC, USA
| | - Jamil Haider
- The NCCU, JLC-BBRI North Carolina Research Campus, Kannapolis, NC, USA
| | - Derek C Norford
- The Julius L. Chambers Biomedical/Biotechnology Research Institute (JLC-BBRI), North Carolina Central University (NCCU), Durham, NC, USA
| | - TinChung Leung
- The NCCU, JLC-BBRI North Carolina Research Campus, Kannapolis, NC, USA
| | - Keith E Levine
- NCCU-RTI Center for Applied Research in Environmental Sciences (CARES), RTI International, Research Triangle Park, Durham, NC, USA
| | - Deepak Kumar
- The Julius L. Chambers Biomedical/Biotechnology Research Institute (JLC-BBRI), North Carolina Central University (NCCU), Durham, NC, USA; NCCU-RTI Center for Applied Research in Environmental Sciences (CARES), RTI International, Research Triangle Park, Durham, NC, USA.
| |
Collapse
|
18
|
Li D, Li Y, Ding H, Wang Y, Xie Y, Zhang X. Cellular Senescence in Cardiovascular Diseases: From Pathogenesis to Therapeutic Challenges. J Cardiovasc Dev Dis 2023; 10:439. [PMID: 37887886 PMCID: PMC10607269 DOI: 10.3390/jcdd10100439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 10/17/2023] [Accepted: 10/19/2023] [Indexed: 10/28/2023] Open
Abstract
Cellular senescence (CS), classically considered a stable cell cycle withdrawal, is hallmarked by a progressive decrease in cell growth, differentiation, and biological activities. Senescent cells (SNCs) display a complicated senescence-associated secretory phenotype (SASP), encompassing a variety of pro-inflammatory factors that exert influence on the biology of both the cell and surrounding tissue. Among global mortality causes, cardiovascular diseases (CVDs) stand out, significantly impacting the living quality and functional abilities of patients. Recent data suggest the accumulation of SNCs in aged or diseased cardiovascular systems, suggesting their potential role in impairing cardiovascular function. CS operates as a double-edged sword: while it can stimulate the restoration of organs under physiological conditions, it can also participate in organ and tissue dysfunction and pave the way for multiple chronic diseases under pathological states. This review explores the mechanisms that underlie CS and delves into the distinctive features that characterize SNCs. Furthermore, we describe the involvement of SNCs in the progression of CVDs. Finally, the study provides a summary of emerging interventions that either promote or suppress senescence and discusses their therapeutic potential in CVDs.
Collapse
Affiliation(s)
- Dan Li
- Department of Cardiovascular Medicine, Lanzhou University Second Hospital, Lanzhou 730030, China; (D.L.); (H.D.); (Y.W.); (Y.X.)
| | - Yongnan Li
- Department of Cardiac Surgery, Lanzhou University Second Hospital, Lanzhou 730030, China;
| | - Hong Ding
- Department of Cardiovascular Medicine, Lanzhou University Second Hospital, Lanzhou 730030, China; (D.L.); (H.D.); (Y.W.); (Y.X.)
| | - Yuqin Wang
- Department of Cardiovascular Medicine, Lanzhou University Second Hospital, Lanzhou 730030, China; (D.L.); (H.D.); (Y.W.); (Y.X.)
| | - Yafei Xie
- Department of Cardiovascular Medicine, Lanzhou University Second Hospital, Lanzhou 730030, China; (D.L.); (H.D.); (Y.W.); (Y.X.)
| | - Xiaowei Zhang
- Department of Cardiovascular Medicine, Lanzhou University Second Hospital, Lanzhou 730030, China; (D.L.); (H.D.); (Y.W.); (Y.X.)
| |
Collapse
|
19
|
Hela F, Aguayo-Mazzucato C. Interaction between Autophagy and Senescence in Pancreatic Beta Cells. BIOLOGY 2023; 12:1205. [PMID: 37759604 PMCID: PMC10525299 DOI: 10.3390/biology12091205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 09/01/2023] [Accepted: 09/02/2023] [Indexed: 09/29/2023]
Abstract
Aging leads to an increase in cellular stress due to the fragility of the organism and the inability to cope with it. In this setting, there is a higher chance of developing different cardiometabolic diseases like diabetes. Cellular senescence and autophagy, both hallmarks of aging and stress-coping mechanisms, have gained increased attention for their role in the pathophysiology of diabetes. Studies show that impairing senescence dampens and even prevents diabetes while the role of autophagy is more contradictory, implying a context- and disease-stage-dependent effect. Reports show conflicting data about the effect of autophagy on senescence while the knowledge about this interaction in beta cells remains scarce. Elucidating this interaction between autophagy and senescence in pancreatic beta cells will lead to an identification of their respective roles and the extent of the effect each mechanism has on beta cells and open new horizons for developing novel therapeutic agents. To help illuminate this relationship we will review the latest findings of cellular senescence and autophagy with a special emphasis on pancreatic beta cells and diabetes.
Collapse
Affiliation(s)
| | - Cristina Aguayo-Mazzucato
- Section on Islet Cell Biology and Regenerative Medicine, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
20
|
Zhong M, Wu Z, Chen Z, Ren Q, Zhou J. Advances in the interaction between endoplasmic reticulum stress and osteoporosis. Biomed Pharmacother 2023; 165:115134. [PMID: 37437374 DOI: 10.1016/j.biopha.2023.115134] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 07/04/2023] [Accepted: 07/07/2023] [Indexed: 07/14/2023] Open
Abstract
The endoplasmic reticulum (ER) is the main site for protein synthesis, folding, and secretion, and accumulation of the unfolded/misfolded proteins in the ER may induce ER stress. ER stress is an important participant in various intracellular signaling pathways. Prolonged- or high-intensity ER stress may induce cell apoptosis. Osteoporosis, characterized by imbalanced bone remodeling, is a global disease caused by many factors, such as ER stress. ER stress stimulates osteoblast apoptosis, increases bone loss, and promotes osteoporosis development. Many factors, such as the drug's adverse effects, metabolic disorders, calcium ion imbalance, bad habits, and aging, have been reported to activate ER stress, resulting in the pathological development of osteoporosis. Increasing evidence shows that ER stress regulates osteogenic differentiation, osteoblast activity, and osteoclast formation and function. Various therapeutic agents have been developed to counteract ER stress and thereby suppress osteoporosis development. Thus, inhibition of ER stress has become a potential target for the therapeutic management of osteoporosis. However, the in-depth understanding of ER stress in the pathogenesis of osteoporosis still needs more effort.
Collapse
Affiliation(s)
- Mingliang Zhong
- College of Rehabilitation, Gannan Medical University, Ganzhou 341000, China
| | - Zhenyu Wu
- First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
| | - Zhixi Chen
- College of Pharmacy, Gannan Medical University, Ganzhou 341000, China
| | - Qun Ren
- College of Pharmacy, Gannan Medical University, Ganzhou 341000, China
| | - Jianguo Zhou
- Department of Joint Surgery, Ganzhou People's Hospital, Ganzhou 341000, China.
| |
Collapse
|
21
|
Kang T, Moore EC, Kopania EEK, King CD, Schilling B, Campisi J, Good JM, Brem RB. A natural variation-based screen in mouse cells reveals USF2 as a regulator of the DNA damage response and cellular senescence. G3 (BETHESDA, MD.) 2023; 13:jkad091. [PMID: 37097016 PMCID: PMC10320765 DOI: 10.1093/g3journal/jkad091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 03/30/2023] [Accepted: 04/06/2023] [Indexed: 04/26/2023]
Abstract
Cellular senescence is a program of cell cycle arrest, apoptosis resistance, and cytokine release induced by stress exposure in metazoan cells. Landmark studies in laboratory mice have characterized a number of master senescence regulators, including p16INK4a, p21, NF-κB, p53, and C/EBPβ. To discover other molecular players in senescence, we developed a screening approach to harness the evolutionary divergence between mouse species. We found that primary cells from the Mediterranean mouse Mus spretus, when treated with DNA damage to induce senescence, produced less cytokine and had less-active lysosomes than cells from laboratory Mus musculus. We used allele-specific expression profiling to catalog senescence-dependent cis-regulatory variation between the species at thousands of genes. We then tested for correlation between these expression changes and interspecies sequence variants in the binding sites of transcription factors. Among the emergent candidate senescence regulators, we chose a little-studied cell cycle factor, upstream stimulatory factor 2 (USF2), for molecular validation. In acute irradiation experiments, cells lacking USF2 had compromised DNA damage repair and response. Longer-term senescent cultures without USF2 mounted an exaggerated senescence regulatory program-shutting down cell cycle and DNA repair pathways, and turning up cytokine expression, more avidly than wild-type. We interpret these findings under a model of pro-repair, anti-senescence regulatory function by USF2. Our study affords new insights into the mechanisms by which cells commit to senescence, and serves as a validated proof of concept for natural variation-based regulator screens.
