1
|
Wang D, Bi X, Zhao L, Xiang S, Xi W, Yang S, Wu W, Chen T, Zheng L, Chi X, Kang Y. Targeting SphK1/S1PR3 axis ameliorates sepsis-induced multiple organ injury via orchestration of macrophage polarization and glycolysis. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025; 1872:119877. [PMID: 39549732 DOI: 10.1016/j.bbamcr.2024.119877] [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: 08/28/2024] [Revised: 10/21/2024] [Accepted: 11/01/2024] [Indexed: 11/18/2024]
Abstract
Sepsis is a heterogeneous and imprecise disorder characterized by aberrant response to infection which has been accredited for detrimental impact on immune homeostasis. Recently, macrophage metabolism has been recognized as attractive targets to develop novel immunomodulatory therapy for sepsis research. However, the fine-tuning regulators dictating macrophage functions and the specific mechanisms underlying macrophage metabolic reprogramming remain largely obscure. Sphingosine-1-phosphate (S1P), a metabolic mediator of sphingolipid catabolism, predominantly formed through sphingosine kinase 1 (SphK1) catalyzing, mediates inflammation in sepsis by binding to S1P receptor 3 (S1PR3) expressed in macrophages. Here we demonstrate that SphK1/S1PR3 axis was upregulated in lipopolysaccharide (LPS)-induced macrophages and septic mice lungs, cascading the activation of proglycolytic signaling such as HIF-1α, HK2 and PFKFB3. Targeted inhibition of Sphk1 by PF-543 effectively abrogated upregulated SphK1/S1PR3 axis in vitro and in vivo. In addition, PF-543 significantly suppressed sepsis-related inflammation and multi-organ injury in vivo. Furthermore, PF-543 not only blunted key glycolytic enzymes HIF-1α, HK2, and PFKFB3 in LPS-treated macrophages but also inhibited HK2 and PFKFB3 in septic mice. Silencing or inhibiting SphK1 tempered pro-inflammatory M1 macrophages while boosted anti-inflammatory M2 macrophages. Intriguingly, S1PR3 knockdown proficiently dampened glycolysis-associated markers, retrieved LPS-modulated M1/M2 polarization and attenuated NF-κB p65 activation. In conclusion, our study provides the first evidence that PF-543 orchestrates proportional imbalance of macrophage polarization and the Warburg effect in a SphK1/S1PR3 dependent manner during sepsis, mitigating both hyperinflammation and multi-organ failure, adding a novel puzzle piece to pharmacologically exploitable therapy for sepsis.
Collapse
Affiliation(s)
- Dan Wang
- Surgical Anesthesia Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China; Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Xinwen Bi
- Surgical Anesthesia Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China; Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Le Zhao
- Surgical Anesthesia Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China; Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Shijian Xiang
- Department of Pharmacy, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China; Shenzhen Key Laboratory of Chinese Medicine Active Substance Screening and Translational Research, Shenzhen, China
| | - Wenjie Xi
- Surgical Anesthesia Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Shushu Yang
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Weijie Wu
- Surgical Anesthesia Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Tufeng Chen
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Lei Zheng
- Surgical Anesthesia Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China.
| | - Xinjin Chi
- Surgical Anesthesia Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China.
| | - Yang Kang
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China.
| |
Collapse
|
2
|
Zhang Y, Lu H, Hou L, Zhang X, Guo T, Wang R, Wang Q, Xing M. GPR120 exacerbates the immune-inflammatory response in chicken liver by mediating acetochlor induced macrophage M1 polarization. JOURNAL OF HAZARDOUS MATERIALS 2024; 485:136928. [PMID: 39709819 DOI: 10.1016/j.jhazmat.2024.136928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 12/11/2024] [Accepted: 12/16/2024] [Indexed: 12/24/2024]
Abstract
Acetochlor is a widely used and highly effective herbicide. Its overuse poses significant threats to biosecurity and ecological integrity, particularly affecting free-ranging birds. Data on its impact, especially mechanisms of liver toxicity in chickens, are lacking. Thus, we established an animal-cell-animal model to explore intrinsic mechanisms at multiple levels. We found that acetochlor exposure caused liver cell swelling, inflammatory cell accumulation, and lipid deposition. Transcriptomic analyses revealed that differential gene were mainly enriched in hepatic immune, inflammatory, and programmed cell death pathways. We next focused on the gene GPR120, conducting transfection and agonism experiments in LMH, HD11, and co-cultured cells. Acetochlor significantly increased ROS accumulation, activated the NLRP3 inflammasome, and which induced PANoptosis. HD11 cells exhibited M1 polarization with upregulated pro-inflammatory factors. Silencing GPR120 exacerbated cellular damage and immune responses, whereas its agonist, GSK7A, dramatically reduced macrophage M1 polarization and mitigated immune damage to LMH cells. Finally, we returned to animal studies, adding Omega-3-a known GPR120 agonist-to the diet. Omega-3 effectively reversed acetochlor-induced hepatitis and PANoptosis. Given that acetochlor residues pose potential threats to ecosystems and avian health, it is crucial to strengthen residue control, conduct risk assessments, and explore targeted pathways and nutritional supplementation to counteract these negative impacts.
Collapse
Affiliation(s)
- Yue Zhang
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin, Heilongjiang 150040, PR China
| | - Hongmin Lu
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin, Heilongjiang 150040, PR China
| | - Lulu Hou
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin, Heilongjiang 150040, PR China
| | - Xin Zhang
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin, Heilongjiang 150040, PR China
| | - Tiantian Guo
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin, Heilongjiang 150040, PR China
| | - Ruoqi Wang
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin, Heilongjiang 150040, PR China
| | - Qi Wang
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin, Heilongjiang 150040, PR China
| | - Mingwei Xing
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin, Heilongjiang 150040, PR China.
| |
Collapse
|
3
|
Chakraborty S, Banerjee M, Jayaraman G, Rajeswari V D. Evaluation of the health impacts and deregulation of signaling pathways in humans induced by microplastics. CHEMOSPHERE 2024; 369:143881. [PMID: 39631686 DOI: 10.1016/j.chemosphere.2024.143881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 11/29/2024] [Accepted: 12/01/2024] [Indexed: 12/07/2024]
Abstract
This review assesses the diverse health risk factors associated with microplastic (MP) exposure and their impact on cellular signaling pathways. MPs induce chronic inflammation, oxidative stress, endocrine disruption, apoptosis, and immune dysregulation. They activate signaling pathways such as NF-κB, MAPK, and Nrf2, exacerbating inflammatory responses, oxidative damage, and hormonal imbalances. Understanding the interplay between MPs and signaling pathways is crucial for elucidating the mechanisms underlying MP-induced health effects. Effective risk assessment and management strategies are essential to mitigate the adverse health impacts of MPs on human populations. This research underscores the urgent need for interdisciplinary collaboration to safeguard human health and environmental sustainability in the face of rising MP pollution. In this paper, we also assess the risk factors caused by the microplastics in the pregnant women and the development of the fetus. This review explores the potential risks and challenges associated with MP exposure in newborn babies. It is quite concerning that microplastic particles were recently found in the placental tissue of newborn children for the first time. Although it is unclear how these tiny particles affect different organs, researchers believe that these tiny particles could potentially carry harmful chemicals or disrupt the developing immune system of the fetus. This review overall focuses on the impact of microplastic disrupting different signaling including reproductive health in humans.
