1
|
Kemna K, van der Burg M, Lankester A, Giera M. Hematopoietic stem cell metabolism within the bone marrow niche - insights and opportunities. Bioessays 2024:e2400154. [PMID: 39506498 DOI: 10.1002/bies.202400154] [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: 06/30/2024] [Revised: 10/25/2024] [Accepted: 10/28/2024] [Indexed: 11/08/2024]
Abstract
Hematopoiesis unfolds within the bone marrow niche where hematopoietic stem cells (HSCs) play a central role in continually replenishing blood cells. The hypoxic bone marrow environment imparts peculiar metabolic characteristics to hematopoietic processes. Here, we discuss the internal metabolism of HSCs and describe external influences exerted on HSC metabolism by the bone marrow niche environment. Importantly, we suggest that the metabolic environment and metabolic cues are intertwined with HSC cell fate, and are crucial for hematopoietic processes. Metabolic dysregulation within the bone marrow niche during acute stress, inflammation, and chronic inflammatory conditions can lead to reduced HSC vitality. Additionally, we raise questions regarding metabolic stresses imposed on HSCs during implementation of stem cell protocols such as allo-SCT and gene therapy, and the potential ramifications. Enhancing our comprehension of metabolic influences on HSCs will expand our understanding of pathophysiology in the bone marrow and improve the application of stem cell therapies.
Collapse
Affiliation(s)
- Koen Kemna
- Department of Pediatrics, Laboratory for Pediatric Immunology, Willem-Alexander Children's Hospital, Leiden University Medical Center, Leiden, The Netherlands
| | - Mirjam van der Burg
- Department of Pediatrics, Laboratory for Pediatric Immunology, Willem-Alexander Children's Hospital, Leiden University Medical Center, Leiden, The Netherlands
| | - Arjan Lankester
- Department of Pediatrics, Laboratory for Pediatric Immunology, Willem-Alexander Children's Hospital, Leiden University Medical Center, Leiden, The Netherlands
| | - Martin Giera
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
2
|
Pal C. Small Molecules Targeting Mitochondria: A Mechanistic Approach to Combating Doxorubicin-Induced Cardiotoxicity. Cardiovasc Toxicol 2024:10.1007/s12012-024-09941-7. [PMID: 39495464 DOI: 10.1007/s12012-024-09941-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 10/29/2024] [Indexed: 11/05/2024]
Abstract
Doxorubicin (Dox) is a commonly used chemotherapy drug effective against a range of cancers, but its clinical application is greatly limited by dose-dependent and cumulative cardiotoxicity. Mitochondrial dysfunction is recognized as a key factor in Dox-induced cardiotoxicity, leading to oxidative stress, disrupted calcium balance, and activation of apoptotic pathways. Recent research has emphasized the potential of small molecules that specifically target mitochondria to alleviate these harmful effects. This review provides a comprehensive analysis of small molecules that offer cardioprotection by preserving mitochondrial function in the context of doxorubicin-induced cardiotoxicity (DIC). The mechanisms of action include the reduction of reactive oxygen species (ROS) production, stabilization of mitochondrial membrane potential, enhancement of mitochondrial biogenesis, and modulation of key signaling pathways involved in cell survival and apoptosis. By targeting mitochondria, these small molecules present a promising therapeutic strategy to prevent or reduce the cardiotoxic effects associated with Dox treatment. This review not only discusses the mechanistic actions of these agents but also emphasizes their potential in improving cardiovascular outcomes for cancer patients. Gaining insight into these mechanisms can help in creating more effective strategies to safeguard the heart during chemotherapy, allowing for the ongoing use of Dox with a lower risk to the patient's cardiovascular health. This review highlights the critical role of mitochondria-targeted therapies as a promising approach in addressing DIC.
Collapse
Affiliation(s)
- Chinmay Pal
- Department of Chemistry, Gobardanga Hindu College, North 24 Parganas, West Bengal, 743273, India.
| |
Collapse
|
3
|
Ye L, Zhao J, Xiao Z, Gu W, Liu X, Ajuyo NMC, Min Y, Pei Y, Wang D. Integrative Human Genetic and Cellular Analysis of the Pathophysiological Roles of AnxA2 in Alzheimer's Disease. Antioxidants (Basel) 2024; 13:1274. [PMID: 39456526 PMCID: PMC11504888 DOI: 10.3390/antiox13101274] [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: 09/02/2024] [Revised: 10/14/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024] Open
Abstract
Alzheimer's disease (AD) is an intractable and progressive neurodegenerative disease. Amyloid beta (Aβ) aggregation is the hallmark of AD. Aβ induces neurotoxicity through a variety of mechanisms, including interacting with membrane receptors to alter downstream signaling, damaging cellular or organelle membranes, interfering with protein degradation and synthesis, and inducing an excessive immune-inflammatory response, all of which lead to neuronal death and other pathological changes associated with AD. In this study, we extracted gene expression profiles from the GSE5281 and GSE97760 microarray datasets in the GEO (Gene Expression Omnibus) database, as well as from the Human Gene Database. We identified differentially expressed genes in the brain tissues of AD patients and healthy persons. Through GO, KEGG, and ROC analyses, annexin A2 (AnxA2) was identified as a putative target gene. Notably, accumulating evidence suggests that intracellular AnxA2 is a key regulator in various biological processes, including endocytosis, transmembrane transport, neuroinflammation, and apoptosis. Thus, we conducted a series of cell biology experiments to explore the biological function of AnxA2 in AD. The results indicate that AnxA2 gene knockdown primarily affects oxidative phosphorylation, cell cycle, AD, protein processing in the endoplasmic reticulum, SNARE interactions in vesicular transport, and autophagy. In SH-SY5Y cells secreting Aβ42, AnxA2 gene knockdown exacerbated Aβ42-induced cytotoxicity, including cell death, intracellular ROS levels, and neuronal senescence, altered cell cycle, and reduced ATP levels, suggesting its critical role in mitochondrial function maintenance. AnxA2 gene knockdown also exacerbated the inhibitory effect of Aβ42 on cell migration. AnxA2 overexpression reduced the inflammatory response induced by Aβ42, while its absence increased pro-inflammatory and decreased anti-inflammatory responses. Furthermore, AnxA2 gene knockdown facilitated apoptosis and decreased autophagy. These results indicated potential pathophysiological roles of AnxA2 in AD.
Collapse
Affiliation(s)
- Lianmeng Ye
- Laboratory of Biopharmaceuticals and Molecular Pharmacology, Key Laboratory of Tropical Biological Resources of the Ministry of Education of China, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China
- One Health Cooperative Innovation Center, Hainan University, Haikou 570228, China
- Department of Biotechnology, School of Life and Health Sciences, Hainan University, Haikou 570228, China
| | - Jiazheng Zhao
- Laboratory of Biopharmaceuticals and Molecular Pharmacology, Key Laboratory of Tropical Biological Resources of the Ministry of Education of China, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China
- One Health Cooperative Innovation Center, Hainan University, Haikou 570228, China
- Department of Biotechnology, School of Life and Health Sciences, Hainan University, Haikou 570228, China
| | - Zhengpan Xiao
- Laboratory of Biopharmaceuticals and Molecular Pharmacology, Key Laboratory of Tropical Biological Resources of the Ministry of Education of China, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China
- One Health Cooperative Innovation Center, Hainan University, Haikou 570228, China
- Department of Biotechnology, School of Life and Health Sciences, Hainan University, Haikou 570228, China
| | - Wenyu Gu
- Laboratory of Biopharmaceuticals and Molecular Pharmacology, Key Laboratory of Tropical Biological Resources of the Ministry of Education of China, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China
- One Health Cooperative Innovation Center, Hainan University, Haikou 570228, China
- Department of Biotechnology, School of Life and Health Sciences, Hainan University, Haikou 570228, China
| | - Xiaoxuan Liu
- Laboratory of Biopharmaceuticals and Molecular Pharmacology, Key Laboratory of Tropical Biological Resources of the Ministry of Education of China, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China
- One Health Cooperative Innovation Center, Hainan University, Haikou 570228, China
- Department of Biotechnology, School of Life and Health Sciences, Hainan University, Haikou 570228, China
| | - Nuela Manka’a Che Ajuyo
- Laboratory of Biopharmaceuticals and Molecular Pharmacology, Key Laboratory of Tropical Biological Resources of the Ministry of Education of China, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China
- One Health Cooperative Innovation Center, Hainan University, Haikou 570228, China
| | - Yi Min
- One Health Cooperative Innovation Center, Hainan University, Haikou 570228, China
- Department of Biotechnology, School of Life and Health Sciences, Hainan University, Haikou 570228, China
| | - Yechun Pei
- One Health Cooperative Innovation Center, Hainan University, Haikou 570228, China
- Department of Biotechnology, School of Life and Health Sciences, Hainan University, Haikou 570228, China
| | - Dayong Wang
- Laboratory of Biopharmaceuticals and Molecular Pharmacology, Key Laboratory of Tropical Biological Resources of the Ministry of Education of China, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China
- One Health Cooperative Innovation Center, Hainan University, Haikou 570228, China
| |
Collapse
|
4
|
Long B, Zhou H, Xiao L, Jiang X, Li J, Ma Z, He N, Xin W, Zhang B, Zhu X, Yu Z, Jiao Z. Targeting NUF2 suppresses gastric cancer progression through G2/M phase arrest and apoptosis induction. Chin Med J (Engl) 2024; 137:2437-2451. [PMID: 39193700 PMCID: PMC11479523 DOI: 10.1097/cm9.0000000000003006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Indexed: 08/29/2024] Open
Abstract
BACKGROUND Gastric cancer (GC), a malignant tumor with poor prognosis, is one of the leading causes of cancer-related deaths worldwide; consequently, identifying novel therapeutic targets is crucial for its corresponding treatment. NUF2 , a component of the NDC80 kinetochore complex, promotes cancer progression in multiple malignancies. Therefore, this study aimed to explore the potential of NUF2 as a therapeutic target to inhibit GC progression. METHODS Clinical samples were obtained from patients who underwent radical resection of GC at Lanzhou University Second Hospital from 2016 to 2021. Cell count assays, colony formation assays, and cell-derived xenotransplantation (CDX) models were used to determine the effects of NUF2 on GC progression. Flow cytometry was used to detect the effect of NUF2 or quercetin on cell cycle progression and apoptosis. A live-cell time-lapse imaging assay was performed to determine the effect of NUF2 on the regulation of mitotic progression. Transcriptomics was used to investigate the NUF2 -associated molecular mechanisms. Virtual docking and microscale thermophoresis were used to identify NUF2 inhibitors. Finally, CDX, organoid, and patient-derived xenograft (PDX) models were used to examine the efficacy of the NUF2 inhibitor in GC. RESULTS NUF2 expression was significantly increased in GC and was negatively correlated with prognosis. The deletion of NUF2 suppressed GC progression both in vivo and in vitro . NUF2 significantly regulated the mitogen-activated protein kinase (MAPK) pathway, promoted G2/M phase transition, and inhibited apoptosis in GC cells. Additionally, quercetin was identified as a selective NUF2 inhibitor with low toxicity that significantly suppressed tumor growth in GC cells, organoids, CDX, and PDX models. CONCLUSIONS Collectively, NUF2 -mediated G2/M phase transition and apoptosis inhibition promoted GC progression; additionally, NUF2 inhibitors exhibited potent anti-GC activity. This study provides a new strategy for targeting NUF2 to suppress GC progression in clinical settings.