Collapse
Affiliation(s)
- Taekyu Kang
- Buck Institute for Research on Aging, Novato, CA 94945, USA
- Department of Plant and Microbial Biology, University of California, Berkeley, CA 94720, USA
| | - Emily C Moore
- Division of Biological Sciences, University of Montana, Missoula, MT 59812, USA
| | - Emily E K Kopania
- Division of Biological Sciences, University of Montana, Missoula, MT 59812, USA
| | | | | | - Judith Campisi
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Jeffrey M Good
- Division of Biological Sciences, University of Montana, Missoula, MT 59812, USA
| | - Rachel B Brem
- Buck Institute for Research on Aging, Novato, CA 94945, USA
- Department of Plant and Microbial Biology, University of California, Berkeley, CA 94720, USA
| |
Collapse
|
22
|
Gerosa L, Malvandi AM, Malavolta M, Provinciali M, Lombardi G. Exploring cellular senescence in the musculoskeletal system: Any insights for biomarkers discovery? Ageing Res Rev 2023; 88:101943. [PMID: 37142059 DOI: 10.1016/j.arr.2023.101943] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 04/21/2023] [Accepted: 05/01/2023] [Indexed: 05/06/2023]
Abstract
The locomotor system comprises skeletal muscles and bones with active metabolism and cellular turnover. Chronic locomotor system disorders gradually arising with aging are inversely associated with the correct function of bone and muscles. Senescent cells appear more frequently in advanced ages or pathological conditions, and the accumulation of senescent cells in muscle tissue negatively correlates with muscle regeneration, which is crucial for maintaining strength and preventing frailty. Senescence in the bone microenvironment, osteoblasts, and osteocytes affects bone turnover favoring osteoporosis. It is likely that in response to injury and age-related damage over the lifetime, a subset of niche cells accumulates oxidative stress and DNA damage beyond the threshold that primes the onset of cellular senescence. These senescent cells may acquire resistance to apoptosis that, combined with the weakened immune system, results in impaired clearance of senescent cells and their accumulation. The secretory profile of senescent cells causes local inflammation, further spreading senescence in neighboring niche cells and impairing tissue homeostasis. The resulting impairment of turnover/tissue repair in the musculoskeletal system reduces the efficiency of the organ in response to environmental needs that finally lead to functional decline. Management of the musculoskeletal system at the cellular level can benefit the quality of life and reduce early aging. This work discusses current knowledge of cellular senescence of musculoskeletal tissues to conclude with biologically active biomarkers effective enough to reveal the underlying mechanisms of tissue flaws at the earliest possible.
Collapse
Affiliation(s)
- Laura Gerosa
- Laboratory of Experimental Biochemistry and Molecular Biology, IRCCS Istituto Ortopedico Galeazzi, Milano, Italy
| | - Amir Mohammad Malvandi
- Laboratory of Experimental Biochemistry and Molecular Biology, IRCCS Istituto Ortopedico Galeazzi, Milano, Italy.
| | - Marco Malavolta
- Advanced Technology Center for Aging Research, IRCCS INRCA, 60121 Ancona, Italy
| | - Mauro Provinciali
- Advanced Technology Center for Aging Research, IRCCS INRCA, 60121 Ancona, Italy
| | - Giovanni Lombardi
- Laboratory of Experimental Biochemistry and Molecular Biology, IRCCS Istituto Ortopedico Galeazzi, Milano, Italy; Department of Athletics, Strength and Conditioning, Poznań University of Physical Education, Poznań, Poland
| |
Collapse
|
23
|
Giroud J, Bouriez I, Paulus H, Pourtier A, Debacq-Chainiaux F, Pluquet O. Exploring the Communication of the SASP: Dynamic, Interactive, and Adaptive Effects on the Microenvironment. Int J Mol Sci 2023; 24:10788. [PMID: 37445973 DOI: 10.3390/ijms241310788] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 06/20/2023] [Accepted: 06/25/2023] [Indexed: 07/15/2023] Open
Abstract
Cellular senescence is a complex cell state that can occur during physiological ageing or after exposure to stress signals, regardless of age. It is a dynamic process that continuously evolves in a context-dependent manner. Senescent cells interact with their microenvironment by producing a heterogenous and plastic secretome referred to as the senescence-associated secretory phenotype (SASP). Hence, understanding the cross-talk between SASP and the microenvironment can be challenging due to the complexity of signal exchanges. In this review, we first aim to update the definition of senescence and its associated biomarkers from its discovery to the present day. We detail the regulatory mechanisms involved in the expression of SASP at multiple levels and develop how SASP can orchestrate microenvironment modifications, by focusing on extracellular matrix modifications, neighboring cells' fate, and intercellular communications. We present hypotheses on how these microenvironmental events may affect dynamic changes in SASP composition in return. Finally, we discuss the various existing approaches to targeting SASP and clarify what is currently known about the biological effects of these modified SASPs on the cellular environment.
Collapse
Affiliation(s)
- Joëlle Giroud
- Laboratory of Biochemistry and Cell Biology (URBC), Namur Research Institute for Life Sciences (NARILIS), University of Namur, 5000 Namur, Belgium
- University of Lille, CNRS, Inserm, Pasteur Institute of Lille, UMR9020-U1277-CANTHER-Cancer Heterogeneity Plasticity and Resistance to Therapies, 59000 Lille, France
| | - Inès Bouriez
- Laboratory of Biochemistry and Cell Biology (URBC), Namur Research Institute for Life Sciences (NARILIS), University of Namur, 5000 Namur, Belgium
| | - Hugo Paulus
- Laboratory of Biochemistry and Cell Biology (URBC), Namur Research Institute for Life Sciences (NARILIS), University of Namur, 5000 Namur, Belgium
| | - Albin Pourtier
- University of Lille, CNRS, Inserm, Pasteur Institute of Lille, UMR9020-U1277-CANTHER-Cancer Heterogeneity Plasticity and Resistance to Therapies, 59000 Lille, France
| | - Florence Debacq-Chainiaux
- Laboratory of Biochemistry and Cell Biology (URBC), Namur Research Institute for Life Sciences (NARILIS), University of Namur, 5000 Namur, Belgium
| | - Olivier Pluquet
- University of Lille, CNRS, Inserm, Pasteur Institute of Lille, UMR9020-U1277-CANTHER-Cancer Heterogeneity Plasticity and Resistance to Therapies, 59000 Lille, France
| |
Collapse
|
24
|
Ma DJ, Hwang JS, Noh KB, Oh SH, Kim KW, Shin YJ. Role of NADPH Oxidase 4 in Corneal Endothelial Cells Is Mediated by Endoplasmic Reticulum Stress and Autophagy. Antioxidants (Basel) 2023; 12:1228. [PMID: 37371958 DOI: 10.3390/antiox12061228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 06/03/2023] [Accepted: 06/05/2023] [Indexed: 06/29/2023] Open
Abstract
Human corneal-endothelial cells (hCEnCs) are located on the inner layer of the cornea. Injury to CEnCs leads to permanent corneal edema, requiring corneal transplantation. NADPH oxidase 4 (NOX4) has been reported to be implicated in the pathogenesis of CEnCs diseases. Thus, we investigated the role of NOX4 in CEnCs in this study. In an animal study, siRNA for NOX4 (siNOX4) or plasmid for NOX4 (pNOX4) was introduced into the corneal endothelium of rats by electroporation, using a square-wave electroporator (ECM830, Havard apparatus) to decrease or increase the expression of NOX4, respectively, and the rat corneas were cryoinjured through contact with a metal rod of 3 mm diameter frozen in liquid nitrogen for 10 min. The immunofluorescence staining of NOX4 and 8-OHdG showed that the levels of NOX4 and 8-OHdG were decreased in the siNOX4 group compared to the siControl, and increased in the pNOX4 group compared to the pControl at one week after treatment. Without cryoinjury, corneal opacity was more severe, and the density of CEnCs was lower, in pNOX4-treated rats compared to pControl. After cryoinjury, the corneas were more transparent, and the CEnC density was higher, in siNOX4-treated rats. The hCEnCs were cultured and transfected with siNOX4 and pNOX4. The silencing of NOX4 in hCEnCs resulted in a normal cell shape, higher viability, and higher proliferation rate than those transfected with the siControl, while NOX4 overexpression had the opposite effect. NOX4 overexpression increased the number of senescent cells and intracellular oxidative stress levels. NOX4 overexpression increased ATF4 and ATF6 levels, and nuclear translocation of XBP-1, which is the endoplasmic reticulum (ER) stress marker, while the silencing of NOX4 had the opposite effect. Additionally, the mitochondrial membrane potential was hyperpolarized by the silencing of NOX4, and depolarized by NOX4 overexpression. The LC3II levels, a marker of autophagy, were decreased by the silencing of NOX4, and increased by NOX4 overexpression. In conclusion, NOX4 plays a pivotal role in the wound-healing and senescence of hCEnCs, by modulating oxidative stress, ER stress, and autophagy. The regulation of NOX4 may be a potential therapeutic strategy for regulating the homeostasis of CEnCs, and treating corneal-endothelial diseases.
Collapse
Affiliation(s)
- Dae Joong Ma
- Department of Ophthalmology, Hallym University Medical Center, College of Medicine, Hallym University, Seoul 07442, Republic of Korea
- Hallym BioEyeTech Research Center, College of Medicine, Hallym University, Seoul 07442, Republic of Korea
| | - Jin Sun Hwang
- Department of Ophthalmology, Hallym University Medical Center, College of Medicine, Hallym University, Seoul 07442, Republic of Korea
- Hallym BioEyeTech Research Center, College of Medicine, Hallym University, Seoul 07442, Republic of Korea
| | - Kyung Bo Noh
- Department of Ophthalmology, Hallym University Medical Center, College of Medicine, Hallym University, Seoul 07442, Republic of Korea
- Hallym BioEyeTech Research Center, College of Medicine, Hallym University, Seoul 07442, Republic of Korea
| | - Sun-Hee Oh
- Department of Ophthalmology, Hallym University Medical Center, College of Medicine, Hallym University, Seoul 07442, Republic of Korea
- Hallym BioEyeTech Research Center, College of Medicine, Hallym University, Seoul 07442, Republic of Korea
| | - Kyoung Wook Kim
- Department of Ophthalmology, Hallym University Medical Center, College of Medicine, Hallym University, Seoul 07442, Republic of Korea
- Hallym BioEyeTech Research Center, College of Medicine, Hallym University, Seoul 07442, Republic of Korea
| | - Young Joo Shin
- Department of Ophthalmology, Hallym University Medical Center, College of Medicine, Hallym University, Seoul 07442, Republic of Korea
- Hallym BioEyeTech Research Center, College of Medicine, Hallym University, Seoul 07442, Republic of Korea
| |
Collapse
|
25
|
Eleftheriadis T, Pissas G, Golfinopoulos S, Efthymiadi M, Poulianiti C, Polyzou Konsta MA, Liakopoulos V, Stefanidis I. Routes of Albumin Overload Toxicity in Renal Tubular Epithelial Cells. Int J Mol Sci 2023; 24:ijms24119640. [PMID: 37298591 DOI: 10.3390/ijms24119640] [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: 05/05/2023] [Revised: 05/29/2023] [Accepted: 05/31/2023] [Indexed: 06/12/2023] Open
Abstract
Besides being a marker of kidney disease severity, albuminuria exerts a toxic effect on renal proximal tubular epithelial cells (RPTECs). We evaluated whether an unfolded protein response (UPR) or DNA damage response (DDR) is elicited in RPTECs exposed to high albumin concentration. The deleterious outcomes of the above pathways, apoptosis, senescence, or epithelial-to-mesenchymal transition (EMT) were evaluated. Albumin caused reactive oxygen species (ROS) overproduction and protein modification, and a UPR assessed the level of crucial molecules involved in this pathway. ROS also induced a DDR evaluated by critical molecules involved in this pathway. Apoptosis ensued through the extrinsic pathway. Senescence also occurred, and the RPTECs acquired a senescence-associated secretory phenotype since they overproduced IL-1β and TGF-β1. The latter may contribute to the observed EMT. Agents against endoplasmic reticulum stress (ERS) only partially alleviated the above changes, while the inhibition of ROS upregulation prevented both UPR and DDR and all the subsequent harmful effects. Briefly, albumin overload causes cellular apoptosis, senescence, and EMT in RPTECs by triggering UPR and DDR. Promising anti-ERS factors are beneficial but cannot eliminate the albumin-induced deleterious effects because DDR also occurs. Factors that suppress ROS overproduction may be more effective since they could halt UPR and DDR.