Collapse
Affiliation(s)
- Shreya Chakraborty
- Department of Bio-Medical Sciences, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India
| | - Manosi Banerjee
- Department of Bio-Medical Sciences, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India
| | - Gurunathan Jayaraman
- Department of Bio-Sciences, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India
| | - Devi Rajeswari V
- Department of Bio-Medical Sciences, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India.
| |
Collapse
|
4
|
Fox AC, Blazeck J. Applying metabolic control strategies to engineered T cell cancer therapies. Metab Eng 2024; 86:250-261. [PMID: 39490640 PMCID: PMC11611646 DOI: 10.1016/j.ymben.2024.10.009] [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: 07/25/2024] [Accepted: 10/22/2024] [Indexed: 11/05/2024]
Abstract
Chimeric antigen receptor (CAR) T cells are an engineered immunotherapy that express synthetic receptors to recognize and kill cancer cells. Despite their success in treating hematologic cancers, CAR T cells have limited efficacy against solid tumors, in part due to the altered immunometabolic profile within the tumor environment, which hinders T cell proliferation, infiltration, and anti-tumor activity. For instance, CAR T cells must compete for essential nutrients within tumors, while resisting the impacts of immunosuppressive metabolic byproducts. In this review, we will describe the altered metabolic features within solid tumors that contribute to immunosuppression of CAR T cells. We'll discuss how overexpression of key metabolic enzymes can enhance the ability of CAR T cells to resist corresponding tumoral metabolic changes or even revert the metabolic profile of a tumor to a less inhibitory state. In addition, metabolic remodeling is intrinsically linked to T cell activity, differentiation, and function, such that metabolic engineering strategies can also promote establishment of more or less efficacious CAR T cell phenotypes. Overall, we will show how applying metabolic engineering strategies holds significant promise in improving CAR T cells for the treatment of solid tumors.
Collapse
Affiliation(s)
- Andrea C Fox
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta GA 303332, USA
| | - John Blazeck
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta GA 303332, USA.
| |
Collapse
|
5
|
Chen Q, Shao B, Xu YN, Li X, Ren SH, Wang HD, Zhang JY, Sun CL, Liu T, Xiao YY, Zhao PY, Yang GM, Liu X, Wang H. IGF2 contributes to the immunomodulatory effects of exosomes from endometrial regenerative cells on experimental colitis. Int Immunopharmacol 2024; 140:112825. [PMID: 39079347 DOI: 10.1016/j.intimp.2024.112825] [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: 05/13/2024] [Revised: 07/19/2024] [Accepted: 07/25/2024] [Indexed: 09/01/2024]
Abstract
BACKGROUND Exosomes derived from endometrial regenerative cells (ERC-Exos) can inherit the immunomodulatory function from ERCs, however, whether ERC-Exos exhibit such effect on inflammatory bowel diseases with mucosal immune dysregulation has not been explored. Insulin-like growth factor-Ⅱ (IGF2) is considered to possess the potential to induce an anti-inflammatory phenotype in immune cells. In this study, the contribution of IGF2 in mediating the protective efficacy of ERC-Exos on colitis was investigated. METHODS Lentiviral transfection was employed to obtain IGF2-specific knockout ERC-Exos (IGF2-/--ERC-Exos). Experimental colitis mice induced by dextran sulfate sodium (DSS) were divided into the phosphate-buffered saline (untreated), ERC-Exos-treated and IGF2-/--ERC-Exos-treated groups. Colonic histopathological analysis and intestinal barrier function were explored. The infiltration of CD4+ T cells and dendritic cells (DCs) were analyzed by immunofluorescence staining and flow cytometry. The maturation and function of bone marrow-derived dendritic cells (BMDCs) in different exosome administrations were evaluated by flow cytometry, ELISA and the coculture system, respectively. RESULTS Compared with the untreated group, ERC-Exos treatment significantly attenuated DSS-induced weight loss, bloody stools, shortened colon length, pathological damage, as well as repaired the weakened intestinal mucosal barrier, including promoting the goblet cells retention, restoring the intestinal barrier integrity and enhancing the expression of tight junction proteins, while the protective effect of exosomes was impaired with the knockout of IGF2 in ERC-Exos. Additionally, IGF2-expressing ERC-Exos decreased the proportions of Th1 and Th17, increased the proportions of Treg, as well as attenuated DC infiltration and maturation in mesenteric lymph nodes and lamina propria of the colitis mice. ERC-Exos were also observed to be phagocytosed by BMDCs and IGF2 is responsible for the modulating effect of ERC-Exos on BMDCs in vitro. CONCLUSIONS Exosomes derived from ERCs can exert a therapeutic effect on experimental colitis with remarkable alleviation of the intestinal barrier damage and the abnormal mucosal immune responses. We emphasized that IGF2 plays a critical role for ERC-Exos mediated immunomodulatory function and protection against colitis.