Collapse
Affiliation(s)
- Bo Long
- The First Department of General Surgery, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, Gansu 730000, China
| | - Huinian Zhou
- The First Department of General Surgery, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, Gansu 730000, China
| | - Lixia Xiao
- The First Department of General Surgery, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, Gansu 730000, China
| | - Xiangyan Jiang
- The First Department of General Surgery, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, Gansu 730000, China
| | - Jian Li
- The First Department of General Surgery, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, Gansu 730000, China
| | - Zhijian Ma
- The First Department of General Surgery, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, Gansu 730000, China
| | - Na He
- The First Department of General Surgery, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, Gansu 730000, China
| | - Wei Xin
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, Gansu 730000, China
| | - Boya Zhang
- The Second Clinical Medical School of Lanzhou University, Lanzhou University, Lanzhou, Gansu 730000, China
| | - Xiaoqin Zhu
- The Second Clinical Medical School of Lanzhou University, Lanzhou University, Lanzhou, Gansu 730000, China
| | - Zeyuan Yu
- The First Department of General Surgery, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, Gansu 730000, China
| | - Zuoyi Jiao
- The First Department of General Surgery, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, Gansu 730000, China
| |
Collapse
|
5
|
Duan BT, Zhang HY, Song ZH, Han XY, Cui KL, Xu T, Zhang Y, Zhao YJ, Lei X, Tan F, Guo LL, Yang HL, Zhang L, Bai R, Lv XL, Zheng MX. EtROP38 suppresses apoptosis of host cells infected with Eimeria tenella by inhibition of the p38MAPK pathway. Vet Parasitol 2024; 331:110296. [PMID: 39217762 DOI: 10.1016/j.vetpar.2024.110296] [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: 03/20/2024] [Revised: 08/22/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
Coccidiosis is an important parasitic disease that has serious adverse effects on the global poultry industry. The mechanism by which the pathogenic factors of Eimeria tenella damage host cells is unknown. Some kinases from the rhoptry compartment can regulate apoptosis of host cells. This study focused on revealing the role and critical nodes of E. tenella rhoptry protein (EtROP) 38 in controlling the apoptosis of host cells via the P38 mitogen-activated protein kinase (MAPK) signaling pathway. The cells were treated with EtROP38 protein, siRNA p38MAPK, or both. The rate of infection, apoptosis, and the dynamic changes in the expression and activation of key factor genes of the P38MAPK signaling pathway in host cells infected with E. tenella were measured. The results showed that the addition of EtROP38 and/or knockdown of the host cells p38 gene reduced the apoptosis rate of cecal epithelial cells (CECS), decreased the mRNA expressions of p38, p53, c-myc, c-fos, and c-jun and increased the expression of p65, decreased the protein expressions of c-myc, c-fos, and c-jun, decreased the p38 protein phosphorylation level, and increased the p65 protein phosphorylation level in CECS. When E. tenella was inoculated for 4-96 h, the addition of Et ROP38 and/or host cell p38 knockdown both increased the infection rate of host cells, and this effect was more pronounced with the addition of EtROP38 with the host cell p38 knockdown. These observations indicate that E. tenella can inhibits the activation of the p38MAPK signaling pathway in host cells via EtROP38, which suppresses apoptosis in host cells.
Collapse
Affiliation(s)
- Bu-Ting Duan
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong, China
| | - Hao-Yu Zhang
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong, China
| | - Zi-Hao Song
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong, China
| | - Xiao-Yi Han
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong, China
| | - Kai-Ling Cui
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong, China
| | - Tong Xu
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong, China
| | - Yu Zhang
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong, China
| | - Yong-Juan Zhao
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong, China; School of Food and Environment, Jinzhong College of Information, Taigu, Jinzhong, China
| | - Xuan Lei
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong, China
| | - Fan Tan
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong, China
| | - Lu-Lu Guo
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong, China
| | - Hui-Lin Yang
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong, China
| | - Li Zhang
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong, China
| | - Rui Bai
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong, China
| | - Xiao-Ling Lv
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong, China
| | - Ming-Xue Zheng
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong, China.
| |
Collapse
|
6
|
Huang Z, Xu L, Wu Z, Xiong X, Luo L, Wen Z. CDC25B Is a Prognostic Biomarker Associated With Immune Infiltration and Drug Sensitivity in Hepatocellular Carcinoma. Int J Genomics 2024; 2024:8922878. [PMID: 39371450 PMCID: PMC11455594 DOI: 10.1155/2024/8922878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 09/09/2024] [Indexed: 10/08/2024] Open
Abstract
Cell division cycle 25B (CDC25B), a member of the CDC25 phosphatase family, plays a key role in cell cycle regulation. Studies have suggested its carcinogenic potential in various cancers, but the role of CDC25B in the development of hepatocellular carcinoma (HCC) remains poorly understood. The aim of this study was to clarify the role of CDC25B in HCC using bioinformatics and experiments. CDC25B expression data of HCC cancer tissues and paracancerous normal samples were obtained from The Cancer Gene Atlas (TCGA) and Gene Expression Omnibus (GEO) databases, and the relationship between CDC25B expression and the prognosis and degree of tumor differentiation of HCC patients was analyzed. CDC25B expression was verified in clinical HCC tissue samples using fluorescence quantitative polymerase chain reaction (q-PCR) and protein immunoblotting (Western blot). Gene set enrichment analysis (GSEA) was used to identify signaling pathways enriched in CDC25B expression, and differential genes (DEGs) were used to screen out coexpressed hub genes and construct protein-protein interaction (PPI) networks. 5-Ethynyl-2'-deoxyuridine (EDU) staining was used to compare the proliferation and differentiation ability of the HCC cell line (HCC-LM3) after knockdown of CDC25B. Finally, we investigated the mutation of CDC25B in HCC and the relationship between CDC25B expression and tumor cell infiltration of lymphocytes and some immune checkpoints as well as drug sensitivity. CDC25B was overexpressed in HCC tissues and correlated with poor prognosis and the degree of tumor differentiation in patients with HCC. The GSEA and PPI networks together revealed significantly upregulated signaling pathways, as well as functions, associated with the development of HCC when CDC25B was overexpressed. The EDU assay demonstrated that the ability of cells to differentiate value addedly was markedly reduced following the downregulation of CDC25B expression in HCC-LM3s. CDC25B was also involved in the formation of the tumor microenvironment (TME) and immune processes in HCC, and the high expression of CDC25B made patients less sensitive to some drugs. CDC25B can be used as a biomarker and immunotherapeutic target for poor prognosis and partial drug sensitivity in HCC, providing new ideas for HCC treatment.