Collapse
Affiliation(s)
- Theodoros Eleftheriadis
- Department of Nephrology, Faculty of Medicine, University of Thessaly, Biopolis, Mezourlo Hill, 41110 Larissa, Greece
| | - Georgios Pissas
- Department of Nephrology, Faculty of Medicine, University of Thessaly, Biopolis, Mezourlo Hill, 41110 Larissa, Greece
| | - Spyridon Golfinopoulos
- Department of Nephrology, Faculty of Medicine, University of Thessaly, Biopolis, Mezourlo Hill, 41110 Larissa, Greece
| | - Maria Efthymiadi
- Department of Nephrology, Faculty of Medicine, University of Thessaly, Biopolis, Mezourlo Hill, 41110 Larissa, Greece
| | - Christina Poulianiti
- Department of Nephrology, Faculty of Medicine, University of Thessaly, Biopolis, Mezourlo Hill, 41110 Larissa, Greece
| | - Maria Anna Polyzou Konsta
- Department of Nephrology, Faculty of Medicine, University of Thessaly, Biopolis, Mezourlo Hill, 41110 Larissa, Greece
| | - Vassilios Liakopoulos
- Department of Nephrology, Faculty of Medicine, University of Thessaly, Biopolis, Mezourlo Hill, 41110 Larissa, Greece
| | - Ioannis Stefanidis
- Department of Nephrology, Faculty of Medicine, University of Thessaly, Biopolis, Mezourlo Hill, 41110 Larissa, Greece
| |
Collapse
|
26
|
Kusuma FK, Prabhu A, Tieo G, Ahmed SM, Dakle P, Yong WK, Pathak E, Madan V, Jiang YY, Tam WL, Kappei D, Dröge P, Koeffler HP, Jeitany M. Signalling inhibition by ponatinib disrupts productive alternative lengthening of telomeres (ALT). Nat Commun 2023; 14:1919. [PMID: 37024489 PMCID: PMC10079688 DOI: 10.1038/s41467-023-37633-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 03/24/2023] [Indexed: 04/08/2023] Open
Abstract
Alternative lengthening of telomeres (ALT) supports telomere maintenance in 10-15% of cancers, thus representing a compelling target for therapy. By performing anti-cancer compound library screen on isogenic cell lines and using extrachromosomal telomeric C-circles, as a bona fide marker of ALT activity, we identify a receptor tyrosine kinase inhibitor ponatinib that deregulates ALT mechanisms, induces telomeric dysfunction, reduced ALT-associated telomere synthesis, and targets, in vivo, ALT-positive cells. Using RNA-sequencing and quantitative phosphoproteomic analyses, combined with C-circle level assessment, we find an ABL1-JNK-JUN signalling circuit to be inhibited by ponatinib and to have a role in suppressing telomeric C-circles. Furthermore, transcriptome and interactome analyses suggest a role of JUN in DNA damage repair. These results are corroborated by synergistic drug interactions between ponatinib and either DNA synthesis or repair inhibitors, such as triciribine. Taken together, we describe here a signalling pathway impacting ALT which can be targeted by a clinically approved drug.
Collapse
Affiliation(s)
- Frances Karla Kusuma
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Aishvaryaa Prabhu
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Galen Tieo
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Syed Moiz Ahmed
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Pushkar Dakle
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Wai Khang Yong
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Elina Pathak
- Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Vikas Madan
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Yan Yi Jiang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
- Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, P. R. China
| | - Wai Leong Tam
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Dennis Kappei
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Peter Dröge
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - H Phillip Koeffler
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
- Cedars-Sinai Medical Center, Division of Hematology/Oncology, UCLA School of Medicine, Los Angeles, CA, USA
- Department of Hematology-Oncology, National University Cancer Institute of Singapore (NCIS), National University Hospital, Singapore, Singapore
| | - Maya Jeitany
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore.
| |
Collapse
|
27
|
Menezes PR, Trufen CEM, Lichtenstein F, Pellegrina DVDS, Reis EM, Onuki J. Transcriptome profile analysis reveals putative molecular mechanisms of 5-aminolevulinic acid toxicity. Arch Biochem Biophys 2023; 738:109540. [PMID: 36746260 DOI: 10.1016/j.abb.2023.109540] [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: 08/16/2022] [Revised: 12/23/2022] [Accepted: 02/03/2023] [Indexed: 02/05/2023]
Abstract
5-aminolevulinic acid (5-ALA) is the first precursor of the heme biosynthesis pathway, accumulated in acute intermittent porphyria (AIP), an inherited metabolic disease characterized by porphobilinogen deaminase deficiency. An increased incidence of hepatocellular carcinoma (HCC) has been reported as a long-term manifestation in symptomatic AIP patients. 5-ALA is an α-aminoketone prone to oxidation, yielding reactive oxygen species and 4,5-dioxovaleric acid. A high concentration of 5-ALA presents deleterious pro-oxidant potential. It can induce apoptosis, DNA damage, mitochondrial dysfunction, and altered expression of carcinogenesis-related proteins. Several hypotheses of the increased risk of HCC rely on the harmful effect of elevated 5-ALA in the liver of AIP patients, which could promote a pro-carcinogenic environment. We investigated the global transcriptional changes and perturbed molecular pathways in HepG2 cells following exposure to 5-ALA 25 mM for 2 h and 24 h using DNA microarray. Distinct transcriptome profiles were observed. 5-ALA '25 mM-2h' upregulated 10 genes associated with oxidative stress response and carcinogenesis. Enrichment analysis of differentially expressed genes by KEGG, Reactome, MetaCore™, and Gene Ontology, showed that 5-ALA '25 mM-24h' enriched pathways involved in drug detoxification, oxidative stress, DNA damage, cell death/survival, cell cycle, and mitochondria dysfunction corroborating the pro-oxidant properties of 5-ALA. Furthermore, our results disclosed other possible processes such as senescence, immune responses, endoplasmic reticulum stress, and also some putative effectors, such as sequestosome, osteopontin, and lon peptidase 1. This study provided additional knowledge about molecular mechanisms of 5-ALA toxicity which is essential to a deeper understanding of AIP and HCC pathophysiology. Furthermore, our findings can contribute to improving the efficacy of current therapies and the development of novel biomarkers and targets for diagnosis, prognosis, and therapeutic strategies for AHP/AIP and associated HCC.
Collapse
Affiliation(s)
- Patricia Regina Menezes
- Laboratório de Desenvolvimento e Inovação, Instituto Butantan, Av. Vital Brazil, 1500, 05503-900, São Paulo, SP, Brazil
| | - Carlos Eduardo Madureira Trufen
- Laboratório de Desenvolvimento e Inovação, Instituto Butantan, Av. Vital Brazil, 1500, 05503-900, São Paulo, SP, Brazil; Centro de Excelência para Descoberta de Novos Alvos Moleculares, Instituto Butantan, Av. Vital Brazil, 1500, 05503-900, São Paulo, SP, Brazil; Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, Průmyslová 595, 252 50, Vestec, Czech Republic
| | - Flavio Lichtenstein
- Laboratório de Desenvolvimento e Inovação, Instituto Butantan, Av. Vital Brazil, 1500, 05503-900, São Paulo, SP, Brazil; Centro de Excelência para Descoberta de Novos Alvos Moleculares, Instituto Butantan, Av. Vital Brazil, 1500, 05503-900, São Paulo, SP, Brazil
| | | | - Eduardo Moraes Reis
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, Av. Prof. Lineu Prestes, 748, 05508-900, São Paulo, SP, Brazil
| | - Janice Onuki
- Laboratório de Desenvolvimento e Inovação, Instituto Butantan, Av. Vital Brazil, 1500, 05503-900, São Paulo, SP, Brazil; Centro de Excelência para Descoberta de Novos Alvos Moleculares, Instituto Butantan, Av. Vital Brazil, 1500, 05503-900, São Paulo, SP, Brazil; Laboratório de Herpetologia, Instituto Butantan, Av. Vital Brazil, 1500, 05503-900, São Paulo, SP, Brazil.
| |
Collapse
|
28
|
Hogan KA, Zeidler JD, Beasley HK, Alsaadi AI, Alshaheeb AA, Chang YC, Tian H, Hinton AO, McReynolds MR. Using mass spectrometry imaging to visualize age-related subcellular disruption. Front Mol Biosci 2023; 10:906606. [PMID: 36968274 PMCID: PMC10032471 DOI: 10.3389/fmolb.2023.906606] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 01/24/2023] [Indexed: 03/10/2023] Open
Abstract
Metabolic homeostasis balances the production and consumption of energetic molecules to maintain active, healthy cells. Cellular stress, which disrupts metabolism and leads to the loss of cellular homeostasis, is important in age-related diseases. We focus here on the role of organelle dysfunction in age-related diseases, including the roles of energy deficiencies, mitochondrial dysfunction, endoplasmic reticulum (ER) stress, changes in metabolic flux in aging (e.g., Ca2+ and nicotinamide adenine dinucleotide), and alterations in the endoplasmic reticulum-mitochondria contact sites that regulate the trafficking of metabolites. Tools for single-cell resolution of metabolite pools and metabolic flux in animal models of aging and age-related diseases are urgently needed. High-resolution mass spectrometry imaging (MSI) provides a revolutionary approach for capturing the metabolic states of individual cells and cellular interactions without the dissociation of tissues. mass spectrometry imaging can be a powerful tool to elucidate the role of stress-induced cellular dysfunction in aging.