Collapse
Affiliation(s)
- Qiang Chen
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China; Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China.
| | - Bo Shao
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China; Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China.
| | - Yi-Ni Xu
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China; Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China.
| | - Xiang Li
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China; Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China.
| | - Shao-Hua Ren
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China; Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China; Department of General Surgery, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China.
| | - Hong-da Wang
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China; Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China.
| | - Jing-Yi Zhang
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China; Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China.
| | - Cheng-Lu Sun
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China; Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China.
| | - Tong Liu
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China; Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China.
| | - Yi-Yi Xiao
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China; Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China.
| | - Peng-Yu Zhao
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China; Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China.
| | - Guang-Mei Yang
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China; Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China.
| | - Xu Liu
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China; Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China.
| | - Hao Wang
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China; Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China; Tianjin Key Laboratory of Precise Vascular Reconstruction and Organ Function Repair, Tianjin, China.
| |
Collapse
|
6
|
Lee SK, Kwon JH, Jang JW, Bae SH, Yoon SK, Jung ES, Choi JY. The Critical Role of Regulatory T Cells in Immune Tolerance and Rejection Following Liver Transplantation: Interactions With the Gut Microbiome. Transplantation 2024:00007890-990000000-00891. [PMID: 39375899 DOI: 10.1097/tp.0000000000005220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/09/2024]
Abstract
Liver transplantation (LT) is the ultimate treatment for patients with end-stage liver disease or early hepatocellular carcinoma. In the context of LT, because of the unique immunological characteristics of human liver allograft, 5%-20% of selected LT recipients can achieve operational tolerance. Nonetheless, there remains a risk of rejection in LT patients. Maintaining immune homeostasis is thus crucial for improving clinical outcomes in these patients. In mechanism, several immune cells, including dendritic cells, Kupffer cells, myeloid-derived suppressor cells, hepatic stellate cells, regulatory B cells, and CD4+ regulatory T cells (Treg), contribute to achieving tolerance following LT. In terms of Treg, it plays a role in successfully minimizing immunosuppression or achieving tolerance post-LT while also reducing the risk of rejection. Furthermore, the gut microbiome modulates systemic immune functions along the gut-liver axis. Recent studies have explored changes in the microbiome and its metabolites under various conditions, including post-LT, acute rejection, and tolerance. Certain functional microbiomes and metabolites exhibit immunomodulatory functions, such as the augmentation of Treg, influencing immune homeostasis. Therefore, understanding the mechanisms of tolerance in LT, the role of Treg in tolerance and rejection, as well as their interactions with gut microbiome, is vital for the management of LT patients.
Collapse
Affiliation(s)
- Soon Kyu Lee
- Division of Hepatology, Department of Internal Medicine, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- The Catholic University Liver Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jung Hyun Kwon
- Division of Hepatology, Department of Internal Medicine, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- The Catholic University Liver Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jeong Won Jang
- The Catholic University Liver Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Division of Hepatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Si Hyun Bae
- The Catholic University Liver Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Division of Hepatology, Department of Internal Medicine, Eunpyeong St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Seung Kew Yoon
- The Catholic University Liver Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Division of Hepatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Eun Sun Jung
- Department of Pathology, Eunpyeong St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jong Young Choi
- The Catholic University Liver Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Division of Hepatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| |
Collapse
|
7
|
Mashayekhi-Sardoo H, Rezaee R, Yarmohammadi F, Karimi G. Targeting Endoplasmic Reticulum Stress by Natural and Chemical Compounds Ameliorates Cisplatin-Induced Nephrotoxicity: A Review. Biol Trace Elem Res 2024:10.1007/s12011-024-04351-w. [PMID: 39212819 DOI: 10.1007/s12011-024-04351-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 08/18/2024] [Indexed: 09/04/2024]
Abstract
Cisplatin is a chemotherapeutic that dose-dependently causes renal complications such as decreased kidney function and acute kidney injury. The endoplasmic reticulum (ER) is responsible for calcium homeostasis and protein folding and plays a major part in cisplatin's nephrotoxicity. The current article reviews how chemical and natural compounds modulate cisplatin-induced apoptosis, autophagy, and inflammation by inhibiting ER stress signaling pathways. The available evidence indicates that natural compounds (Achyranthes aspera water-soluble extract, morin hydrate, fucoidan, isoliquiritigenin, leonurine, epigallocatechin-3-gallate, grape seed proanthocyanidin, and ginseng polysaccharide) and chemicals (Sal003, NSC228155, TUG891, dorsomorphin (compound C), HC-030031, dexmedetomidine, and recombinant human erythropoietin (rHuEpo)) can alleviate cisplatin nephrotoxicity by suppression of ER stress signaling pathways including IRE1α/ASK1/JNK, PERK-eIF2α-ATF4, and ATF6, as well as PI3K/AKT signaling pathway. Since ER and related signaling pathways are important in cisplatin nephrotoxicity, agents that can inhibit the abovementioned signaling pathways may hold promise in alleviating this untoward adverse effect.
Collapse
Affiliation(s)
- Habibeh Mashayekhi-Sardoo
- Bio Environmental Health Hazards Research Center, Jiroft University of Medical Sciences, Jiroft, Iran
- Student Research Committee, Jiroft University of Medical Sciences, Jiroft, Iran
| | - Ramin Rezaee
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh Yarmohammadi
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Gholamreza Karimi
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
- Pharmaceutical Research Center, Institute of Pharmaceutical Technology, Mashhad University of Medical, P. O. Box, Sciences, Mashhad, 1365-91775, Iran.
| |
Collapse
|
8
|
Aria H, Azizi M, Nazem S, Mansoori B, Darbeheshti F, Niazmand A, Daraei A, Mansoori Y. Competing endogenous RNAs regulatory crosstalk networks: The messages from the RNA world to signaling pathways directing cancer stem cell development. Heliyon 2024; 10:e35208. [PMID: 39170516 PMCID: PMC11337742 DOI: 10.1016/j.heliyon.2024.e35208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Revised: 07/08/2024] [Accepted: 07/24/2024] [Indexed: 08/23/2024] Open
Abstract
Cancer stem cells (CSCs) are one of the cell types that account for cancer heterogeneity. The cancer cells arrest in G0 and generate non-CSC progeny through self-renewal and pluripotency, resulting in tumor recurrence, metastasis, and resistance to chemotherapy. They can stimulate tumor relapse and re-grow a metastatic tumor. So, CSCs is a promising target for eradicating tumors, and developing an anti-CSCs therapy has been considered. In recent years competing endogenous RNA (ceRNA) has emerged as a significant class of post-transcriptional regulators that affect gene expression via competition for microRNA (miRNA) binding. Furthermore, aberrant ceRNA expression is associated with tumor progression. Recent findings show that ceRNA network can cause tumor progression through the effect on CSCs. To overcome therapeutic resistance due to CSCs, we need to improve our current understanding of the mechanisms by which ceRNAs are implicated in CSC-related relapse. Thus, this review was designed to discuss the role of ceRNAs in CSCs' function. Targeting ceRNAs may open the path for new cancer therapeutic targets and can be used in clinical research.