Collapse
Affiliation(s)
- Zixiang Huang
- Department of GastroenterologyThe Second Affiliated Hospital of Jiangxi Medical CollegeNanchang University, Nanchang, China
| | - Liangzhi Xu
- Department of Hepatobiliary SurgeryEzhou Central Hospital, Ezhou, Hubei, China
| | - Zhengqiang Wu
- Department of GastroenterologyThe Second Affiliated Hospital of Jiangxi Medical CollegeNanchang University, Nanchang, China
| | - Xiaofeng Xiong
- Department of GastroenterologyThe Second Affiliated Hospital of Jiangxi Medical CollegeNanchang University, Nanchang, China
| | - Linfei Luo
- Department of GastroenterologyThe Second Affiliated Hospital of Jiangxi Medical CollegeNanchang University, Nanchang, China
| | - Zhili Wen
- Department of GastroenterologyThe Second Affiliated Hospital of Jiangxi Medical CollegeNanchang University, Nanchang, China
| |
Collapse
|
7
|
Lu J, Yu M, Li J. PKC-δ Promotes IL-1β-Induced Apoptosis of Rat Chondrocytes and Via Activating JNK and P38 MAPK Pathways. Cartilage 2024; 15:315-327. [PMID: 37491820 PMCID: PMC11418514 DOI: 10.1177/19476035231181446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 03/17/2023] [Accepted: 05/26/2023] [Indexed: 07/27/2023] Open
Abstract
OBJECTIVE Protein kinase C-delta (PKC-δ) is involved in apoptosis. This study aimed to establish whether PKC-δ can further promote IL-1β-induced chondrocyte apoptosis by mediating the phosphorylation of the JNK and p38 mitogen-activated protein kinase (MAPK) signaling pathways In osteoarthritis (OA). METHODS We employed chondrocyte staining to determine the extent of cartilage degeneration. PKC-δ and p38 signal expressions were used in the immunohistochemical (IHC) test and apoptosis was assayed at the TUNEL test in human osteoarthritic and controls. We stimulated rat cartilage cells using IL-1β (10 ng/ml)/rottlerin (10 μM) or lentivirus. To determine the apoptosis rate, we employed flow cytometry. The mRNA of both BCL2-related X (BAX) and cysteine aspartate protease 3 (caspase-3) could be measured via qRT-PCR. Western blot measured the protein levels of BAX, caspase-3, PKC-δ, p-JNK/JNK and p-p38/p38. RESULTS The positive rate of PKC-δ and the apoptotic rate of chondrocytes in OA were higher than controls. The manifestation of PKC-δ was positively related to the degree of cartilage degeneration, p38 protein expression, and apoptosis rate. IL-1β exposure upregulated PKC-δ expression in chondrocytes in a dose-dependent manner. Decreasing PKC-δ expression and its phosphorylation in OA can inhibit MAPK signaling pathway activation (phosphorylation) by downregulating JNK and p38 protein phosphorylation and expression. This inhibition decreases caspase-3 and BAX levels, consequently lowering the apoptosis rate in chondrocytes. CONCLUSION PKC-δ activation by IL-1β in OA promotes chondrocyte apoptosis via activation of the JNK and p38 MAPK signal pathways, thereby promoting the OA progression.
Collapse
Affiliation(s)
- Jinfeng Lu
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Miao Yu
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jia Li
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
8
|
Zhan Y, Wang R, Huang C, Xu X, Xiao X, Wu L, Wei J, Long T, Gao C. Digitoxin inhibits ICC cell properties via the NF‑κB/ST6GAL1 signaling pathway. Oncol Rep 2024; 52:103. [PMID: 38940341 PMCID: PMC11229393 DOI: 10.3892/or.2024.8762] [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: 03/12/2024] [Accepted: 05/30/2024] [Indexed: 06/29/2024] Open
Abstract
Intrahepatic cholangiocarcinoma (ICC) is a type of liver cancer associated with poor prognosis and increased mortality; the limited treatment strategy highlights the urgent need for investigation. Traditional Chinese Medicine (TCM), used alone or in combination with other treatments, can enhance therapeutic efficacy, improve life quality of patients and extend overall survival. In total, two rounds of screening of a TCM library of 2,538 active compounds were conducted using a Cell Counting Kit‑8 assay and ICC cell lines. Cell proliferation and migration abilities were assessed through colony formation, 5‑ethynyl‑2'‑deoxyuridine, would healing and Transwell assays. The impact of digitoxin (DT) on signaling pathways was initially investigated using RNA sequencing and further validated using reverse transcription‑quantitative PCR, western blotting, lectin blotting and flow cytometry. ICC cells stably overexpressing ST6 β‑galactoside α‑2,6‑sialyltransferase 1 (ST6GAL1) were generated through lentiviral transfection. It was shown that DT emerged as a highly effective anti‑ICC candidate from two rounds high‑throughput library screening. DT could inhibit the proliferation and migration of ICC cells by suppressing NF‑κB activation and reducing nuclear phosphorylated‑NF‑κB levels, along with diminishing ST6GAL1 mRNA and protein expression. The aforementioned biological effects and signal pathways of DT could be counteracted by overexpressing ST6GAL1 in ICC cells. In conclusion, DT suppressed ICC cell proliferation and migration by targeting the NF‑κB/ST6GAL1 signaling axis. The findings of the present study indicated the promising therapeutic effects of DT in managing ICC, offering new avenues for treatment strategies.
Collapse
Affiliation(s)
- Yueping Zhan
- Clinical Laboratory Medicine Center, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, P.R. China
| | - Rong Wang
- Clinical Laboratory Medicine Center, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, P.R. China
| | - Chenjun Huang
- Clinical Laboratory Medicine Center, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, P.R. China
| | - Xuewen Xu
- Clinical Laboratory Medicine Center, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, P.R. China
| | - Xiao Xiao
- Clinical Laboratory Medicine Center, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, P.R. China
| | - Linlin Wu
- Clinical Laboratory Medicine Center, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, P.R. China
| | - Jiao Wei
- Clinical Laboratory Medicine Center, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, P.R. China
| | - Tian Long
- Clinical Laboratory Medicine Center, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, P.R. China
| | - Chunfang Gao
- Clinical Laboratory Medicine Center, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, P.R. China
| |
Collapse
|
9
|
Dorry S, Perla S, Bennett AM. MAPK Phosphatase-5 is required for TGF-β signaling through a JNK-dependent pathway. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.27.600976. [PMID: 38979264 PMCID: PMC11230413 DOI: 10.1101/2024.06.27.600976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Mitogen-activated protein kinase (MAPK) phosphatases (MKPs) constitute members of the dual-specificity family of protein phosphatases that dephosphorylate the MAPKs. MKP-5 dephosphorylates the stress-responsive MAPKs, p38 MAPK and JNK, and has been shown to promote tissue fibrosis. Here, we provide insight into how MKP-5 regulates the transforming growth factor-β (TGF-β) pathway, a well-established driver of fibrosis. We show that MKP-5-deficient fibroblasts in response to TGF-β are impaired in SMAD2 phosphorylation at canonical and non-canonical sites, nuclear translocation, and transcriptional activation of fibrogenic genes. Consistent with this, pharmacological inhibition of MKP-5 is sufficient to block TGF-β signaling, and that this regulation occurs through a JNK-dependent pathway. By utilizing RNA sequencing and transcriptomic analysis, we identify TGF-β signaling activators regulated by MKP-5 in a JNK-dependent manner, providing mechanistic insight into how MKP-5 promotes TGF-β signaling. This study elucidates a novel mechanism whereby MKP-5-mediated JNK inactivation is required for TGF-β signaling and provides insight into the role of MKP-5 in fibrosis.
Collapse
Affiliation(s)
- Sam Dorry
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Sravan Perla
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Anton M. Bennett
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut, USA
- Yale Center for Molecular and Systems Metabolism, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
10
|
He TQ, Wang Z, Li CY, Zhao YW, Tong XY, Liu JH, Ouyang JM. Sulfated Laminarin Polysaccharides Reduce the Adhesion of Nano-COM Crystals to Renal Epithelial Cells by Inhibiting Oxidative and Endoplasmic Reticulum Stress. Pharmaceuticals (Basel) 2024; 17:805. [PMID: 38931471 PMCID: PMC11206474 DOI: 10.3390/ph17060805] [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: 04/12/2024] [Revised: 05/19/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
Purpose: Adhesion between calcium oxalate crystals and renal tubular epithelial cells is a vital cause of renal stone formation; however, the drugs that inhibit crystal adhesion and the mechanism of inhibition have yet to be explored. Methods: The cell injury model was constructed using nano-COM crystals, and changes in oxidative stress levels, endoplasmic reticulum (ER) stress levels, downstream p38 MAPK protein expression, apoptosis, adhesion protein osteopontin expression, and cell-crystal adhesion were examined in the presence of Laminarin polysaccharide (DLP) and sulfated DLP (SDLP) under protected and unprotected conditions. Results: Both DLP and SDLP inhibited nano-COM damage to human kidney proximal tubular epithelial cell (HK-2), increased cell viability, decreased ROS levels, reduced the opening of mitochondrial membrane permeability transition pore, markedly reduced ER Ca2+ ion concentration and adhesion molecule OPN expression, down-regulated the expression of ER stress signature proteins including CHOP, Caspase 12, and p38 MAPK, and decreased the apoptosis rate of cells. SDLP has a better protective effect on cells than DLP. Conclusions: SDLP protects HK-2 cells from nano-COM crystal-induced apoptosis by reducing oxidative and ER stress levels and their downstream factors, thereby reducing crystal-cell adhesion interactions and the risks of kidney stone formation.