Collapse
Affiliation(s)
- Kelly A. Hogan
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA, United States
- Signal Transduction and Molecular Nutrition Laboratory, Kogod Aging Center, Department of Anesthesiology and Perioperative Medicine, Mayo Clinic College of Medicine, Rochester, MN, United States
- Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, United States
| | - Julianna D. Zeidler
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Heather K. Beasley
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, United States
| | - Abrar I. Alsaadi
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA, United States
| | - Abdulkareem A. Alshaheeb
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA, United States
| | - Yi-Chin Chang
- Department of Chemistry, Pennsylvania State University, University Park, PA, United States
| | - Hua Tian
- Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, United States
- Department of Chemistry, Pennsylvania State University, University Park, PA, United States
- *Correspondence: Hua Tian, ; Antentor O. Hinton Jr, ; Melanie R. McReynolds,
| | - Antentor O. Hinton
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, United States
- *Correspondence: Hua Tian, ; Antentor O. Hinton Jr, ; Melanie R. McReynolds,
| | - Melanie R. McReynolds
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA, United States
- Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, United States
- *Correspondence: Hua Tian, ; Antentor O. Hinton Jr, ; Melanie R. McReynolds,
| |
Collapse
|
29
|
Azizi M, Salehi-Mazandarani S, Nikpour P, Andalib A, Rezaei M. The role of unfolded protein response-associated miRNAs in immunogenic cell death amplification: A literature review and bioinformatics analysis. Life Sci 2023; 314:121341. [PMID: 36586572 DOI: 10.1016/j.lfs.2022.121341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 12/19/2022] [Accepted: 12/26/2022] [Indexed: 12/30/2022]
Abstract
Immunogenic cell death (ICD) is a type of cellular death that is elicited in response to the specific types of anti-cancer therapies and enhances the anti-tumor immune responses by the combination of antigenicity and adjuvanticity of dying tumor cells. There is a well-established interlink between endoplasmic reticulum stress (ERS) and ICD elicited by anti-cancer therapies. Most recent evidences support that unfolded protein response (UPR)-associated miRNAs can be key players in the ERS-induced ICD. Hence, in the present study, we conducted a literature review on the role of these miRNAs and associated molecular pathways that may regulate ICD. We first collected UPR-associated miRNAs that promote ERS-induced apoptosis and then focused on microRNAs (miRNAs) that promote ERS-induced apoptosis via PERK/eIF2α/ATF4/CHOP pathway activation, as the main core for ICD and release of damage-associated molecular patterns. To better identify PERK/eIF2α/ATF4/CHOP pathway-inducing miRNAs that can be used as potential therapeutic targets for improving ICD in cancer treatment, we did a comprehensive bioinformatics analysis and network construction. Our results showed that "pathways in cancer", "MAPK signaling pathway", "PI3K-Akt signaling pathway", and "Cellular senescence", which correlate with UPR components and ERS induction, were among the significant signaling pathways related to the target genes of these miRNAs. Furthermore, a protein-protein interaction (PPI) network was constructed, which revealed the involvement of the PPI-extracted hub genes in the regulation of proliferation and apoptosis. In conclusion, we propose that these types of miRNAs can be considered as the potential cancer therapy options for better induction of ICD in combination with other ICD inducers.
Collapse
Affiliation(s)
- Mahdieh Azizi
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Sadra Salehi-Mazandarani
- Department of Genetics and Molecular Biology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Parvaneh Nikpour
- Department of Genetics and Molecular Biology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Alireza Andalib
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Marzieh Rezaei
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
30
|
Wang T, Chen P, Weir S, Baltezor M, Schoenen FJ, Chen Q. Novel compound C150 inhibits pancreatic cancer through induction of ER stress and proteosome assembly. Front Oncol 2022; 12:870473. [PMID: 36276125 PMCID: PMC9579335 DOI: 10.3389/fonc.2022.870473] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 09/13/2022] [Indexed: 11/24/2022] Open
Abstract
Pancreatic cancer is a devastating disease with a dismal prognosis and poor treatment outcomes. Searching for new agents for pancreatic cancer treatment is of great significance. We previously identified a novel activity of compound C150 to inhibit pancreatic cancer epithelial-to-mesenchymal transition (EMT). Here, we further revealed its mechanism of action. C150 induced ER stress in pancreatic cancer cells and subsequently increased proteasome activity by enhancing proteasome assembly, which subsequently enhanced the degradation of critical EMT transcription factors (EMT-TFs). In addition, as cellular responses to ER stress, autophagy was elevated, and general protein synthesis was inhibited in pancreatic cancer cells. Besides EMT inhibition, the C150-induced ER stress resulted in G2/M cell cycle arrest, which halted cell proliferation and led to cellular senescence. In an orthotopic syngeneic mouse model, an oral dose of C150 at 150 mg/kg 3× weekly significantly increased survival of mice bearing pancreatic tumors, and reduced tumor growth and ascites occurrence. These results suggested that compound C150 holds promises in comprehensively inhibiting pancreatic cancer progression.
Collapse
Affiliation(s)
- Tao Wang
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas, KS, United States
| | - Ping Chen
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas, KS, United States
| | - Scott Weir
- Department of Cancer Biology, University of Kansas Medical Center, Kansas, KS, United States
| | - Michael Baltezor
- Biotechnology Innovation and Optimization Center, University of Kansas, Lawrence, KS, United States
| | - Frank J. Schoenen
- Higuchi Biosciences Center, University of Kansas, Lawrence, KS, United States
- Medicinal Chemistry Core Laboratory, Lead Development and Optimization Shared Resource, University of Kansas Cancer Center, Lawrence, KS, United States
| | - Qi Chen
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas, KS, United States
- *Correspondence: Qi Chen,
| |
Collapse
|
31
|
Piskorz WM, Cechowska-Pasko M. Senescence of Tumor Cells in Anticancer Therapy—Beneficial and Detrimental Effects. Int J Mol Sci 2022; 23:ijms231911082. [PMID: 36232388 PMCID: PMC9570404 DOI: 10.3390/ijms231911082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 09/16/2022] [Accepted: 09/18/2022] [Indexed: 01/10/2023] Open
Abstract
Cellular senescence process results in stable cell cycle arrest, which prevents cell proliferation. It can be induced by a variety of stimuli including metabolic stress, DNA damage, telomeres shortening, and oncogenes activation. Senescence is generally considered as a process of tumor suppression, both by preventing cancer cells proliferation and inhibiting cancer progression. It can also be a key effector mechanism for many types of anticancer therapies such as chemotherapy and radiotherapy, both directly and through bioactive molecules released by senescent cells that can stimulate an immune response. Senescence is characterized by a senescence-associated secretory phenotype (SASP) that can have both beneficial and detrimental impact on cancer progression. Despite the negatives, attempts are still being made to use senescence to fight cancer, especially when it comes to senolytics. There is a possibility that a combination of prosenescence therapy—which targets tumor cells and causes their senescence—with senotherapy—which targets senescent cells, can be promising in cancer treatment. This review provides information on cellular senescence, its connection with carcinogenesis and therapeutic possibilities linked to this process.
Collapse
|
32
|
Protection of zero-valent iron nanoparticles against sepsis and septic heart failure. J Nanobiotechnology 2022; 20:405. [PMID: 36064371 PMCID: PMC9444118 DOI: 10.1186/s12951-022-01589-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Accepted: 08/08/2022] [Indexed: 11/23/2022] Open
Abstract
Background Septic heart failure accounts for high mortality rates globally. With a strong reducing capacity, zero-valent iron nanoparticles (nanoFe) have been applied in many fields. However, the precise roles and mechanisms of nanoFe in septic cardiomyopathy remain unknown. Results NanoFe was prepared via the liquid-phase reduction method and functionalized with the biocompatible polymer sodium carboxymethylcellulose (CMC). We then successfully constructed a mouse model of septic myocardial injury by challenging with cecal ligation and puncture (CLP). Our findings demonstrated that nanoFe has a significant protective effect on CLP-induced septic myocardial injury. This may be achieved by attenuating inflammation and oxidative stress, improving mitochondrial function, regulating endoplasmic reticulum stress, and activating the AMPK pathway. The RNA-seq results supported the role of nanoFe treatment in regulating a transcriptional profile consistent with its role in response to sepsis. Conclusions The results provide a theoretical basis for the application strategy and combination of nanoFe in sepsis and septic myocardial injury. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12951-022-01589-1.