Collapse
Affiliation(s)
- Hamid Aria
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mahdieh Azizi
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Shima Nazem
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Behnam Mansoori
- Pediatrics Department, School of Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Farzaneh Darbeheshti
- Department of Radiation Oncology, Dana-Farber Cancer Institute and Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Anoosha Niazmand
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Abdolreza Daraei
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
- Department of Medical Genetics, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Yaser Mansoori
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
- Department of Medical Genetics, Fasa University of Medical Sciences, Fasa, Iran
| |
Collapse
|
9
|
Ghorbani A, Chatanaka MK, Avery LM, Wang M, Brown J, Cohen R, Gorham T, Misaghian S, Padmanabhan N, Romero D, Stengelin M, Mathew A, Sigal G, Wohlstadter J, Horbinski C, McCortney K, Xu W, Zadeh G, Mansouri A, Yousef GM, Diamandis EP, Prassas I. Glial fibrillary acidic protein, neurofilament light, matrix metalloprotease 3 and fatty acid binding protein 4 as non-invasive brain tumor biomarkers. Clin Proteomics 2024; 21:41. [PMID: 38879494 PMCID: PMC11179213 DOI: 10.1186/s12014-024-09492-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 05/29/2024] [Indexed: 06/19/2024] Open
Abstract
BACKGROUND Gliomas are aggressive malignant tumors, with poor prognosis. There is an unmet need for the discovery of new, non-invasive biomarkers for differential diagnosis, prognosis, and management of brain tumors. Our objective is to validate four plasma biomarkers - glial fibrillary acidic protein (GFAP), neurofilament light (NEFL), matrix metalloprotease 3 (MMP3) and fatty acid binding protein 4 (FABP4) - and compare them with established brain tumor molecular markers and survival. METHODS Our cohort consisted of patients with benign and malignant brain tumors (GBM = 77, Astrocytomas = 26, Oligodendrogliomas = 23, Secondary tumors = 35, Meningiomas = 70, Schwannomas = 15, Pituitary adenomas = 15, Normal individuals = 30). For measurements, we used ultrasensitive electrochemiluminescence multiplexed immunoassays. RESULTS High plasma GFAP concentration was associated with GBM, low GFAP and high FABP4 were associated with meningiomas, and low GFAP and low FABP4 were associated with astrocytomas and oligodendrogliomas. NEFL was associated with progression of disease. Several prognostic genetic alterations were significantly associated with all plasma biomarker levels. We found no independent associations between plasma GFAP, NEFL, FABP4 and MMP3, and overall survival. The candidate biomarkers could not reliably discriminate GBM from primary or secondary CNS lymphomas. CONCLUSIONS GFAP, NEFL, FABP4 and MMP3 are useful for differential diagnosis and prognosis, and are associated with molecular changes in gliomas.
Collapse
Affiliation(s)
- Atefeh Ghorbani
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Miyo K Chatanaka
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Lisa M Avery
- Biostatistics Division, Dalla Lana School of Public Health, University of Toronto, Toronto, Canada
- Department of Biostatistics, The Princess Margaret Cancer Centre, University of Toronto, Toronto, Canada
| | - Mingyue Wang
- Meso Scale Diagnostics, LLC., Rockville, MD, USA
| | | | - Rachel Cohen
- Meso Scale Diagnostics, LLC., Rockville, MD, USA
| | - Taron Gorham
- Meso Scale Diagnostics, LLC., Rockville, MD, USA
| | | | | | | | | | - Anu Mathew
- Meso Scale Diagnostics, LLC., Rockville, MD, USA
| | - George Sigal
- Meso Scale Diagnostics, LLC., Rockville, MD, USA
| | | | - Craig Horbinski
- Feinberg School of Medicine, Northwestern Medicine, Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, USA
| | - Katy McCortney
- Feinberg School of Medicine, Northwestern Medicine, Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, USA
| | - Wei Xu
- Biostatistics Division, Dalla Lana School of Public Health, University of Toronto, Toronto, Canada
- Department of Biostatistics, The Princess Margaret Cancer Centre, University of Toronto, Toronto, Canada
| | - Gelareh Zadeh
- MacFeeters Hamilton Neuro-Oncology Program, Princess Margaret Cancer Centre, University Health Network and University of Toronto, Toronto, ON, Canada
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, ON, Canada
| | - Alireza Mansouri
- Department of Neurosurgery, Hershey Medical Center, Hershey, PA, USA
- Penn State Cancer Institute, Hershey Medical Center, Hershey, PA, USA
| | - George M Yousef
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
- Laboratory Medicine Program, University Health Network, Toronto, Canada
| | - Eleftherios P Diamandis
- Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, Canada.
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Canada.
| | - Ioannis Prassas
- Laboratory Medicine Program, University Health Network, Toronto, Canada.
| |
Collapse
|
10
|
Zhang J, Li H, Zhong H, Chen X, Hu ZX. Omega-3 polyunsaturated fatty acids protect peritoneal mesothelial cells from hyperglycolysis and mesothelial-mesenchymal transition through the FFAR4/CaMKKβ/AMPK/mTOR signaling pathway. Int Immunopharmacol 2024; 128:111561. [PMID: 38262160 DOI: 10.1016/j.intimp.2024.111561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 01/14/2024] [Accepted: 01/15/2024] [Indexed: 01/25/2024]
Abstract
Peritoneal fibrosis is a severe clinical complication associated with peritoneal dialysis (PD) and impacts its efficacy and patient outcomes. The process of mesothelial-mesenchymal transition (MMT) in peritoneal mesothelial cells plays a pivotal role in fibrogenesis, whereas metabolic reprogramming, characterized by excessive glycolysis, is essential in MMT development. No reliable therapies are available despite substantial progress made in understanding the mechanisms underlying peritoneal fibrosis. Protective effect of omega-3 polyunsaturated fatty acids (ω3 PUFAs) has been described in PD-induced peritoneal fibrosis, although the detailed mechanisms remain unknown. It is known that ω3 PUFAs bind to and activate the free fatty acid receptor 4 (FFAR4). However, the expression and role of FFAR4 in the peritoneum have not been investigated. Thus, we hypothesized that ω3 PUFAs would alleviate peritoneal fibrosis by inhibiting hyperglycolysis and MMT through FFAR4 activation. First, we determined FFAR4 expression in peritoneal mesothelium in humans and mice. FFAR4 expression was abnormally decreased in patients on PD and mice and HMrSV5 mesothelial cells exposed to PD fluid (PDF); this change was restored by the ω3 PUFAs (EPA and DHA). ω3 PUFAs significantly inhibited peritoneal hyperglycolysis, MMT, and fibrosis in PDF-treated mice and HMrSV5 mesothelial cells; these changes induced by ω3 PUFAs were blunted by treatment with the FFAR4 antagonist AH7614 and FFAR4 siRNA. Additionally, ω3 PUFAs induced FFAR4, Ca2+/calmodulin-dependent protein kinase kinase β (CaMKKβ), and AMPK and suppressed mTOR, leading to the inhibition of hyperglycolysis, demonstrating that the ω3 PUFAs-mediated FFAR4 activation ameliorated peritoneal fibrosis by inhibiting hyperglycolysis and MMT via CaMKKβ/AMPK/mTOR signaling. As natural FFAR4 agonists, ω3 PUFAs may be considered for the treatment of PD-associated peritoneal fibrosis.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Nephrology, West China Hospital, Sichuan University, Chengdu, China