Collapse
Affiliation(s)
- Tian-Qu He
- Department of Urology, The Affiliated Children’s Hospital of Xiangya School of Medicine, Central South University (Hunan Children’s Hospital), Changsha 410007, China
| | - Zhi Wang
- Department of Urology, The Affiliated Children’s Hospital of Xiangya School of Medicine, Central South University (Hunan Children’s Hospital), Changsha 410007, China
| | - Chuang-Ye Li
- Department of Urology, The Affiliated Children’s Hospital of Xiangya School of Medicine, Central South University (Hunan Children’s Hospital), Changsha 410007, China
| | - Yao-Wang Zhao
- Department of Urology, The Affiliated Children’s Hospital of Xiangya School of Medicine, Central South University (Hunan Children’s Hospital), Changsha 410007, China
| | - Xin-Yi Tong
- Institute of Biomineralization and Lithiasis Research, College of Chemistry and Materials Science, Jinan University, Guangzhou 510632, China
| | - Jing-Hong Liu
- Institute of Biomineralization and Lithiasis Research, College of Chemistry and Materials Science, Jinan University, Guangzhou 510632, China
| | - Jian-Ming Ouyang
- Institute of Biomineralization and Lithiasis Research, College of Chemistry and Materials Science, Jinan University, Guangzhou 510632, China
| |
Collapse
|
11
|
Alzu'bi A, Abu-El-Rub E, Almahasneh F, Tahat L, Athamneh RY, Khasawneh R, Alzoubi H, Ghorab DS, Almazari R, Zoubi MSA, Al-Zoubi RM. Delineating the molecular mechanisms of hippocampal neurotoxicity induced by chronic administration of synthetic cannabinoid AB-FUBINACA in mice. Neurotoxicology 2024; 103:50-59. [PMID: 38823587 DOI: 10.1016/j.neuro.2024.05.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 05/28/2024] [Accepted: 05/28/2024] [Indexed: 06/03/2024]
Abstract
Chronic use of synthetic cannabinoids (SCs) has been associated with cognitive and behavioural deficits and an increased risk of neuropsychiatric disorders. The underlying molecular and cellular mechanisms of the neurotoxic effects of long-term use of SCs have not been well investigated in the literature. Herein, we evaluated the in vivo effects of chronic administration of AB-FUBINACA on the hippocampus in mice. Our results revealed that the administration of AB-FUBINACA induced a significant impairment in recognition memory associated with histopathological changes in the hippocampus. These findings were found to be correlated with increased level of oxidative stress, neuroinflammation, and apoptosis markers, and reduced expression of brain-derived neurotrophic factor (BDNF), which plays an essential role in modulating synaptic plasticity integral for promoting learning and memory in the hippocampus. Additionally, we showed that AB-FUBINACA significantly decreased the expression of NR1, an important functional subunit of glutamate/NMDA receptors and closely implicated in the development of toxic psychosis. These findings shed light on the long-term neurotoxic effects of SCs on hippocampus and the underlying mechanisms of these effects. This study provided new targets for possible medical interventions to improve the treatment guidelines for SCs addiction.
Collapse
Affiliation(s)
- Ayman Alzu'bi
- Department of Basic Medical Sciences, Faculty of Medicine, Yarmouk University, Irbid 211-63, Jordan.
| | - Ejlal Abu-El-Rub
- Department of Basic Medical Sciences, Faculty of Medicine, Yarmouk University, Irbid 211-63, Jordan
| | - Fatimah Almahasneh
- Department of Basic Medical Sciences, Faculty of Medicine, Yarmouk University, Irbid 211-63, Jordan
| | - Lena Tahat
- Department of Biological Sciences, Faculty of Science, Yarmouk University, Irbid 211-63, Jordan
| | - Rabaa Y Athamneh
- Department of Medical Laboratory Sciences, Faculty of Allied Science, Zarqa University, Zarqa 13110, Jordan
| | - Ramada Khasawneh
- Department of Basic Medical Sciences, Faculty of Medicine, Yarmouk University, Irbid 211-63, Jordan
| | - Hiba Alzoubi
- Department of Basic Medical Sciences, Faculty of Medicine, Yarmouk University, Irbid 211-63, Jordan
| | - Doaa S Ghorab
- Department of Basic Medical Sciences, Faculty of Medicine, Yarmouk University, Irbid 211-63, Jordan; Pathology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Rawan Almazari
- Department of Basic Medical Sciences, Faculty of Medicine, Yarmouk University, Irbid 211-63, Jordan
| | - Mazhar Salim Al Zoubi
- Department of Basic Medical Sciences, Faculty of Medicine, Yarmouk University, Irbid 211-63, Jordan
| | - Raed M Al-Zoubi
- Surgical Research Section, Department of Surgery, Hamad Medical Corporation, Doha, Qatar; Department of Biomedical Sciences, QU-Health, College of Health Sciences, Qatar University, Doha 2713, Qatar; Department of Chemistry, Jordan University of Science and Technology, P.O.Box 3030, Irbid 22110, Jordan.
| |
Collapse
|
12
|
Abstract
Eryptosis is a regulated cell death (RCD) of mature erythrocytes initially described as a counterpart of apoptosis for enucleated cells. However, over the recent years, a growing number of studies have emphasized certain differences between both cell death modalities. In this review paper, we underline the hallmarks of eryptosis and apoptosis and highlight resemblances and dissimilarities between both RCDs. We summarize and critically discuss differences in the impact of caspase-3, Ca2+ signaling, ROS signaling pathways, opposing roles of casein kinase 1α, protein kinase C, Janus kinase 3, cyclin-dependent kinase 4, and AMP-activated protein kinase to highlight a certain degree of divergence between apoptosis and eryptosis. This review emphasizes the crucial importance of further studies that focus on deepening our knowledge of cell death machinery and identifying novel differences between cell death of nucleated and enucleated cells. This might provide evidence that erythrocytes can be defined as viable entities capable of programmed cell destruction. Additionally, the revealed cell type-specific patterns in cell death can facilitate the development of cell death-modulating therapeutic agents.
Collapse
Affiliation(s)
- Anton Tkachenko
- 1st Faculty of Medicine, BIOCEV, Charles University, Průmyslová 595, 25250, Vestec, Czech Republic.
| |
Collapse
|
13
|
Liu Y, Tang Q, Tao Q, Dong H, Shi Z, Zhou L. Low-frequency magnetic field therapy for glioblastoma: Current advances, mechanisms, challenges and future perspectives. J Adv Res 2024:S2090-1232(24)00125-5. [PMID: 38565404 DOI: 10.1016/j.jare.2024.03.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 03/10/2024] [Accepted: 03/29/2024] [Indexed: 04/04/2024] Open
Abstract
BACKGROUND Glioblastoma (GBM) is the most common malignant tumour of the central nervous system. Despite recent advances in multimodal GBM therapy incorporating surgery, radiotherapy, systemic therapy (chemotherapy, targeted therapy), and supportive care, the overall survival (OS) remains poor, and long-term survival is rare. Currently, the primary obstacles hindering the effectiveness of GBM treatment are still the blood-brain barrier and tumor heterogeneity. In light of its substantial advantages over conventional therapies, such as strong penetrative ability and minimal side effects, low-frequency magnetic fields (LF-MFs) therapy has gradually caught the attention of scientists. AIM OF REVIEW In this review, we shed the light on the current status of applying LF-MFs in the treatment of GBM. We specifically emphasize our current understanding of the mechanisms by which LF-MFs mediate anticancer effects and the challenges faced by LF-MFs in treating GBM cells. Furthermore, we discuss the prospective applications of magnetic field therapy in the future treatment of GBM. Key scientific concepts of review: The review explores the current progress on the use of LF-MFs in the treatment of GBM with a special focus on the potential underlying mechanisms of LF-MFs in anticancer effects. Additionally, we also discussed the complex magnetic field features and biological characteristics related to magnetic bioeffects. Finally, we proposed a promising magnetic field treatment strategy for future applications in GBM therapy.
Collapse
Affiliation(s)
- Yinlong Liu
- Department of Neurosurgery, Huashan Hospital, Fudan University, China
| | - Qisheng Tang
- Department of Neurosurgery, Huashan Hospital, Fudan University, China; National Center for Neurological Disorders, China; Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, China; Neurosurgical Institute of Fudan University, Shanghai, China; Shanghai Clinical Medical Center of Neurosurgery, China
| | - Quan Tao
- Shanghai Institute of Microsystem and Information Technology, China
| | - Hui Dong
- Shanghai Institute of Microsystem and Information Technology, China
| | - Zhifeng Shi
- Department of Neurosurgery, Huashan Hospital, Fudan University, China; National Center for Neurological Disorders, China; Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, China; Neurosurgical Institute of Fudan University, Shanghai, China; Shanghai Clinical Medical Center of Neurosurgery, China.
| | - Liangfu Zhou
- Department of Neurosurgery, Huashan Hospital, Fudan University, China; National Center for Neurological Disorders, China; Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, China; Neurosurgical Institute of Fudan University, Shanghai, China; Shanghai Clinical Medical Center of Neurosurgery, China.