Collapse
|
33
|
Zhang Q, Zhou D, Liang Y. Single-Cell Analyses of Heterotopic Ossification: Characteristics of Injury-Related Senescent Fibroblasts. J Inflamm Res 2022; 15:5579-5593. [PMID: 36185637 PMCID: PMC9519125 DOI: 10.2147/jir.s369376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 08/17/2022] [Indexed: 11/23/2022] Open
Abstract
Introduction Injury-related cellular senescence may be involved in heterotopic ossification, and no research has been performed about this before. Methods and Results The study utilized integrated single-cell RNA-sequencing (scRNA-seq) data from heterotopic ossification samples. The number of senescent cells increased from day 3 and reached the highest level at day 21. However, the expression level of Cyclin Dependent Kinase Inhibitor 2A (Cdkn2a) has no such tendency as the change of cell amount, indicating that the expression level of Cdkn2a may be different in different types of senescent cells or the same time of senescent cell at different time points. The expression level of SASPs (senescence associated secret phenotypes) was also different in different types of senescent cells or at different time points. The GO (gene ontology) analysis revealed that the senescent cells were significantly correlated with the ossification processes, like ECM organization, cell adhesion, ossification, cartilage development, etc. Trajectory analysis showed that injury-related senescent fibroblasts (day 7 and 21) and age-related senescent fibroblasts (day 0 and 42) were in different branches. GO analysis demonstrated that injury-related senescent fibroblasts were mainly related to ossification and ECM remodeling. The KEGG (Kyoto Encyclopedia of Genes and Genomes) results revealed that the ossification was significantly corrected with protein processing in PI3K-Akt signaling, MAPK signaling, focal adhesion, etc. Conclusion Consequently, we demonstrated that, unlike age-related senescence, the injury-related senescence demonstrated significantly different SASP phenotypes. The injury-related senescence of fibroblasts is associated with heterotopic ossification formation and may act through PI3K/Akt-induced SASPs.
Collapse
Affiliation(s)
- Qiang Zhang
- Department of Orthopedics, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People’s Republic of China
| | - Dong Zhou
- Department of Orthopaedic, The Affiliated Changzhou No.2 People’s Hospital of Nanjing Medical University, Changzhou, People’s Republic of China
- Correspondence: Dong Zhou, Department of Orthopaedic, The Affiliated Hospital of Nanjing Medical University, Changzhou No. 2 People’s Hospital, Xinglong Road 29#, Changzhou, Jiangsu, 213003, People’s Republic of China, Email
| | - Yu Liang
- Department of Orthopedics, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People’s Republic of China
- Yu Liang, Department of Orthopedics, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, People’s Republic of China, Email
| |
Collapse
|
34
|
Schmitt CA, Wang B, Demaria M. Senescence and cancer - role and therapeutic opportunities. Nat Rev Clin Oncol 2022; 19:619-636. [PMID: 36045302 PMCID: PMC9428886 DOI: 10.1038/s41571-022-00668-4] [Citation(s) in RCA: 232] [Impact Index Per Article: 116.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/18/2022] [Indexed: 01/10/2023]
Abstract
Cellular senescence is a state of stable, terminal cell cycle arrest associated with various macromolecular changes and a hypersecretory, pro-inflammatory phenotype. Entry of cells into senescence can act as a barrier to tumorigenesis and, thus, could in principle constitute a desired outcome for any anticancer therapy. Paradoxically, studies published in the past decade have demonstrated that, in certain conditions and contexts, malignant and non-malignant cells with lastingly persistent senescence can acquire pro-tumorigenic properties. In this Review, we first discuss the major mechanisms involved in the antitumorigenic functions of senescent cells and then consider the cell-intrinsic and cell-extrinsic factors that participate in their switch towards a tumour-promoting role, providing an overview of major translational and emerging clinical findings. Finally, we comprehensively describe various senolytic and senomorphic therapies and their potential to benefit patients with cancer. The entry of cells into senescence can act as a barrier to tumorigenesis; however, in certain contexts senescent malignant and non-malignant cells can acquire pro-tumorigenic properties. The authors of this Review discuss the cell-intrinsic and cell-extrinsic mechanisms involved in both the antitumorigenic and tumour-promoting roles of senescent cells, and describe the potential of various senolytic and senomorphic therapeutic approaches in oncology. Cellular senescence is a natural barrier to tumorigenesis; senescent cells are widely detected in premalignant lesions from patients with cancer. Cellular senescence is induced by anticancer therapy and can contribute to some treatment-related adverse events (TRAEs). Senescent cells exert both protumorigenic and antitumorigenic effects via cell-autonomous and paracrine mechanisms. Pharmacological modulation of senescence-associated phenotypes has the potential to improve therapy efficacy and reduce the incidence of TRAEs.
Collapse
Affiliation(s)
- Clemens A Schmitt
- Charité Universitätsmedizin Berlin, Medical Department of Hematology, Oncology and Tumour Immunology, and Molekulares Krebsforschungszentrum-MKFZ, Campus Virchow Klinikum, Berlin, Germany.,Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.,Johannes Kepler University, Linz, Austria.,Kepler University Hospital, Department of Hematology and Oncology, Linz, Austria.,Deutsches Konsortium für Translationale Krebsforschung (German Cancer Consortium), Partner site Berlin, Berlin, Germany
| | - Boshi Wang
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen (UMCG), University of Groningen (RUG), Groningen, the Netherlands
| | - Marco Demaria
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen (UMCG), University of Groningen (RUG), Groningen, the Netherlands.
| |
Collapse
|
35
|
New Trends in Aging Drug Discovery. Biomedicines 2022; 10:biomedicines10082006. [PMID: 36009552 PMCID: PMC9405986 DOI: 10.3390/biomedicines10082006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 08/13/2022] [Accepted: 08/15/2022] [Indexed: 11/17/2022] Open
Abstract
Aging is considered the main risk factor for many chronic diseases that frequently appear at advanced ages. However, the inevitability of this process is being questioned by recent research that suggests that senescent cells have specific features that differentiate them from younger cells and that removal of these cells ameliorates senescent phenotype and associated diseases. This opens the door to the design of tailored therapeutic interventions aimed at reducing and delaying the impact of senescence in life, that is, extending healthspan and treating aging as another chronic disease. Although these ideas are still far from reaching the bedside, it is conceivable that they will revolutionize the way we understand aging in the next decades. In this review, we analyze the main and well-validated cellular pathways and targets related to senescence as well as their implication in aging-associated diseases. In addition, the most relevant small molecules with senotherapeutic potential, with a special emphasis on their mechanism of action, ongoing clinical trials, and potential limitations, are discussed. Finally, a brief overview of alternative strategies that go beyond the small molecule field, together with our perspectives for the future of the field, is provided.
Collapse
|
36
|
Artesunate Inhibits the Cell Growth in Colorectal Cancer by Promoting ROS-Dependent Cell Senescence and Autophagy. Cells 2022; 11:cells11162472. [PMID: 36010550 PMCID: PMC9406496 DOI: 10.3390/cells11162472] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 08/01/2022] [Accepted: 08/02/2022] [Indexed: 01/10/2023] Open
Abstract
Although artesunate has been reported to be a promising candidate for colorectal cancer (CRC) treatment, the underlying mechanisms and molecular targets of artesunate are yet to be explored. Here, we report that artesunate acts as a senescence and autophagy inducer to exert its inhibitory effect on CRC in a reactive oxygen species (ROS)-dependent manner. In SW480 and HCT116 cells, artesunate treatment led to mitochondrial dysfunction, drastically promoted mitochondrial ROS generation, and consequently inhibited cell proliferation by causing cell cycle arrest at G0/G1 phase as well as subsequent p16- and p21-mediated cell senescence. Senescent cells underwent endoplasmic reticulum stress (ERS), and the unfolded protein response (UPR) was activated via IRE1α signaling, with upregulated BIP, IRE1α, phosphorylated IRE1α (p-IRE1α), CHOP, and DR5. Further experiments revealed that autophagy was induced by artesunate treatment due to oxidative stress and ER stress. In contrast, N-Acetylcysteine (NAC, an ROS scavenger) and 3-Methyladenine (3-MA, an autophagy inhibitor) restored cell viability and attenuated autophagy in artesunate-treated cells. Furthermore, cellular free Ca2+ levels were increased and could be repressed by NAC, 3-MA, and GSK2350168 (an IRE1α inhibitor). In vivo, artesunate administration reduced the growth of CT26 cell-derived tumors in BALB/c mice. Ki67 and cyclin D1 expression was downregulated in tumor tissue, while p16, p21, p-IRE1α, and LC3B expression was upregulated. Taken together, artesunate induces senescence and autophagy to inhibit cell proliferation in colorectal cancer by promoting excessive ROS generation.
Collapse
|
37
|
Transmissible Endoplasmic Reticulum Stress Mediated by Extracellular Vesicles from Adipocyte Promoting the Senescence of Adipose-Derived Mesenchymal Stem Cells in Hypertrophic Obesity. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:7175027. [PMID: 36035215 PMCID: PMC9410860 DOI: 10.1155/2022/7175027] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 07/05/2022] [Accepted: 07/13/2022] [Indexed: 11/24/2022]
Abstract
Hypertrophic obesity, characterized by an excessive expansion of subcutaneous adipocytes, causes chronic inflammation and insulin resistance. It is the primary feature of obesity in middle-aged and elderly individuals. In the adipose microenvironment, a high level of endoplasmic reticulum (ER) stress and changes in the extracellular vesicle (EV) composition of adipocytes may cause the senescence and restrained differentiation of progenitor cells of adipose, including adipose-derived mesenchymal stem cells (ASCs). In this study, a hypertrophic obesity mouse model was established, and the effects of adipocytes on the ER stress and senescence of ASCs were observed in a coculture of control ASCs and hypertrophic obesity mouse adipocytes or their derived EVs. The adipocytes of hypertrophic obesity mice were treated with GW4869 or an iron chelation agent to observe the effects of EVs secreted by adipocytes and their iron contents on the ER stress and senescence of ASCs. Results showed higher ER stress level and senescence phenotypes in the ASCs from the hypertrophic obesity mice than in those from the control mice. The ER stress, senescence phenotypes, and ferritin level of ASCs can be aggravated by the coculture of ASCs with adipocytes or EVs released by them from the hypertrophic obesity mice. GW4869 or iron chelator treatment improved the ER stress and senescence of the ASCs cocultured with EVs released by the adipocytes of the hypertrophic obesity mice. Our findings suggest that EV-mediated transmissible ER stress is responsible for the senescence of ASCs in hypertrophic obesity mice.