| | - Hao Li
- Department of Nephrology, West China Hospital, Sichuan University, Chengdu, China
| | - Hui Zhong
- Department of Nephrology, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaoting Chen
- Animal Experimental Center, West China Hospital, Sichuan University, Chengdu, China
| | - Zhang-Xue Hu
- Department of Nephrology, West China Hospital, Sichuan University, Chengdu, China; National Clinical Research Center for Geriatrics and Kidney Research Institute, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
11
|
Kim S, Lee S, Kim TY, Lee SH, Seo SU, Kweon MN. Newly isolated Lactobacillus paracasei strain modulates lung immunity and improves the capacity to cope with influenza virus infection. MICROBIOME 2023; 11:260. [PMID: 37996951 PMCID: PMC10666316 DOI: 10.1186/s40168-023-01687-8] [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: 05/23/2023] [Accepted: 10/01/2023] [Indexed: 11/25/2023]
Abstract
BACKGROUND The modulation of immune responses by probiotics is crucial for local and systemic immunity. Recent studies have suggested a correlation between gut microbiota and lung immunity, known as the gut-lung axis. However, the evidence and mechanisms underlying this axis remain elusive. RESULTS In this study, we screened various Lactobacillus (L.) strains for their ability to augment type I interferon (IFN-I) signaling using an IFN-α/β reporter cell line. We identified L. paracasei (MI29) from the feces of healthy volunteers, which showed enhanced IFN-I signaling in vitro. Oral administration of the MI29 strain to wild-type B6 mice for 2 weeks resulted in increased expression of IFN-stimulated genes and pro-inflammatory cytokines in the lungs. We found that MI29-treated mice had significantly increased numbers of CD11c+PDCA-1+ plasmacytoid dendritic cells and Ly6Chi monocytes in the lungs compared with control groups. Pre-treatment with MI29 for 2 weeks resulted in less weight loss and lower viral loads in the lung after a sub-lethal dose of influenza virus infection. Interestingly, IFNAR1-/- mice did not show enhanced viral resistance in response to oral MI29 administration. Furthermore, metabolic profiles of MI29-treated mice revealed changes in fatty acid metabolism, with MI29-derived fatty acids contributing to host defense in a Gpr40/120-dependent manner. CONCLUSIONS These findings suggest that the newly isolated MI29 strain can activate host defense immunity and prevent infections caused by the influenza virus through the gut-lung axis. Video Abstract.
Collapse
Affiliation(s)
- Seungil Kim
- Mucosal Immunology Laboratory, Department of Convergence Medicine, University of Ulsan College of Medicine/Asan Medical Center, Seoul, Republic of Korea
- Digestive Diseases Research Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Sohyeon Lee
- Mucosal Immunology Laboratory, Department of Convergence Medicine, University of Ulsan College of Medicine/Asan Medical Center, Seoul, Republic of Korea
| | - Tae-Young Kim
- Mucosal Immunology Laboratory, Department of Convergence Medicine, University of Ulsan College of Medicine/Asan Medical Center, Seoul, Republic of Korea
| | - Su-Hyun Lee
- Mucosal Immunology Laboratory, Department of Convergence Medicine, University of Ulsan College of Medicine/Asan Medical Center, Seoul, Republic of Korea
| | - Sang-Uk Seo
- Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Mi-Na Kweon
- Mucosal Immunology Laboratory, Department of Convergence Medicine, University of Ulsan College of Medicine/Asan Medical Center, Seoul, Republic of Korea.
- Digestive Diseases Research Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
12
|
Mottaghi S, Abbaszadeh H. Grape seed extract in combination with deferasirox ameliorates iron overload, oxidative stress, inflammation, and liver dysfunction in beta thalassemia children. Complement Ther Clin Pract 2023; 53:101804. [PMID: 37832335 DOI: 10.1016/j.ctcp.2023.101804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 09/28/2023] [Accepted: 10/01/2023] [Indexed: 10/15/2023]
Abstract
BACKGROUND AND PURPOSE Iron overload in the body is associated with serious and irreversible tissue damage. This study aimed to investigate the iron-chelating, antioxidant, anti-inflammatory, and hepatoprotective activities of grape seed extract (GSE) supplement as well as its safety in β-thalassemia major (β-TM) pediatric patients receiving deferasirox as a standard iron-chelation therapy. MATERIALS AND METHODS The children were randomly allocated to either GSE group (n = 30) or control group (n = 30) to receive GSE (100 mg/day) or placebo capsules, respectively, for 4 weeks. The serum levels of iron, ferritin, total iron-binding capacity (TIBC), alanine transaminase (ALT), aspartate aminotransferase (AST), tumor necrosis factor alpha (TNF-α), high-sensitivity C-reactive protein (hs-CRP), malondialdehyde (MDA), and glutathione (GSH) as well as superoxide dismutase (SOD) activity and hemoglobin (Hb) concentration were measured pre-and post-intervention. RESULTS GSE supplement significantly attenuated the serum levels of iron (p = 0.030), ferritin (p = 0.017), ALT (p = 0.000), AST (p = 0.000), TNF-α (p = 0.000), and hs-CRP (p = 0.001). The TIBC level (p = 0.020) significantly enhanced in the GSE group compared with the placebo group. Moreover, GSE supplement remarkably improved the oxidative stress markers, MDA (p = 0.000) and GSH (p = 0.001). The changes in the SOD activity (p = 0.590) and Hb concentration (p = 0.670) were not statistically different between the groups. CONCLUSION GSE supplement possesses several health beneficial influences on children with β-TM by alleviating iron burden, oxidative stress, inflammation, and liver dysfunction.