| |
Collapse
|
14
|
Abooshahab R, Al-Salami H, Dass CR. Synergy between PEDF and Doxorubicin in Breast Cancer Cells: Effects on Metastatic and Metabolic Pathways. Int J Mol Sci 2024; 25:2755. [PMID: 38474001 DOI: 10.3390/ijms25052755] [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: 02/01/2024] [Revised: 02/19/2024] [Accepted: 02/24/2024] [Indexed: 03/14/2024] Open
Abstract
Pigment epithelium-derived factor (PEDF), a serine protease inhibitor (Serpin) family member, shows promise in inhibiting tumour growth. In our study, we explored the effects of PEDF on the efficacy of the frontline chemotherapy agent doxorubicin (Dox) in BC cells. We found that Dox+PEDF treatment significantly reduced glucose uptake in MDA-MB-231 cells compared to the control (p = 0.0005), PEDF (p = 0.0137), and Dox (p = 0.0171) alone but paradoxically increased it in MCF-7 cells. Our findings further revealed that PEDF, Dox, and Dox+PEDF substantially hindered tumour cell migration from tumour spheroids, with Dox+PEDF showing the most significant impact (p < 0.0001). We also observed notable decreases in the expression of metastatic markers (uPAR, uPA, CXCR4, MT1-MMP, TNF-α) across all treatment groups (p < 0.0001) in both cell lines. When it comes to metabolic pathways, PEDF increased phosphorylated IRS-1 (p-IRS1) levels in MDA-MB-231 and MCF-7 (p < 0.0001), while Dox decreased it, and the combination led to an increase. In MDA-MB-231 cells, treatment with PEDF, Dox, and the combination led to a notable decrease in both phosphorylated AKT (p-AKT) and total AKT levels. In MCF-7, while PEDF, Dox, and their combination led to a reduction in p-AKT, total levels of AKT increased in the presence of Dox and Dox+PEDF. Combining PEDF with Dox enhances the targeting of metastatic and metabolic pathways in breast cancer cell lines. This synergy, marked by PEDF's increasing roles in cancer control, may pave the way for more effective cancer treatments.
Collapse
Affiliation(s)
- Raziyeh Abooshahab
- Curtin Medical School, Curtin University, Bentley 6102, Australia
- Curtin Health Innovation Research Institute, Bentley 6102, Australia
| | - Hani Al-Salami
- Curtin Medical School, Curtin University, Bentley 6102, Australia
- Biotechnology and Drug Development Research Laboratory, Curtin Health Innovation Research Institute, Bentley 6102, Australia
| | - Crispin R Dass
- Curtin Medical School, Curtin University, Bentley 6102, Australia
- Curtin Health Innovation Research Institute, Bentley 6102, Australia
| |
Collapse
|
15
|
Wang HH, Fan SQ, Zhan YT, Peng SP, Wang WY. Suppression of the SLC7A11/glutathione axis causes ferroptosis and apoptosis and alters the mitogen-activated protein kinase pathway in nasopharyngeal carcinoma. Int J Biol Macromol 2024; 254:127976. [PMID: 37951442 DOI: 10.1016/j.ijbiomac.2023.127976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 07/24/2023] [Accepted: 11/06/2023] [Indexed: 11/14/2023]
Abstract
SLC7A11 is a unit of the glutamate cystine antiporter Xc- system. It functions to import cystine for glutathione biosynthesis and maintains the redox balance in cells. Sorafenib inhibits the transporter activity of SLC7A11. The use of sorafenib has been approved in the treatment of multiple cancers. However, at present, our understanding of the mechanism of SLC7A11 and sorafenib in nasopharyngeal carcinoma (NPC) remains limited. We found that the expression of SLC7A11 was upregulated in NPC. A high SLC7A11 expression was associated with poor prognosis, metastasis, and an advanced T stage, which can be used as an independent prognostic indicator of NPC. In vitro, we observed that NPC cells relied on cystine for survival. Targeting SLC7A11 resulted in glutathione biosynthesis limitation, intracellular reactive oxygen species accumulation, lipid peroxides, ferroptosis, and apoptosis. Meanwhile, it altered mitogen activated protein kinase pathway, including p38 activation but ERK inhibition in NPC. This limited the proliferation of NPC cells. Sorafenib inhibited the proliferation and induced the death of NPC cells in vivo. In conclusion, SLC7A11 plays an important role in the occurrence and progression of NPC and may be a novel target for NPC treatment.
Collapse
Affiliation(s)
- Hai-Hua Wang
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Song-Qing Fan
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Yu-Ting Zhan
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Shu-Ping Peng
- Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
| | - Wei-Yuan Wang
- Department of Pathology, The Xiangya Hospital, Central South University, Changsha, Hunan 410008, China.
| |
Collapse
|
16
|
Ma J, Li J, Wei S, Ge Q, Wu J, Xue L, Qi Y, Xu S, Jin H, Gao C, Lin J. Delivery of dental pulp stem cells by an injectable ROS-responsive hydrogel promotes temporomandibular joint cartilage repair via enhancing anti-apoptosis and regulating microenvironment. J Tissue Eng 2024; 15:20417314241260436. [PMID: 38911101 PMCID: PMC11193934 DOI: 10.1177/20417314241260436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 05/23/2024] [Indexed: 06/25/2024] Open
Abstract
Temporomandibular joint (TMJ) cartilage repair poses a considerable clinical challenge, and tissue engineering has emerged as a promising solution. In this study, we developed an injectable reactive oxygen species (ROS)-responsive multifunctional hydrogel (RDGel) to encapsulate dental pulp stem cells (DPSCs/RDGel in short) for the targeted repair of condylar cartilage defect. The DPSCs/RDGel composite exhibited a synergistic effect in the elimination of TMJ OA (osteoarthritis) inflammation via the interaction between the hydrogel component and the DPSCs. We first demonstrated the applicability and biocompatibility of RDGel. RDGel encapsulation could enhance the anti-apoptotic ability of DPSCs by inhibiting P38/P53 mitochondrial apoptotic signal in vitro. We also proved that the utilization of DPSCs/RDGel composite effectively enhanced the expression of TMJOA cartilage matrix and promoted subchondral bone structure in vivo. Subsequently, we observed the synergistic improvement of DPSCs/RDGel composite on the oxidative stress microenvironment of TMJOA and its regulation and promotion of M2 polarization, thereby confirmed that M2 macrophages further promoted the condylar cartilage repair of DPSCs. This is the first time application of DPSCs/RDGel composite for the targeted repair of TMJOA condylar cartilage defects, presenting a novel and promising avenue for cell-based therapy.
Collapse
Affiliation(s)
- Jinjin Ma
- Department of Stomatology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Juan Li
- Department of Stomatology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shibo Wei
- Innovation Center for Smart Medical Technologies & Devices, Binjiang Institute of Zhejiang University, Hangzhou, China
| | - Qinwen Ge
- Institute of Orthopaedics and Traumatology of Zhejiang Province, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Jie Wu
- School of Stomatology, Zhejiang Chinese Medical University, Hangzhou, China
| | - Leilei Xue
- Department of Stomatology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yezi Qi
- Department of Stomatology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Siyi Xu
- Department of Stomatology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hongting Jin
- Institute of Orthopaedics and Traumatology of Zhejiang Province, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Changyou Gao
- Innovation Center for Smart Medical Technologies & Devices, Binjiang Institute of Zhejiang University, Hangzhou, China
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China
| | - Jun Lin
- Department of Stomatology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
17
|
Afsar B, Afsar RE. Salt Behind the Scenes of Systemic Lupus Erythematosus and Rheumatoid Arthritis. Curr Nutr Rep 2023; 12:830-844. [PMID: 37980312 DOI: 10.1007/s13668-023-00509-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/09/2023] [Indexed: 11/20/2023]
Abstract
PURPOSE OF REVIEW Sodium is vital for human health. High salt intake is a global health problem and is associated with cardiovascular morbidity and mortality. Recent evidence suggests that both innate and adaptive immune systems are affected by sodium. In general, excess salt intake drives immune cells toward a pro-inflammatory phenotype. The incidence of autoimmune diseases, including systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA), is steadily increasing. As excess salt induces a pro-inflammatory state, increased salt intake may have impacts on autoimmune diseases. The relationship between salt intake and autoimmune diseases is most widely studied in patients with SLE or RA. This review aimed to summarize the relationship between salt intake and SLE and RA. RECENT FINDINGS Most, but not all, of these studies showed that high salt intake might promote SLE by M1 macrophage shift, increase in Th17/Treg cell ratio, activation of dendritic and follicular helper T cells, and increased secretion of pro-inflammatory cytokines. In RA, apart from driving immune cells toward a pro-inflammatory state, high salt intake also influences cellular signaling pathways, including receptor activator of nuclear factor κB ligand (RANKL), Rho GTPases, and MAPK (mitogen-activated protein kinase). There is now sufficient evidence that excess salt intake may be related to the development and progression of SLE and RA, although there are still knowledge gaps. More studies are warranted to further highlight the relationship between excess salt intake, SLE, and RA. Salt intake may affect cell types and pro-inflammatory cytokines and signaling pathways associated with the development and progression of systemic lupus erythematosus and rheumatoid arthritis. Bcl-6 B-cell lymphoma, 6 Erk extracellular signal-regulated kinases, IFN-γ interferon-gamma, JNK c-Jun N-terminal kinase, IL-4 interleukin 4, IL-6 interleukin 6, MAPK mitogen-activated protein kinase, STAT signal transducer and activator of transcription, Tnf-α tumor necrosis factor, Treg T regulatory cell.