Collapse
|
38
|
Fatima S, Ambreen S, Mathew A, Elwakiel A, Gupta A, Singh K, Krishnan S, Rana R, Khawaja H, Gupta D, Manoharan J, Besler C, Laufs U, Kohli S, Isermann B, Shahzad K. ER-Stress and Senescence Coordinately Promote Endothelial Barrier Dysfunction in Diabetes-Induced Atherosclerosis. Nutrients 2022; 14:2786. [PMID: 35889743 PMCID: PMC9323824 DOI: 10.3390/nu14142786] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 07/01/2022] [Accepted: 07/04/2022] [Indexed: 01/01/2023] Open
Abstract
Diabetes mellitus is hallmarked by accelerated atherosclerosis, a major cause of mortality among patients with diabetes. Efficient therapies for diabetes-associated atherosclerosis are absent. Accelerated atherosclerosis in diabetic patients is associated with reduced endothelial thrombomodulin (TM) expression and impaired activated protein C (aPC) generation. Here, we directly compared the effects of high glucose and oxidized LDL, revealing that high glucose induced more pronounced responses in regard to maladaptive unfolded protein response (UPR), senescence, and vascular endothelial cell barrier disruption. Ex vivo, diabetic ApoE-/- mice displayed increased levels of senescence and UPR markers within atherosclerotic lesions compared with nondiabetic ApoE-/- mice. Activated protein C pretreatment maintained barrier permeability and prevented glucose-induced expression of senescence and UPR markers in vitro. These data suggest that high glucose-induced maladaptive UPR and associated senescence promote vascular endothelial cell dysfunction, which-however-can be reversed by aPC. Taken together, current data suggest that reversal of glucose-induced vascular endothelial cell dysfunction is feasible.
Collapse
Affiliation(s)
- Sameen Fatima
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostic, University Hospital, 04103 Leipzig, Germany; (S.F.); (S.A.); (A.M.); (A.E.); (A.G.); (K.S.); (S.K.); (R.R.); (H.K.); (D.G.); (J.M.); (S.K.); (B.I.)
- Institute of Experimental Internal Medicine, Medical Faculty, Otto von Guericke University, 39120 Magdeburg, Germany
| | - Saira Ambreen
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostic, University Hospital, 04103 Leipzig, Germany; (S.F.); (S.A.); (A.M.); (A.E.); (A.G.); (K.S.); (S.K.); (R.R.); (H.K.); (D.G.); (J.M.); (S.K.); (B.I.)
| | - Akash Mathew
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostic, University Hospital, 04103 Leipzig, Germany; (S.F.); (S.A.); (A.M.); (A.E.); (A.G.); (K.S.); (S.K.); (R.R.); (H.K.); (D.G.); (J.M.); (S.K.); (B.I.)
| | - Ahmed Elwakiel
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostic, University Hospital, 04103 Leipzig, Germany; (S.F.); (S.A.); (A.M.); (A.E.); (A.G.); (K.S.); (S.K.); (R.R.); (H.K.); (D.G.); (J.M.); (S.K.); (B.I.)
| | - Anubhuti Gupta
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostic, University Hospital, 04103 Leipzig, Germany; (S.F.); (S.A.); (A.M.); (A.E.); (A.G.); (K.S.); (S.K.); (R.R.); (H.K.); (D.G.); (J.M.); (S.K.); (B.I.)
| | - Kunal Singh
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostic, University Hospital, 04103 Leipzig, Germany; (S.F.); (S.A.); (A.M.); (A.E.); (A.G.); (K.S.); (S.K.); (R.R.); (H.K.); (D.G.); (J.M.); (S.K.); (B.I.)
| | - Shruthi Krishnan
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostic, University Hospital, 04103 Leipzig, Germany; (S.F.); (S.A.); (A.M.); (A.E.); (A.G.); (K.S.); (S.K.); (R.R.); (H.K.); (D.G.); (J.M.); (S.K.); (B.I.)
| | - Rajiv Rana
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostic, University Hospital, 04103 Leipzig, Germany; (S.F.); (S.A.); (A.M.); (A.E.); (A.G.); (K.S.); (S.K.); (R.R.); (H.K.); (D.G.); (J.M.); (S.K.); (B.I.)
| | - Hamzah Khawaja
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostic, University Hospital, 04103 Leipzig, Germany; (S.F.); (S.A.); (A.M.); (A.E.); (A.G.); (K.S.); (S.K.); (R.R.); (H.K.); (D.G.); (J.M.); (S.K.); (B.I.)
| | - Dheerendra Gupta
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostic, University Hospital, 04103 Leipzig, Germany; (S.F.); (S.A.); (A.M.); (A.E.); (A.G.); (K.S.); (S.K.); (R.R.); (H.K.); (D.G.); (J.M.); (S.K.); (B.I.)
| | - Jayakumar Manoharan
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostic, University Hospital, 04103 Leipzig, Germany; (S.F.); (S.A.); (A.M.); (A.E.); (A.G.); (K.S.); (S.K.); (R.R.); (H.K.); (D.G.); (J.M.); (S.K.); (B.I.)
| | - Christian Besler
- Cardiology, Leipzig Heart Center, University of Leipzig, 04289 Leipzig, Germany;
| | - Ulrich Laufs
- Klinik und Poliklinik für Kardiologie, University Hospital Leipzig, 04103 Leipzig, Germany;
| | - Shrey Kohli
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostic, University Hospital, 04103 Leipzig, Germany; (S.F.); (S.A.); (A.M.); (A.E.); (A.G.); (K.S.); (S.K.); (R.R.); (H.K.); (D.G.); (J.M.); (S.K.); (B.I.)
| | - Berend Isermann
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostic, University Hospital, 04103 Leipzig, Germany; (S.F.); (S.A.); (A.M.); (A.E.); (A.G.); (K.S.); (S.K.); (R.R.); (H.K.); (D.G.); (J.M.); (S.K.); (B.I.)
| | - Khurrum Shahzad
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostic, University Hospital, 04103 Leipzig, Germany; (S.F.); (S.A.); (A.M.); (A.E.); (A.G.); (K.S.); (S.K.); (R.R.); (H.K.); (D.G.); (J.M.); (S.K.); (B.I.)
| |
Collapse
|
39
|
Hao L, Ling YY, Huang ZX, Pan ZY, Tan CP, Mao ZW. Real-time tracking of ER turnover during ERLAD by a rhenium complex via lifetime imaging. Natl Sci Rev 2022; 9:nwab194. [PMID: 35958681 PMCID: PMC9362766 DOI: 10.1093/nsr/nwab194] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 09/23/2021] [Accepted: 09/23/2021] [Indexed: 11/15/2022] Open
Abstract
Endoplasmic reticulum (ER) degradation by autophagy (ER-phagy) is a recently revealed selective autophagy pathway that plays important roles in organelle turnover and protein degradation, but the biological functions of ER-phagy are largely unknown. Here, we present an ER-targeting Re(I) tricarbonyl complex (Re-ERLAD) that can accumulate in the ER, induce ER-to-lysosome-associated degradation (ERLAD) upon visible light irradiation, and label ER buds and track their morphological alterations during ER-phagy. The emission of Re-ERLAD is sensitive to viscosity, which is a key parameter reflecting the amount of unfolded protein in the ER. Quantitative detection using two-photon fluorescence lifetime imaging microscopy shows that ER viscosity initially increases and then decreases during ERLAD, which reveals that ERLAD is a pathway for alleviating ER stress caused by unfolded proteins. In conclusion, our work presents the first specific photoinducer and tracker of ERLAD, which can be used in studying the regulatory mechanism and function of this process.
Collapse
Affiliation(s)
- Liang Hao
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, State Key Laboratory of Oncology in South China, Sun Yat-Sen University, Guangzhou 510275, China
| | - Yu-Yi Ling
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, State Key Laboratory of Oncology in South China, Sun Yat-Sen University, Guangzhou 510275, China
| | - Zhi-Xin Huang
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, State Key Laboratory of Oncology in South China, Sun Yat-Sen University, Guangzhou 510275, China
| | - Zheng-Yin Pan
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, State Key Laboratory of Oncology in South China, Sun Yat-Sen University, Guangzhou 510275, China
| | - Cai-Ping Tan
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, State Key Laboratory of Oncology in South China, Sun Yat-Sen University, Guangzhou 510275, China
| | - Zong-Wan Mao
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, State Key Laboratory of Oncology in South China, Sun Yat-Sen University, Guangzhou 510275, China
| |
Collapse
|
40
|
Li L, Hu G, Xie R, Yang J, Shi X, Jia Z, Qu X, Wang M, Wu Y. Salubrinal-mediated activation of eIF2α signaling improves oxidative stress-induced BMSCs senescence and senile osteoporosis. Biochem Biophys Res Commun 2022; 610:70-76. [DOI: 10.1016/j.bbrc.2022.04.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 04/09/2022] [Indexed: 11/26/2022]
|
41
|
Abstract
Senescence is a cellular response to a variety of stress signals that is characterized by a stable withdrawal from the cell cycle and major changes in cell morphology and physiology. While most research on senescence has been performed on non-cancer cells, it is evident that cancer cells can also mount a senescence response. In this Review, we discuss how senescence can be induced in cancer cells. We describe the distinctive features of senescent cancer cells and how these changes in cellular physiology might be exploited for the selective eradication of these cells (senolysis). We discuss activation of the host immune system as a particularly attractive way to clear senescent cancer cells. Finally, we consider the challenges and opportunities provided by a 'one-two punch' sequential treatment of cancer with pro-senescence therapy followed by senolytic therapy.