Collapse
Affiliation(s)
- Sayeh Mottaghi
- Department of Pediatrics, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Hassan Abbaszadeh
- Department of Pharmacology, Faculty of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
13
|
Singh RK, Kumar S, Kumar S, Shukla A, Kumar N, Patel AK, Yadav LK, Kaushalendra, Antiwal M, Acharya A. Potential implications of protein kinase Cα in pathophysiological conditions and therapeutic interventions. Life Sci 2023; 330:121999. [PMID: 37536614 DOI: 10.1016/j.lfs.2023.121999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 07/31/2023] [Accepted: 07/31/2023] [Indexed: 08/05/2023]
Abstract
PKCα is a molecule with many functions that play an important role in cell survival and death to maintain cellular homeostasis. Alteration in the normal functioning of PKCα is responsible for the complicated etiology of many pathologies, including cancer, cardiovascular diseases, kidney complications, neurodegenerative diseases, diabetics, and many others. Several studies have been carried out over the years on this kinase's function, and regulation in normal physiology and pathological conditions. A lot of data with antithetical results have therefore accumulated over time to create a complex framework of physiological implications connected to the PKCα function that needs comprehensive elucidation. In light of this information, we critically analyze the multiple roles played by PKCα in basic cellular processes and their molecular mechanism during various pathological conditions. This review further discusses the current approaches to manipulating PKCα signaling amplitude in the patient's favour and proposed PKCα as a therapeutic target to reverse pathological states.
Collapse
Affiliation(s)
- Rishi Kant Singh
- Lab of Hematopoiesis and Leukemia, KSBS, Indian Institute of Technology, Delhi, New Delhi 110016, India; Cancer Immunology Lab, Department of Zoology, Banaras Hindu University, Varanasi 221005, India
| | - Sanjay Kumar
- Cancer Immunology Lab, Department of Zoology, Banaras Hindu University, Varanasi 221005, India
| | - Sandeep Kumar
- Cancer Immunology Lab, Department of Zoology, Banaras Hindu University, Varanasi 221005, India
| | - Alok Shukla
- Cancer Immunology Lab, Department of Zoology, Banaras Hindu University, Varanasi 221005, India
| | - Naveen Kumar
- Cancer Immunology Lab, Department of Zoology, Banaras Hindu University, Varanasi 221005, India
| | - Anand Kumar Patel
- Cancer Immunology Lab, Department of Zoology, Banaras Hindu University, Varanasi 221005, India
| | - Lokesh Kumar Yadav
- Cancer Immunology Lab, Department of Zoology, Banaras Hindu University, Varanasi 221005, India
| | - Kaushalendra
- Department of Zoology, Pachhunga University College Campus, Mizoram University, Aizawl 796001, India
| | - Meera Antiwal
- Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| | - Arbind Acharya
- Cancer Immunology Lab, Department of Zoology, Banaras Hindu University, Varanasi 221005, India.
| |
Collapse
|
14
|
Babaei Z, Keyvanloo Shahrestanaki M, Aghaei M. MiR-1236: Key controller of tumor development and progression: Focus on the biological functions and molecular mechanisms. Pathol Res Pract 2023; 248:154671. [PMID: 37418995 DOI: 10.1016/j.prp.2023.154671] [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: 05/24/2023] [Accepted: 07/02/2023] [Indexed: 07/09/2023]
Abstract
Combating with the cancer, as one of the leading causes of morbidity and mortality worldwide, scientific community extensively evidenced microRNA 1236 (miR-1236) roles in the pathogenesis of malignant tumors. It has been mentioned that miR-1236 target genes and signal pathways that are key controller of tumor development and progression. Consistently, increasing evidence reports that miR-1236 participates in cancer cell growth, migration, invasion, apoptosis, and drug resistance, as well as tumor diagnosis, and prognosis. MiR-1236 is also implicated in epithelial-mesenchymal transition (EMT), which is a significant indicator of the metastatic process. Moreover, miR-1236 itself is regulated by several newly discovered long non-coding RNAs (lncRNAs) and circular RNAs (circRNAs). Current review aimed to summarize and discuss different dimensions of miR-1236 involvement in the fundamental cellular and molecular mechanisms of tumor progressions. We believe that miR-1236 may serve as a non-invasive diagnostic marker and potential therapeutic target for cancer.
Collapse
Affiliation(s)
- Zeinab Babaei
- Department of Clinical Biochemistry and Biophysics, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | | | - Mahmoud Aghaei
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
15
|
Warrier S, Srinivasan S, Chedere A, Rangarajan A. Inhibition of protein translation under matrix-deprivation stress in breast cancer cells. Front Med (Lausanne) 2023; 10:1124514. [PMID: 37425300 PMCID: PMC10324034 DOI: 10.3389/fmed.2023.1124514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 05/25/2023] [Indexed: 07/11/2023] Open
Abstract
Matrix-deprivation stress leads to cell-death by anoikis, whereas overcoming anoikis is critical for cancer metastasis. Work from our lab and others has identified a crucial role for the cellular energy sensor AMPK in anoikis-resistance, highlighting a key role for metabolic reprogramming in stress survival. Protein synthesis is a major energy-consuming process that is tightly regulated under stress. Although an increase in protein synthesis in AMPK-depleted experimentally-transformed MEFs has been associated with anoikis, the status and regulation of protein translation in epithelial-origin cancer cells facing matrix-detachment remains largely unknown. Our study shows that protein translation is mechanistically abrogated at both initiation and elongation stages by the activation of the unfolded protein response (UPR) pathway and inactivation of elongation factor eEF2, respectively. Additionally, we show inhibition of the mTORC1 pathway known for regulation of canonical protein synthesis. We further functionally assay this inhibition using SUnSET assay, which demonstrates repression of global protein synthesis in MDA-MB-231 and MCF7 breast cancer cells when subjected to matrix-deprivation. In order to gauge the translational status of matrix-deprived cancer cells, we undertook polysome profiling. Our data revealed reduced but continuous mRNA translation under matrix-deprivation stress. An integrated analysis of transcriptomic and proteomic data further identifies novel targets that may aid cellular adaptations to matrix-deprivation stress and can be explored for therapeutic intervention.
Collapse
|
16
|
Carraro M, Bernardi P. The mitochondrial permeability transition pore in Ca 2+ homeostasis. Cell Calcium 2023; 111:102719. [PMID: 36963206 DOI: 10.1016/j.ceca.2023.102719] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/17/2023] [Accepted: 03/18/2023] [Indexed: 03/26/2023]
Abstract
The mitochondrial Permeability Transition Pore (PTP) can be defined as a Ca2+ activated mega-channel involved in mitochondrial damage and cell death, making its inhibition a hallmark for therapeutic purposes in many PTP-related paradigms. Although long-lasting PTP openings have been widely studied, the physiological implications of transient openings (also called "flickering" behavior) are still poorly understood. The flickering activity was suggested to play a role in the regulation of Ca2+ and ROS homeostasis, and yet this hypothesis did not reach general consensus. This state of affairs might arise from the lack of unquestionable experimental evidence, due to limitations of the available techniques for capturing transient PTP activity and to a still partial understanding of its molecular identity. In this review we will focus on possible implications of the PTP in physiology, in particular its role as a Ca2+ release pathway, discussing the consequences of its forced inhibition. We will also consider the recent hypothesis of the existence of more permeability pathways and their potential involvement in mitochondrial physiology.