Collapse
Affiliation(s)
- Baris Afsar
- Department of Nephrology, School of Medicine, Suleyman Demirel University, Isparta, 32260, Turkey.
| | - Rengin Elsurer Afsar
- Department of Nephrology, School of Medicine, Suleyman Demirel University, Isparta, 32260, Turkey
| |
Collapse
|
18
|
Li X, Lao R, Lei J, Chen Y, Zhou Q, Wang T, Tong Y. Natural Products for Acetaminophen-Induced Acute Liver Injury: A Review. Molecules 2023; 28:7901. [PMID: 38067630 PMCID: PMC10708418 DOI: 10.3390/molecules28237901] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 11/07/2023] [Accepted: 11/23/2023] [Indexed: 12/18/2023] Open
Abstract
The liver plays a vital role in metabolism, synthesis, and detoxification, but it is susceptible to damage from various factors such as viral infections, drug reactions, excessive alcohol consumption, and autoimmune diseases. This susceptibility is particularly problematic for patients requiring medication, as drug-induced liver injury often leads to underestimation, misdiagnosis, and difficulties in treatment. Acetaminophen (APAP) is a widely used and safe drug in therapeutic doses but can cause liver toxicity when taken in excessive amounts. This study aimed to investigate the hepatotoxicity of APAP and explore potential treatment strategies using a mouse model of APAP-induced liver injury. The study involved the evaluation of various natural products for their therapeutic potential. The findings revealed that natural products demonstrated promising hepatoprotective effects, potentially alleviating liver damage and improving liver function through various mechanisms such as oxidative stress and inflammation, which cause changes in signaling pathways. These results underscore the importance of exploring novel treatment options for drug-induced liver injury, suggesting that further research in this area could lead to the development of effective preventive and therapeutic interventions, ultimately benefiting patients with liver injury caused by medicine.
Collapse
Affiliation(s)
- Xiaoyangzi Li
- School of Medicine, Taizhou University, Taizhou 318000, China; (X.L.); (R.L.); (J.L.)
| | - Ruyang Lao
- School of Medicine, Taizhou University, Taizhou 318000, China; (X.L.); (R.L.); (J.L.)
| | - Jiawei Lei
- School of Medicine, Taizhou University, Taizhou 318000, China; (X.L.); (R.L.); (J.L.)
| | - Yuting Chen
- College of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian 116000, China;
| | - Qi Zhou
- School of Pharmacy, Taizhou University, Taizhou 318000, China;
| | - Ting Wang
- School of Medicine, Taizhou University, Taizhou 318000, China; (X.L.); (R.L.); (J.L.)
| | - Yingpeng Tong
- School of Pharmacy, Taizhou University, Taizhou 318000, China;
| |
Collapse
|
19
|
Antunes J, Sobral P, Martins M, Branco V. Nanoplastics activate a TLR4/p38-mediated pro-inflammatory response in human intestinal and mouse microglia cells. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2023; 104:104298. [PMID: 37865352 DOI: 10.1016/j.etap.2023.104298] [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: 04/10/2023] [Revised: 10/05/2023] [Accepted: 10/17/2023] [Indexed: 10/23/2023]
Abstract
The crescent presence of nanoplastics in the environment raises concerns regarding their potential impact on health. This study exposed human colon adenocarcinoma cells (HT29) and microglia cells (N9) to nanoplastics (25 nm, 50 nm, and 100 nm Polystyrene) to investigate their inflammatory responses, which are vital for body's defence. Although cytotoxicity remained generally low, HT29 cells exhibited a notable upregulation of p50 and p38 expression, concomitant with elevated TLR4 expression, in contrast with N9 cells that showed a less pronounced upregulation of these proteins. Additionally, nanoplastic exposure increased IL-1ß levels, partially attenuated by pre-exposure to TLR4 or p38 inhibitors. Intriguingly, N9 cells exposed to nanoplastics exhibited substantial increases in iNOS mRNA. This effect was entirely prevented by pre-exposure to TLR4 or p38 inhibitors, while TNF-α mRNA levels remained relatively stable. These findings underscore the potential of nanoplastics to activate inflammatory pathways, with response kinetics varying depending on the cell type.
Collapse
Affiliation(s)
- Joana Antunes
- MARE, Marine and Environmental Sciences Centre & ARNET, Aquatic Research Network Associated Laboratory, Department of Science and Environmental Engineering, NOVA School of Science and Technology (FCT NOVA), University NOVA of Lisbon, Caparica 2829-516, Portugal.
| | - Paula Sobral
- MARE, Marine and Environmental Sciences Centre & ARNET, Aquatic Research Network Associated Laboratory, Department of Science and Environmental Engineering, NOVA School of Science and Technology (FCT NOVA), University NOVA of Lisbon, Caparica 2829-516, Portugal
| | - Marta Martins
- MARE, Marine and Environmental Sciences Centre & ARNET, Aquatic Research Network Associated Laboratory, Department of Science and Environmental Engineering, NOVA School of Science and Technology (FCT NOVA), University NOVA of Lisbon, Caparica 2829-516, Portugal.
| | - Vasco Branco
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; Egas Moniz Center for Interdisciplinary Research (CiiEM), Egas Moniz School of Health & Science, Quinta da Granja, Monte de Caparica, 2829-511 Caparica, Portugal.
| |
Collapse
|
20
|
Khalifa A, Guijarro A, Ravera S, Bertola N, Adorni MP, Papotti B, Raffaghello L, Benelli R, Becherini P, Namatalla A, Verzola D, Reverberi D, Monacelli F, Cea M, Pisciotta L, Bernini F, Caffa I, Nencioni A. Cyclic fasting bolsters cholesterol biosynthesis inhibitors' anticancer activity. Nat Commun 2023; 14:6951. [PMID: 37907500 PMCID: PMC10618279 DOI: 10.1038/s41467-023-42652-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 10/17/2023] [Indexed: 11/02/2023] Open
Abstract
Identifying oncological applications for drugs that are already approved for other medical indications is considered a possible solution for the increasing costs of cancer treatment. Under the hypothesis that nutritional stress through fasting might enhance the antitumour properties of at least some non-oncological agents, by screening drug libraries, we find that cholesterol biosynthesis inhibitors (CBIs), including simvastatin, have increased activity against cancers of different histology under fasting conditions. We show fasting's ability to increase CBIs' antitumour effects to depend on the reduction in circulating insulin, insulin-like growth factor-1 and leptin, which blunts the expression of enzymes from the cholesterol biosynthesis pathway and enhances cholesterol efflux from cancer cells. Ultimately, low cholesterol levels through combined fasting and CBIs reduce AKT and STAT3 activity, oxidative phosphorylation and energy stores in the tumour. Our results support further studies of CBIs in combination with fasting-based dietary regimens in cancer treatment and highlight the value of fasting for drug repurposing in oncology.
Collapse
Affiliation(s)
- Amr Khalifa
- Department of Internal Medicine and Medical Specialties, University of Genoa, Viale Benedetto XV 6, 16132, Genoa, Italy
- Ospedale Policlinico San Martino IRCCS, Largo Rosanna Benzi 10, 16132, Genoa, Italy
| | - Ana Guijarro
- Department of Internal Medicine and Medical Specialties, University of Genoa, Viale Benedetto XV 6, 16132, Genoa, Italy
- Ospedale Policlinico San Martino IRCCS, Largo Rosanna Benzi 10, 16132, Genoa, Italy
| | - Silvia Ravera
- Department of Experimental Medicine, University of Genoa, Via Leon Battista Alberti 2, 16132, Genoa, Italy
| | - Nadia Bertola
- Department of Experimental Medicine, University of Genoa, Via Leon Battista Alberti 2, 16132, Genoa, Italy
| | - Maria Pia Adorni
- Department of Medicine and Surgery, University of Parma, 43125, Parma, Italy
| | - Bianca Papotti
- Department of Food and Drug, University of Parma, 43124, Parma, Italy
| | - Lizzia Raffaghello
- Center of Translational and Experimental Myology, IRCCS Istituto Giannina Gaslini, 16147, Genoa, Italy
| | - Roberto Benelli
- Ospedale Policlinico San Martino IRCCS, Largo Rosanna Benzi 10, 16132, Genoa, Italy
| | - Pamela Becherini
- Department of Internal Medicine and Medical Specialties, University of Genoa, Viale Benedetto XV 6, 16132, Genoa, Italy
| | - Asmaa Namatalla
- Department of Internal Medicine and Medical Specialties, University of Genoa, Viale Benedetto XV 6, 16132, Genoa, Italy
| | - Daniela Verzola
- Department of Internal Medicine and Medical Specialties, University of Genoa, Viale Benedetto XV 6, 16132, Genoa, Italy
| | - Daniele Reverberi
- Ospedale Policlinico San Martino IRCCS, Largo Rosanna Benzi 10, 16132, Genoa, Italy
| | - Fiammetta Monacelli
- Department of Internal Medicine and Medical Specialties, University of Genoa, Viale Benedetto XV 6, 16132, Genoa, Italy
- Ospedale Policlinico San Martino IRCCS, Largo Rosanna Benzi 10, 16132, Genoa, Italy
| | - Michele Cea
- Department of Internal Medicine and Medical Specialties, University of Genoa, Viale Benedetto XV 6, 16132, Genoa, Italy
- Ospedale Policlinico San Martino IRCCS, Largo Rosanna Benzi 10, 16132, Genoa, Italy
| | - Livia Pisciotta
- Department of Internal Medicine and Medical Specialties, University of Genoa, Viale Benedetto XV 6, 16132, Genoa, Italy
- Ospedale Policlinico San Martino IRCCS, Largo Rosanna Benzi 10, 16132, Genoa, Italy
| | - Franco Bernini
- Department of Food and Drug, University of Parma, 43124, Parma, Italy
| | - Irene Caffa
- Department of Internal Medicine and Medical Specialties, University of Genoa, Viale Benedetto XV 6, 16132, Genoa, Italy.