Collapse
Affiliation(s)
- Liqin Wang
- Division of Molecular Carcinogenesis, Oncode Institute, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Lina Lankhorst
- Cancer, Stem Cells & Developmental Biology programme, Utrecht University, Utrecht, The Netherlands
| | - René Bernards
- Division of Molecular Carcinogenesis, Oncode Institute, Netherlands Cancer Institute, Amsterdam, The Netherlands.
| |
Collapse
|
42
|
Changes in the Transcriptome Caused by Mutations in the Ribosomal Protein uS10 Associated with a Predisposition to Colorectal Cancer. Int J Mol Sci 2022; 23:ijms23116174. [PMID: 35682850 PMCID: PMC9181716 DOI: 10.3390/ijms23116174] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 05/24/2022] [Accepted: 05/30/2022] [Indexed: 02/05/2023] Open
Abstract
A number of mutations in the RPS20 gene encoding the ribosomal protein uS10 have been found to be associated with a predisposition to hereditary non-polyposis colorectal carcinoma (CRC). We transfected HEK293T cells with constructs carrying the uS10 minigene with mutations identical to those mentioned above and examined the effects of the produced proteins on the cellular transcriptome. We showed that uS10 with mutations p.V50SfsX23 or p.L61EfsX11 cannot be incorporated into 40S ribosomal subunits, while the protein with the missense mutation p.V54L functionally replaces the respective endogenous protein in the 40S subunit assembly and the translation process. The comparison of RNA-seq data obtained from cells producing aberrant forms of uS10 with data for those producing the wild-type protein revealed overlapping sets of upregulated and downregulated differently expressed genes (DEGs) related to several pathways. Among the limited number of upregulated DEGs, there were genes directly associated with the progression of CRC, e.g., PPM1D and PIGN. Our findings indicate that the accumulation of the mutant forms of uS10 triggers a cascade of cellular events, similar to that which is triggered when the cell responds to a large number of erroneous proteins, suggesting that this may increase the risk of cancer.
Collapse
|
43
|
Pedroza-Diaz J, Arroyave-Ospina JC, Serna Salas S, Moshage H. Modulation of Oxidative Stress-Induced Senescence during Non-Alcoholic Fatty Liver Disease. Antioxidants (Basel) 2022; 11:antiox11050975. [PMID: 35624839 PMCID: PMC9137746 DOI: 10.3390/antiox11050975] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 04/18/2022] [Accepted: 05/05/2022] [Indexed: 01/10/2023] Open
Abstract
Non-alcoholic fatty liver disease is characterized by disturbed lipid metabolism and increased oxidative stress. These conditions lead to the activation of different cellular response mechanisms, including senescence. Cellular senescence constitutes an important response to injury in the liver. Recent findings show that chronic oxidative stress can induce senescence, and this might be a driving mechanism for NAFLD progression, aggravating the disturbance of lipid metabolism, organelle dysfunction, pro-inflammatory response and hepatocellular damage. In this context, the modulation of cellular senescence can be beneficial to ameliorate oxidative stress-related damage during NAFLD progression. This review focuses on the role of oxidative stress and senescence in the mechanisms leading to NAFLD and discusses the possibilities to modulate senescence as a therapeutic strategy in the treatment of NAFLD.
Collapse
Affiliation(s)
- Johanna Pedroza-Diaz
- University Medical Center Groningen, Department of Gastroenterology and Hepatology, University of Groningen, 9712 CP Groningen, The Netherlands; (J.P.-D.); (S.S.S.); (H.M.)
- Grupo de Investigación e Innovación Biomédica GI2B, Facultad de Ciencias Exactas y Aplicadas, Instituto Tecnológico Metropolitano, Medellín 050536, Colombia
| | - Johanna C. Arroyave-Ospina
- University Medical Center Groningen, Department of Gastroenterology and Hepatology, University of Groningen, 9712 CP Groningen, The Netherlands; (J.P.-D.); (S.S.S.); (H.M.)
- Correspondence:
| | - Sandra Serna Salas
- University Medical Center Groningen, Department of Gastroenterology and Hepatology, University of Groningen, 9712 CP Groningen, The Netherlands; (J.P.-D.); (S.S.S.); (H.M.)
| | - Han Moshage
- University Medical Center Groningen, Department of Gastroenterology and Hepatology, University of Groningen, 9712 CP Groningen, The Netherlands; (J.P.-D.); (S.S.S.); (H.M.)
| |
Collapse
|
44
|
Fernandez RJ, Gardner ZJG, Slovik KJ, Liberti DC, Estep KN, Yang W, Chen Q, Santini GT, Perez JV, Root S, Bhatia R, Tobias JW, Babu A, Morley MP, Frank DB, Morrisey EE, Lengner CJ, Johnson FB. GSK3 inhibition rescues growth and telomere dysfunction in dyskeratosis congenita iPSC-derived type II alveolar epithelial cells. eLife 2022; 11:64430. [PMID: 35559731 PMCID: PMC9200405 DOI: 10.7554/elife.64430] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 05/11/2022] [Indexed: 11/27/2022] Open
Abstract
Dyskeratosis congenita (DC) is a rare genetic disorder characterized by deficiencies in telomere maintenance leading to very short telomeres and the premature onset of certain age-related diseases, including pulmonary fibrosis (PF). PF is thought to derive from epithelial failure, particularly that of type II alveolar epithelial (AT2) cells, which are highly dependent on Wnt signaling during development and adult regeneration. We use human induced pluripotent stem cell-derived AT2 (iAT2) cells to model how short telomeres affect AT2 cells. Cultured DC mutant iAT2 cells accumulate shortened, uncapped telomeres and manifest defects in the growth of alveolospheres, hallmarks of senescence, and apparent defects in Wnt signaling. The GSK3 inhibitor, CHIR99021, which mimics the output of canonical Wnt signaling, enhances telomerase activity and rescues the defects. These findings support further investigation of Wnt agonists as potential therapies for DC-related pathologies.
Collapse
Affiliation(s)
- Rafael Jesus Fernandez
- Medical Scientist Training Program, University of Pennsylvania, Philadelphia, United States
| | - Zachary J G Gardner
- Medical Scientist Training Program, University of Pennsylvania, Philadelphia, United States
| | - Katherine J Slovik
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, United States
| | - Derek C Liberti
- Cell and Molecular Biology Graduate Group, University of Pennsylvania, Philadelphia, United States
| | - Katrina N Estep
- Cell and Molecular Biology Graduate Group, University of Pennsylvania, Philadelphia, United States
| | - Wenli Yang
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, United States
| | - Qijun Chen
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, United States
| | - Garrett T Santini
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States
| | - Javier V Perez
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, United States
| | - Sarah Root
- College of Arts and Sciences and Vagelos Scholars Program, University of Pennsylvania, Philadelphia, United States
| | - Ranvir Bhatia
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States
| | - John W Tobias
- Penn Genomic Analysis Core, University of Pennsylvania, Philadelphia, United States
| | - Apoorva Babu
- Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, United States
| | - Michael P Morley
- Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, United States
| | - David B Frank
- Penn-CHOP Lung Biology Institute, Children's Hospital of Philadelphia, Philadelphia, United States
| | - Edward E Morrisey
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, United States
| | - Christopher J Lengner
- Department of Biomedical Sciences, University of Pennsylvania, Philadelphia, United States
| | - F Brad Johnson
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, United States
| |
Collapse
|
45
|
Metabolic Vulnerabilities in Multiple Myeloma. Cancers (Basel) 2022; 14:cancers14081905. [PMID: 35454812 PMCID: PMC9029117 DOI: 10.3390/cancers14081905] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 04/02/2022] [Accepted: 04/07/2022] [Indexed: 02/05/2023] Open
Abstract
Multiple myeloma (MM) remains an incurable malignancy with eventual emergence of refractory disease. Metabolic shifts, which ensure the availability of sufficient energy to support hyperproliferation of malignant cells, are a hallmark of cancer. Deregulated metabolic pathways have implications for the tumor microenvironment, immune cell function, prognostic significance in MM and anti-myeloma drug resistance. Herein, we summarize recent findings on metabolic abnormalities in MM and clinical implications driven by metabolism that may consequently inspire novel therapeutic interventions. We highlight some future perspectives on metabolism in MM and propose potential targets that might revolutionize the field.
Collapse
|
46
|
Goy E, Tomezak M, Facchin C, Martin N, Bouchaert E, Benoit J, de Schutter C, Nassour J, Saas L, Drullion C, Brodin PM, Vandeputte A, Molendi-Coste O, Pineau L, Goormachtigh G, Pluquet O, Pourtier A, Cleri F, Lartigau E, Penel N, Abbadie C. The out-of-field dose in radiation therapy induces delayed tumorigenesis by senescence evasion. eLife 2022; 11:67190. [PMID: 35302491 PMCID: PMC8933005 DOI: 10.7554/elife.67190] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 02/15/2022] [Indexed: 11/13/2022] Open
Abstract
A rare but severe complication of curative-intent radiation therapy is the induction of second primary cancers. These cancers preferentially develop not inside the planning target volume (PTV) but around, over several centimeters, after a latency period of 1–40 years. We show here that normal human or mouse dermal fibroblasts submitted to the out-of-field dose scattering at the margin of a PTV receiving a mimicked patient’s treatment do not die but enter in a long-lived senescent state resulting from the accumulation of unrepaired DNA single-strand breaks, in the almost absence of double-strand breaks. Importantly, a few of these senescent cells systematically and spontaneously escape from the cell cycle arrest after a while to generate daughter cells harboring mutations and invasive capacities. These findings highlight single-strand break-induced senescence as the mechanism of second primary cancer initiation, with clinically relevant spatiotemporal specificities. Senescence being pharmacologically targetable, they open the avenue for second primary cancer prevention.