Collapse
Affiliation(s)
- Michela Carraro
- Department of Biomedical Sciences, University of Padova and CNR Neuroscience Institute, Via Ugo Bassi 58/B, I-35131 Padova, Italy.
| | - Paolo Bernardi
- Department of Biomedical Sciences, University of Padova and CNR Neuroscience Institute, Via Ugo Bassi 58/B, I-35131 Padova, Italy
| |
Collapse
|
17
|
Hong T, Wang R, Yang G, Wang X, Zeng L, Yang S, Wei J, Gao Q, Zhang X. Human umbilical cord mesenchymal stem cells ameliorate acute graft versus host disease by elevating phytosphingosine. Exp Hematol 2023:S0301-472X(23)00070-X. [PMID: 36931619 DOI: 10.1016/j.exphem.2023.03.002] [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: 11/12/2022] [Revised: 02/28/2023] [Accepted: 03/08/2023] [Indexed: 03/17/2023]
Abstract
Acute graft-versus-host disease (aGVHD) is a prominent barrier to allogeneic hematopoietic stem cell transplantation (allo-HSCT) and even leads to death after HSCT. Human umbilical cord mesenchymal stem cells (HUCMSCs) are effective in aGVHD treatment and have mild side effects, but the underlying mechanisms remain unclear. Phytosphingosine (PHS) is known to prevent loss of moisture from the skin; regulate epidermal cell growth, differentiation, and apoptosis; and exert bactericidal and anti-inflammatory effects. In this study, our results revealed the efficacy of HUCMSCs in alleviating aGVHD in a murine model, with striking changes in metabolism and significantly elevated PHS levels due to sphingolipid metabolism. In vitro, PHS reduced CD4+ T cell proliferation, enhanced apoptosis and reduced T helper 1 (Th1) cell differentiation. Transcriptional analysis of donor CD4+ T cells treated with PHS revealed significant decreases in transcripts regulating proinflammatory pathways, such as NF-κB. In vivo, the administration of PHS significantly ameliorated aGVHD development. Collectively, these beneficial effects indicate proof-of-concept that sphingolipid metabolites could be a safe and effective means to prevent aGVHD in the clinic.
Collapse
Affiliation(s)
- Tao Hong
- Medical Center of Hematology, Xinqiao Hospital. State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University, Chongqing, 400037, China
| | - Rui Wang
- Medical Center of Hematology, Xinqiao Hospital. State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University, Chongqing, 400037, China; Jinfeng Laboratory, Chongqing, 401329, China
| | - Guancui Yang
- Medical Center of Hematology, Xinqiao Hospital. State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University, Chongqing, 400037, China; Department of Hematology, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637002, China
| | - Xiaoqi Wang
- Medical Center of Hematology, Xinqiao Hospital. State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University, Chongqing, 400037, China
| | - Lingyu Zeng
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221008, China
| | - Shijie Yang
- Medical Center of Hematology, Xinqiao Hospital. State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University, Chongqing, 400037, China; Jinfeng Laboratory, Chongqing, 401329, China
| | - Jin Wei
- Department of Hematology, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637002, China
| | - Qiangguo Gao
- Department of Cell Biology, College of Basic Medicine, Army Medical University, Chongqing, 400038, China..
| | - Xi Zhang
- Medical Center of Hematology, Xinqiao Hospital. State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University, Chongqing, 400037, China; Jinfeng Laboratory, Chongqing, 401329, China..
| |
Collapse
|
18
|
FFAR4 improves the senescence of tubular epithelial cells by AMPK/SirT3 signaling in acute kidney injury. Signal Transduct Target Ther 2022; 7:384. [PMID: 36450712 PMCID: PMC9712544 DOI: 10.1038/s41392-022-01254-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 11/04/2022] [Accepted: 11/06/2022] [Indexed: 12/02/2022] Open
Abstract
Acute kidney injury (AKI) is a serious clinical complication with high morbidity and mortality rates. Despite substantial progress in understanding the mechanism of AKI, no effective therapy is available for treatment or prevention. We previously found that G protein-coupled receptor (GPCR) family member free fatty acid receptor 4 (FFAR4) agonist TUG891 alleviated kidney dysfunction and tubular injury in AKI mice. However, the versatile role of FFAR4 in kidney has not been well characterized. In the study, the expression of FFAR4 was abnormally decreased in tubular epithelial cells (TECs) of cisplatin, cecal ligation/perforation and ischemia/reperfusion injury-induced AKI mice, respectively. Systemic and conditional TEC-specific knockout of FFAR4 aggravated renal function and pathological damage, whereas FFAR4 activation by TUG-891 alleviated the severity of disease in cisplatin-induced AKI mice. Notably, FFAR4, as a key determinant, was firstly explored to regulate cellular senescence both in injured kidneys of AKI mice and TECs, which was indicated by senescence-associated β-galactosidase (SA-β-gal) activity, marker protein p53, p21, Lamin B1, phospho-histone H2A.X, phospho-Rb expression, and secretory phenotype IL-6 level. Mechanistically, pharmacological activation and overexpression of FFAR4 reversed the decrease of aging-related SirT3 protein, where FFAR4 regulated SirT3 expression to exhibit anti-senescent effect via Gq subunit-mediated CaMKKβ/AMPK signaling in cisplatin-induced mice and TECs. These findings highlight the original role of tubular FFAR4 in cellular senescence via AMPK/SirT3 signaling and identify FFAR4 as a potential drug target against AKI.