- Ospedale Policlinico San Martino IRCCS, Largo Rosanna Benzi 10, 16132, Genoa, Italy.
| | - Alessio Nencioni
- Department of Internal Medicine and Medical Specialties, University of Genoa, Viale Benedetto XV 6, 16132, Genoa, Italy.
- Ospedale Policlinico San Martino IRCCS, Largo Rosanna Benzi 10, 16132, Genoa, Italy.
| |
Collapse
|
21
|
Kesarwani M, Kincaid Z, Azhar M, Menke J, Schwieterman J, Ansari S, Reaves A, Deininger ME, Levine R, Grimes HL, Azam M. MAPK-negative feedback regulation confers dependence to JAK2 V617F signaling. Leukemia 2023; 37:1686-1697. [PMID: 37430058 PMCID: PMC10976185 DOI: 10.1038/s41375-023-01959-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 06/11/2023] [Accepted: 06/27/2023] [Indexed: 07/12/2023]
Abstract
Despite significant advances in developing selective JAK2 inhibitors, JAK2 kinase inhibitor (TKI) therapy is ineffective in suppressing the disease. Reactivation of compensatory MEK-ERK and PI3K survival pathways sustained by inflammatory cytokine signaling causes treatment failure. Concomitant inhibition of MAPK pathway and JAK2 signaling showed improved in vivo efficacy compared to JAK2 inhibition alone but lacked clonal selectivity. We hypothesized that cytokine signaling in JAK2V617F induced MPNs increases the apoptotic threshold that causes TKI persistence or refractoriness. Here, we show that JAK2V617F and cytokine signaling converge to induce MAPK negative regulator, DUSP1. Enhanced DUSP1 expression blocks p38 mediated p53 stabilization. Deletion of Dusp1 increases p53 levels in the context of JAK2V617F signaling that causes synthetic lethality to Jak2V617F expressing cells. However, inhibition of Dusp1 by a small molecule inhibitor (BCI) failed to impart Jak2V617F clonal selectivity due to pErk1/2 rebound caused by off-target inhibition of Dusp6. Ectopic expression of Dusp6 and BCI treatment restored clonal selectively and eradicated the Jak2V617F cells. Our study shows that inflammatory cytokines and JAK2V617F signaling converge to induce DUSP1, which downregulates p53 and establishes a higher apoptotic threshold. These data suggest that selectively targeting DUSP1 may provide a curative response in JAK2V617F-driven MPN.
Collapse
Affiliation(s)
- Meenu Kesarwani
- Division of Pathology, Cincinnati Children's Hospital, Cincinnati, OH, USA
| | - Zachary Kincaid
- Division of Pathology, Cincinnati Children's Hospital, Cincinnati, OH, USA
| | - Mohammad Azhar
- Division of Pathology, Cincinnati Children's Hospital, Cincinnati, OH, USA
| | - Jacob Menke
- Division of Pathology, Cincinnati Children's Hospital, Cincinnati, OH, USA
| | | | - Sekhu Ansari
- Division of Pathology, Cincinnati Children's Hospital, Cincinnati, OH, USA
| | - Angela Reaves
- Division of Pathology, Cincinnati Children's Hospital, Cincinnati, OH, USA
| | - Michael E Deininger
- Versiti Blood Research Institute and Medical College of Wisconsin, Milwaukee, WI, USA
| | - Ross Levine
- Center for Hematologic Malignancies, and Molecular Cancer Medicine Service, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - H Leighton Grimes
- Division of Immunobiology, Cincinnati Children's Hospital, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati, Cincinnati, OH, USA
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital, Cincinnati, OH, USA
| | - Mohammad Azam
- Division of Pathology, Cincinnati Children's Hospital, Cincinnati, OH, USA.
- Department of Pediatrics, University of Cincinnati, Cincinnati, OH, USA.
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital, Cincinnati, OH, USA.
| |
Collapse
|
22
|
Zhai J, Ji J, Liu J. Learning Causal Biological Networks with Parallel Ant Colony Optimization Algorithm. Bioengineering (Basel) 2023; 10:909. [PMID: 37627794 PMCID: PMC10451563 DOI: 10.3390/bioengineering10080909] [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: 06/12/2023] [Revised: 07/22/2023] [Accepted: 07/27/2023] [Indexed: 08/27/2023] Open
Abstract
A wealth of causal relationships exists in biological systems, both causal brain networks and causal protein signaling networks are very classical causal biological networks (CBNs). Learning CBNs from biological signal data reliably is a critical problem today. However, most of the existing methods are not excellent enough in terms of accuracy and time performance, and tend to fall into local optima because they do not take full advantage of global information. In this paper, we propose a parallel ant colony optimization algorithm to learn causal biological networks from biological signal data, called PACO. Specifically, PACO first maps the construction of CBNs to ants, then searches for CBNs in parallel by simulating multiple groups of ants foraging, and finally obtains the optimal CBN through pheromone fusion and CBNs fusion between different ant colonies. Extensive experimental results on simulation data sets as well as two real-world data sets, the fMRI signal data set and the Single-cell data set, show that PACO can accurately and efficiently learn CBNs from biological signal data.
Collapse
Affiliation(s)
| | | | - Jinduo Liu
- Beijing Municipal Key Laboratory of Multimedia and Intelligent Software Technology, Beijing Institute of Artificial Intelligence, Faculty of Information Technology, Beijing University of Technology, Beijing 100124, China (J.J.)
| |
Collapse
|
23
|
Chen H, Li J, Pi C, Guo D, Zhang D, Zhou X, Xie J. FGF19 induces the cell cycle arrest at G2-phase in chondrocytes. Cell Death Discov 2023; 9:250. [PMID: 37454120 DOI: 10.1038/s41420-023-01543-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 06/13/2023] [Accepted: 07/04/2023] [Indexed: 07/18/2023] Open
Abstract
Fibroblast growth factor 19 (FGF19) has appeared as a new possible avenue in the treatment of skeletal metabolic disorders. However, the role of FGF19 on cell cycle progression in skeletal system is poorly understood. Here we demonstrated that FGF19 had the ability to reduce the proliferation of chondrocytes and cause cell cycle G2 phase arrest through its interaction with β-Klotho (KLB), an important accessory protein that helps FGF19 link to its receptor. FGF19-mediated cell cycle arrest by regulating the expressions of cdk1/cylinb1, chk1 and gadd45a. We then confirmed that the binding of FGF19 to the membrane receptor FGFR4 was necessary for FGF19-mediated cell cycle arrest, and further proved that FGF19-mediated cell cycle arrest was via activation of p38/MAPK signaling. Through inhibitor experiments, we discovered that inhibition of FGFR4 led to down-regulation of p38 signaling even in the presence of FGF19. Meanwhile, inhibiting p38 signaling reduced the cell cycle arrest of chondrocytes induced by FGF19. Furthermore, blocking p38 signaling facilitated to retain the expression of cdk1 and cyclinb1 that had been reduced in chondrocytes by FGF19 and decreased the expression of chk1 and gadd45a that had been enhanced by FGF19 in chondrocytes. Taking together, this study is the first to demonstrate that FGF19 induces cell cycle arrest at G2 phase via FGFR4-p38/MAPK axis and enlarges our understanding about the role of FGF19 on cell cycle progression in chondrocytes.
Collapse
Affiliation(s)
- Hao Chen
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, 610041, Chengdu, China
| | - Jiazhou Li
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, 610041, Chengdu, China
| | - Caixia Pi
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, 610041, Chengdu, China
| | - Daimo Guo
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, 610041, Chengdu, China
| | - Demao Zhang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, 610041, Chengdu, China
| | - Xuedong Zhou
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, 610041, Chengdu, China.
- National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, 610041, Chengdu, China.
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, 610041, Chengdu, China.
| | - Jing Xie
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, 610041, Chengdu, China.
- National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, 610041, Chengdu, China.
| |
Collapse
|
24
|
Sularz O, Koronowicz A, Smoleń S, Boycott C, Stefanska B. Iodine-Biofortified Lettuce Can Promote Mitochondrial Dependent Pathway of Apoptosis in Human Gastrointestinal Cancer Cells. Int J Mol Sci 2023; 24:9869. [PMID: 37373017 PMCID: PMC10298746 DOI: 10.3390/ijms24129869] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 05/31/2023] [Accepted: 06/05/2023] [Indexed: 06/29/2023] Open
Abstract
Previously, our research provided evidence that exposure of gastric and colon cancer cells to extracts from iodine-biofortified lettuce leads to a reduction of cell viability and proliferation through cell cycle arrest and upregulation of pro-apoptotic genes. The aim of the present study was to determine the potential cellular mechanisms of induction of cell death in human gastrointestinal cancer cell lines after treatment with iodine-biofortified lettuce. We demonstrated that extracts from lettuce enriched with iodine induce apoptosis in gastric AGS and colon HT-29 cancer cells and the mechanism of programmed cell death may be triggered and executed through different signaling pathways, depending on the type of cells. Western blot analysis revealed that iodine-fortified lettuce leads to cell death through the release of cytochrome c to the cytosolic fraction and activation of the primary drivers of apoptosis: caspase-3, caspase-7, and caspase-9. Furthermore, we have reported that apoptotic effects of lettuce extracts may be mediated by poly (ADP-ribose) polymerase (PARP) and activation of pro-apoptotic Bcl-2 family proteins such as Bad, Bax, and BID. We also observed mitochondrial dysfunction with the dissipation of the mitochondrial membrane potential in cells exposed to lettuce extracts. Taken together, these results indicate that the organic form of iodine such as 5-ISA and 3,5-diISA is an important factor in the activation of intrinsic mitochondrial apoptotic pathway in AGS and HT-29 cancer cells in a p53-independent manner.