Collapse
Affiliation(s)
- Erwan Goy
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity, Plasticity and Resistance to Therapies, F-59000 Lille, France
| | - Maxime Tomezak
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity, Plasticity and Resistance to Therapies, F-59000 Lille, France.,Univ. Lille, CNRS, UMR8520, Institut d'Electronique, Microélectronique et Nanotechnologie, F-59652 Villeneuve d'Ascq, France
| | - Caterina Facchin
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity, Plasticity and Resistance to Therapies, F-59000 Lille, France
| | - Nathalie Martin
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity, Plasticity and Resistance to Therapies, F-59000 Lille, France
| | - Emmanuel Bouchaert
- Oncovet Clinical Research, Plateforme PRECI, F-59120 Loos, France.,Oncovet, Plateforme PRECI, F-59650 Villeneuve d'Ascq, France
| | - Jerome Benoit
- Oncovet Clinical Research, Plateforme PRECI, F-59120 Loos, France.,Oncovet, Plateforme PRECI, F-59650 Villeneuve d'Ascq, France
| | - Clementine de Schutter
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity, Plasticity and Resistance to Therapies, F-59000 Lille, France
| | - Joe Nassour
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity, Plasticity and Resistance to Therapies, F-59000 Lille, France
| | - Laure Saas
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity, Plasticity and Resistance to Therapies, F-59000 Lille, France
| | - Claire Drullion
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity, Plasticity and Resistance to Therapies, F-59000 Lille, France
| | - Priscille M Brodin
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - CIIL - Centre d'Infection et d'Immunité de Lille, F-59000 Lille, France
| | - Alexandre Vandeputte
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - CIIL - Centre d'Infection et d'Immunité de Lille, F-59000 Lille, France
| | - Olivier Molendi-Coste
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011 - EGID, F-59000 Lille, France
| | - Laurent Pineau
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011 - EGID, F-59000 Lille, France
| | - Gautier Goormachtigh
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity, Plasticity and Resistance to Therapies, F-59000 Lille, France
| | - Olivier Pluquet
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity, Plasticity and Resistance to Therapies, F-59000 Lille, France
| | - Albin Pourtier
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity, Plasticity and Resistance to Therapies, F-59000 Lille, France
| | - Fabrizio Cleri
- Univ. Lille, CNRS, UMR8520, Institut d'Electronique, Microélectronique et Nanotechnologie, F-59652 Villeneuve d'Ascq, France
| | - Eric Lartigau
- Lille University, Medical School and Centre Oscar Lambret, Lille, France
| | - Nicolas Penel
- Lille University, Medical School and Centre Oscar Lambret, Lille, France
| | - Corinne Abbadie
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity, Plasticity and Resistance to Therapies, F-59000 Lille, France
| |
Collapse
|
47
|
Fakhri S, Zachariah Moradi S, DeLiberto LK, Bishayee A. Cellular senescence signaling in cancer: A novel therapeutic target to combat human malignancies. Biochem Pharmacol 2022; 199:114989. [DOI: 10.1016/j.bcp.2022.114989] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 03/05/2022] [Accepted: 03/07/2022] [Indexed: 12/26/2022]
|
48
|
Secomandi L, Borghesan M, Velarde M, Demaria M. The role of cellular senescence in female reproductive aging and the potential for senotherapeutic interventions. Hum Reprod Update 2022; 28:172-189. [PMID: 34918084 PMCID: PMC8888999 DOI: 10.1093/humupd/dmab038] [Citation(s) in RCA: 70] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 10/28/2021] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND Advanced maternal age is associated with decreased oocyte quantity and quality as well as uterine and placental dysfunctions. These changes lead to infertility, pregnancy complications and birth defects in the offspring. As the mean age of giving birth is increasing worldwide, prevention of age-associated infertility and pregnancy complications, along with the more frequent use of ART, become extremely important. Currently, significant research is being conducted to unravel the mechanisms underlying female reproductive aging. Among the potential mechanisms involved, recent evidence has suggested a contributing role for cellular senescence, a cellular state of irreversible growth arrest characterized by a hypersecretory and pro-inflammatory phenotype. Elucidating the role of senescence in female reproductive aging holds the potential for developing novel and less invasive therapeutic measures to prevent or even reverse female reproductive aging and increase offspring wellbeing. OBJECTIVE AND RATIONALE The review will summarize the positive and negative implications of cellular senescence in the pathophysiology of the female reproductive organs during aging and critically explore the use of novel senotherapeutics aiming to reverse and/or eliminate their detrimental effects. The focus will be on major senescence mechanisms of the ovaries, the uterus, and the placenta, as well as the potential and risks of using senotherapies that have been discovered in recent years. SEARCH METHODS Data for this review were identified by searches of MEDLINE, PubMed and Google Scholar. References from relevant articles using the search terms 'Cellular Senescence', 'Aging', 'Gestational age', 'Maternal Age', 'Anti-aging', 'Uterus', 'Pregnancy', 'Fertility', 'Infertility', 'Reproduction', 'Implant', 'Senolytic', 'Senostatic', 'Senotherapy' and 'Senotherapeutic' where selected. A total of 182 articles published in English between 2005 and 2020 were included, 27 of which focus on potential senotherapies for reproductive aging. Exclusion criteria were inclusion of the terms 'male' and 'plants'. OUTCOMES Aging is a major determinant of reproductive wellbeing. Cellular senescence is a basic aging mechanism, which can be exploited for therapeutic interventions. Within the last decade, several new strategies for the development and repurposing of drugs targeting senescent cells have emerged, such as modulators of the anti-inflammatory response, oxidative stress, DNA damage, and mitochondria and protein dysfunctions. Several studies of female reproductive aging and senotherapies have been discussed that show promising results for future interventions. WIDER IMPLICATIONS In most countries of the Organization for Economic Co-operation and Development, the average age at which women give birth is above 30 years. Currently, in countries such as the Netherlands, Australia, Spain, Finland, Germany and the UK, birth rates among 30- to 34-year-olds are now higher than in any other age groups. This review will provide new knowledge and scientific advancement on the senescence mechanisms during female reproductive aging, and benefit fundamental and clinical scientists and professionals in the areas of reproduction, cancer, immunobiology and fibrosis.
Collapse
Affiliation(s)
- Laura Secomandi
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen (UMCG), 9713AV Groningen, The Netherlands
| | - Michela Borghesan
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen (UMCG), 9713AV Groningen, The Netherlands
| | - Michael Velarde
- Institute of Biology, College of Science, University of the Philippines Diliman, Quezon City, PH 1101, Philippines
| | - Marco Demaria
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen (UMCG), 9713AV Groningen, The Netherlands
| |
Collapse
|
49
|
Liu D, Liu J, Zhang D, Yang W. Advances in relationship between cell senescence and atherosclerosis. Zhejiang Da Xue Xue Bao Yi Xue Ban 2022; 51:95-101. [PMID: 35576118 PMCID: PMC9109755 DOI: 10.3724/zdxbyxb-2021-0270] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 11/03/2021] [Indexed: 06/15/2023]
Abstract
Cellular senescence is a biological process associated with the degeneration of cell structure and function, which contribute to age-related diseases. Atherosclerosis is a chronic inflammatory disease that can cause a variety of cardiovascular disorders. In this article, we review the effects of cellular senescence on the development of atherosclerosis through diverse physiopathological changes, focusing on the alterations in senescent organelles and the increased senescence-associated secretory phenotype (SASP), and exploring the relevant therapeutic strategies for atherosclerosis by clearing senescent cells and reducing SASP, to provide new insights for the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Dekun Liu
- 1. Faculty of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250300, China
| | - Jiali Liu
- 1. Faculty of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250300, China
| | - Dan Zhang
- 3. Innovation Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250300, China
| | - Wenqing Yang
- 2. Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan 250300, China
| |
Collapse
|
50
|
Pena E, El Alam S, Siques P, Brito J. Oxidative Stress and Diseases Associated with High-Altitude Exposure. Antioxidants (Basel) 2022; 11:267. [PMID: 35204150 PMCID: PMC8868315 DOI: 10.3390/antiox11020267] [Citation(s) in RCA: 60] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/11/2022] [Accepted: 01/13/2022] [Indexed: 12/12/2022] Open
Abstract
Several diseases associated with high-altitude exposure affect unacclimated individuals. These diseases include acute mountain sickness (AMS), high-altitude cerebral edema (HACE), high-altitude pulmonary edema (HAPE), chronic mountain sickness (CMS), and, notably, high-altitude pulmonary hypertension (HAPH), which can eventually lead to right ventricle hypertrophy and heart failure. The development of these pathologies involves different molecules and molecular pathways that might be related to oxidative stress. Studies have shown that acute, intermittent, and chronic exposure to hypobaric hypoxia induce oxidative stress, causing alterations to molecular pathways and cellular components (lipids, proteins, and DNA). Therefore, the aim of this review is to discuss the oxidative molecules and pathways involved in the development of high-altitude diseases. In summary, all high-altitude pathologies are related to oxidative stress, as indicated by increases in the malondialdehyde (MDA) biomarker and decreases in superoxide dismutase (SOD) and glutathione peroxidase (GPx) antioxidant activity. In addition, in CMS, the levels of 8-iso-PGF2α and H2O2 are increased, and evidence strongly indicates an increase in Nox4 activity in HAPH. Therefore, antioxidant treatments seem to be a promising approach to mitigating high-altitude pathologies.
Collapse
Affiliation(s)
- Eduardo Pena
- Institute of Health Studies, Arturo Prat University, Iquique 1100000, Chile; (E.P.); (P.S.); (J.B.)
| | - Samia El Alam
- Institute of Health Studies, Arturo Prat University, Iquique 1100000, Chile; (E.P.); (P.S.); (J.B.)
| | - Patricia Siques
- Institute of Health Studies, Arturo Prat University, Iquique 1100000, Chile; (E.P.); (P.S.); (J.B.)
| | - Julio Brito
- Institute of Health Studies, Arturo Prat University, Iquique 1100000, Chile; (E.P.); (P.S.); (J.B.)
| |
Collapse
|