Collapse
|
19
|
Azizidoost S, Nasrolahi A, Ghaedrahmati F, Kempisty B, Mozdziak P, Radoszkiewicz K, Farzaneh M. The pathogenic roles of lncRNA-Taurine upregulated 1 (TUG1) in colorectal cancer. Cancer Cell Int 2022; 22:335. [PMID: 36333703 PMCID: PMC9636703 DOI: 10.1186/s12935-022-02745-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 10/07/2022] [Indexed: 11/06/2022] Open
Abstract
Colorectal cancer (CRC) is a gastrointestinal tumor that develops from the colon, rectum, or appendix. The prognosis of CRC patients especially those with metastatic lesions remains unsatisfactory. Although various conventional methods have been used for the treatment of patients with CRC, the early detection and identification of molecular mechanisms associated with CRC is necessary. The scientific literature reports that altered expression of long non-coding RNAs (lncRNAs) contributed to the pathogenesis of CRC cells. LncRNA TUG1 was reported to target various miRNAs and signaling pathways to mediate CRC cell proliferation, migration, and metastasis. Therefore, TUG1 might be a potent predictive/prognostic biomarker for diagnosis of CRC.
Collapse
Affiliation(s)
- Shirin Azizidoost
- Atherosclerosis Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Ava Nasrolahi
- Infectious Ophthalmologic Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Farhoodeh Ghaedrahmati
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Bartosz Kempisty
- Graduate Physiology Program, North Carolina State University, 27695, Raleigh, NC, USA
| | - Paul Mozdziak
- Graduate Physiology Program, North Carolina State University, 27695, Raleigh, NC, USA
| | - Klaudia Radoszkiewicz
- Translational Platform for Regenerative Medicine, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106, Warsaw, Poland
| | - Maryam Farzaneh
- Fertility, Infertility and Perinatology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
20
|
Richardson KC, Jung K, Pardo J, Turner CT, Granville DJ. Noncytotoxic Roles of Granzymes in Health and Disease. Physiology (Bethesda) 2022; 37:323-348. [PMID: 35820180 DOI: 10.1152/physiol.00011.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Granzymes are serine proteases previously believed to play exclusive and somewhat redundant roles in lymphocyte-mediated target cell death. However, recent studies have challenged this paradigm. Distinct substrate profiles and functions have since emerged for each granzyme while their dysregulated proteolytic activities have been linked to diverse pathologies.
Collapse
Affiliation(s)
- Katlyn C Richardson
- International Collaboration on Repair Discoveries (ICORD), British Columbia Professional Firefighters' Wound Healing Laboratory, Vancouver Coastal Health Research Institute, Vancouver, British Columbia, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Karen Jung
- International Collaboration on Repair Discoveries (ICORD), British Columbia Professional Firefighters' Wound Healing Laboratory, Vancouver Coastal Health Research Institute, Vancouver, British Columbia, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Julian Pardo
- Fundación Instituto de Investigación Sanitaria Aragón (IIS Aragón), Biomedical Research Centre of Aragon (CIBA), Zaragoza, Spain.,Department of Microbiology, Radiology, Pediatrics and Public Health, University of Zaragoza, Zaragoza, Spain.,CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Zaragoza, Spain
| | - Christopher T Turner
- Faculty of Pharmacy and Pharmaceutical Sciences, Monash University, Melbourne, Victoria, Australia.,Future Industries Institute, University of South Australia, Adelaide, South Australia, Australia
| | - David J Granville
- International Collaboration on Repair Discoveries (ICORD), British Columbia Professional Firefighters' Wound Healing Laboratory, Vancouver Coastal Health Research Institute, Vancouver, British Columbia, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
21
|
Sanati M, Binabaj MM, Ahmadi SS, Aminyavari S, Javid H, Mollazadeh H, Bibak B, Mohtashami E, Jamialahmadi T, Afshari AR, Sahebkar A. Recent advances in glioblastoma multiforme therapy: A focus on autophagy regulation. Biomed Pharmacother 2022; 155:113740. [PMID: 36166963 DOI: 10.1016/j.biopha.2022.113740] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/14/2022] [Accepted: 09/21/2022] [Indexed: 11/02/2022] Open
Abstract
Despite conventional treatment options including chemoradiation, patients with the most aggressive primary brain tumor, glioblastoma multiforme (GBM), experience an average survival time of less than 15 months. Regarding the malignant nature of GBM, extensive research and discovery of novel treatments are urgently required to improve the patients' prognosis. Autophagy, a crucial physiological pathway for the degradation and recycling of cell components, is one of the exciting targets of GBM studies. Interventions aimed at autophagy activation or inhibition have been explored as potential GBM therapeutics. This review, which delves into therapeutic techniques to block or activate autophagy in preclinical and clinical research, aims to expand our understanding of available therapies battling GBM.
Collapse
Affiliation(s)
- Mehdi Sanati
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Birjand University of Medical Sciences, Birjand, Iran; Experimental and Animal Study Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Maryam Moradi Binabaj
- Non-Communicable Diseases Research Center, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Seyed Sajad Ahmadi
- Department of Physiology and Pharmacology, Faculty of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Samaneh Aminyavari
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hossein Javid
- Department of Medical Laboratory Sciences, Varastegan Institute for Medical Sciences, Mashhad, Iran
| | - Hamid Mollazadeh
- Department of Physiology and Pharmacology, Faculty of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Bahram Bibak
- Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Elmira Mohtashami
- Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Tannaz Jamialahmadi
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Surgical Oncology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir R Afshari
- Department of Physiology and Pharmacology, Faculty of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran; Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran.
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
22
|
Liu J, Sun M, Sun Y, Li H. TMEM189 promotes breast cancer through inhibition of autophagy-regulated ferroptosis. Biochem Biophys Res Commun 2022; 622:37-44. [DOI: 10.1016/j.bbrc.2022.06.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 06/08/2022] [Indexed: 11/16/2022]
|
23
|
Zhang C, Liu N. Noncoding RNAs in the Glycolysis of Ovarian Cancer. Front Pharmacol 2022; 13:855488. [PMID: 35431949 PMCID: PMC9005897 DOI: 10.3389/fphar.2022.855488] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Accepted: 03/15/2022] [Indexed: 01/11/2023] Open
Abstract
Energy metabolism reprogramming is the characteristic feature of tumors. The tumorigenesis, metastasis, and drug resistance of ovarian cancer (OC) is dependent on energy metabolism. Even under adequate oxygen conditions, OC cells tend to convert glucose to lactate, and glycolysis can rapidly produce ATP to meet their metabolic energy needs. Non-coding RNAs (ncRNAs) interact directly with DNA, RNA, and proteins to function as an essential regulatory in gene expression and tumor pathology. Studies have shown that ncRNAs regulate the process of glycolysis by interacting with the predominant glycolysis enzyme and cellular signaling pathway, participating in tumorigenesis and progression. This review summarizes the mechanism of ncRNAs regulation in glycolysis in OC and investigates potential therapeutic targets.
Collapse
Affiliation(s)
- Chunmei Zhang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Ning Liu
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|