Collapse
Affiliation(s)
- Olga Sularz
- Department of Human Nutrition and Dietetics, Faculty of Food Technology, University of Agriculture in Krakow, Balicka 122, 31-149 Krakow, Poland;
| | - Aneta Koronowicz
- Department of Human Nutrition and Dietetics, Faculty of Food Technology, University of Agriculture in Krakow, Balicka 122, 31-149 Krakow, Poland;
| | - Sylwester Smoleń
- Department of Plant Biology and Biotechnology, Faculty of Biotechnology and Horticulture, University of Agriculture in Krakow, Al. 29 Listopada 54, 31-425 Krakow, Poland;
| | - Cayla Boycott
- Food, Nutrition and Health Program, Faculty of Land and Food Systems, The University of British Columbia, 2205 East Mall, Vancouver, BC V6T 1Z4, Canada;
| | - Barbara Stefanska
- Food, Nutrition and Health Program, Faculty of Land and Food Systems, The University of British Columbia, 2205 East Mall, Vancouver, BC V6T 1Z4, Canada;
| |
Collapse
|
25
|
Sun J, Xia F, Zhang S, Zhang B, Guan Y, Hu X, Xue P, Yang S, Zhou Y, Ling D, Li F. A Selective Nano Cell Cycle Checkpoint Inhibitor Overcomes Leukemia Chemoresistance. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2300736. [PMID: 37029565 DOI: 10.1002/smll.202300736] [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: 01/26/2023] [Revised: 03/17/2023] [Indexed: 06/19/2023]
Abstract
Cell cycle checkpoint activation promotes DNA damage repair, which is highly associated with the chemoresistance of various cancers including acute myeloid leukemia (AML). Selective cell cycle checkpoint inhibitors are strongly demanded to overcome chemoresistance, but remain unexplored. A selective nano cell cycle checkpoint inhibitor (NCCI: citric acid capped ultra-small iron oxide nanoparticles) that can catalytically inhibit the cell cycle checkpoint of AML to boost the chemotherapeutic efficacy of genotoxic agents is now reported. NCCI can selectively accumulate in AML cells and convert H2 O2 to • OH to cleave heat shock protein 90, leading to the degradation of ataxia telangiectasia and Rad3-related proteinand checkpoint kinase 1, and the subsequent dysfunction of the G2/M checkpoint. Consequently, NCCI revitalizes the anti-AML efficacy of cytarabine that is previously ineffective both in vitro and in vivo. This study offers new insights into designing selective cell cycle checkpoint inhibitors for biomedical applications.
Collapse
Affiliation(s)
- Jie Sun
- Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Liangzhu LaboratoryZhejiang University Medical CenterInstitute of Hematology, Zhejiang University, Hangzhou, 310058, China
| | - Fan Xia
- Institute of Pharmaceutics, Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Shaoqi Zhang
- Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Liangzhu LaboratoryZhejiang University Medical CenterInstitute of Hematology, Zhejiang University, Hangzhou, 310058, China
- Institute of Pharmaceutics, Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Bo Zhang
- Frontiers Science Center for Transformative Molecules, School of Chemistry and Chemical Engineering, National Center for Translational Medicine, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University, Shanghai, 200240, China
- WLA Laboratories, Shanghai, 201203, China
| | - Yunan Guan
- Institute of Pharmaceutics, Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Xi Hu
- Institute of Pharmaceutics, Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Frontiers Science Center for Transformative Molecules, School of Chemistry and Chemical Engineering, National Center for Translational Medicine, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University, Shanghai, 200240, China
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Pengpeng Xue
- Institute of Pharmaceutics, Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Shengfei Yang
- Institute of Pharmaceutics, Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yan Zhou
- Institute of Pharmaceutics, Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Daishun Ling
- Institute of Pharmaceutics, Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Frontiers Science Center for Transformative Molecules, School of Chemistry and Chemical Engineering, National Center for Translational Medicine, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University, Shanghai, 200240, China
- WLA Laboratories, Shanghai, 201203, China
| | - Fangyuan Li
- Institute of Pharmaceutics, Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Frontiers Science Center for Transformative Molecules, School of Chemistry and Chemical Engineering, National Center for Translational Medicine, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University, Shanghai, 200240, China
- WLA Laboratories, Shanghai, 201203, China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, 310009, China
| |
Collapse
|
26
|
Crabtree A, Boehnke N, Bates F, Hackel B. Consequences of poly(ethylene oxide) and poloxamer P188 on transcription in healthy and stressed myoblasts. Proc Natl Acad Sci U S A 2023; 120:e2219885120. [PMID: 37094151 PMCID: PMC10161009 DOI: 10.1073/pnas.2219885120] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 03/26/2023] [Indexed: 04/26/2023] Open
Abstract
Poly(ethylene oxide) (PEO) and poloxamers, a class of poly(ethylene oxide)-b-poly(propylene oxide)-b-poly(ethylene oxide) (PEO-PPO-PEO) triblock copolymers, have many personal and medical care applications, including the stabilization of stressed cellular membranes. Despite the widespread use, the cellular transcriptional response to these molecules is relatively unknown. C2C12 myoblasts, a model muscle cell, were subjected to short-term Poloxamer 188 (P188) and PEO181 (8,000 g/mol) treatment in culture. RNA was extracted and sequenced to quantify transcriptomic impact. The addition of moderate concentrations (14 µM) of either polymer to unstressed cells caused substantial differential gene expression, including at least twofold modulation of 357 and 588 genes, respectively. In addition, evaluation of the transcriptome response to osmotic stress without polymer treatment revealed dramatic change in RNA expression. Interestingly, the addition of polymer to stressed cells-at concentrations that provide physiological protection-did not yield a significant difference in expression of any gene relative to stress alone. Genome-scale expression analysis was corroborated by single-gene quantitative real-time PCR. Changes in protein expression were measured via western blot, which revealed partial alignment with the RNA results. Collectively, the significant changes to expression of multiple genes and resultant protein translation demonstrates an unexpectedly broad biochemical response to these polymers in healthy myoblasts in vitro. Meanwhile, the lack of substantial transcriptional response to polymer treatment in stressed cells highlights the physical nature of that protective mechanism.
Collapse
Affiliation(s)
- Adelyn A. Crabtree
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, MN55455
| | - Natalie Boehnke
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, MN55455
| | - Frank S. Bates
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, MN55455
| | - Benjamin J. Hackel
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, MN55455
| |
Collapse
|
27
|
Chen Y, Wu X, Liu X, Lai J, Gong Q. Comparative transcriptome analysis provides insights into the TDG supersaturation stress response of Schizothorax davidi. Comp Biochem Physiol C Toxicol Pharmacol 2023; 269:109618. [PMID: 37004899 DOI: 10.1016/j.cbpc.2023.109618] [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: 12/23/2022] [Revised: 03/23/2023] [Accepted: 03/27/2023] [Indexed: 04/03/2023]
Abstract
In the dam discharge season, the supersaturation of total dissolved gas (TDG) in the downstream channel can seriously affect the survival of aquatic organisms. However, few studies have revealed the mechanism by which TDG supersaturation affects the physiology of fish thus far. The present study was conducted to study the mechanism of the effect of TDG supersaturation on Schizothorax davidi, a species that is very sensitive to gas bubble disease. S. davidi was exposed to 116 % TDG supersaturation stress for 24 h. Serum biochemical tests showed that the aspartate aminotransferase and alanine aminotransferase levels after TDG supersaturation exposure were significantly decreased compared to those in the control group, while superoxide dismutase activity was significantly increased. RNA-Seq of gill tissues identified 1890 differentially expressed genes (DEGs), which consisted of 862 upregulated genes and 1028 downregulated genes, in the TDG supersaturation group vs. the control group. Pathway enrichment analysis revealed that the cell cycle, apoptosis and immune signaling pathways were affected by TDG stress. The results of this study may contribute to our understanding of the underlying molecular mechanism of environmental stress in fish.
Collapse
Affiliation(s)
- Yeyu Chen
- The Fishery Institute of the Sichuan Academy of Agricultural Sciences, Chengdu 611730, China
| | - Xiaoyun Wu
- The Fishery Institute of the Sichuan Academy of Agricultural Sciences, Chengdu 611730, China
| | - Xiaoqing Liu
- Key Laboratory of Fluid and Power Machinery, Ministry of Education, Xihua University, Chengdu 610039, China
| | - Jiansheng Lai
- The Fishery Institute of the Sichuan Academy of Agricultural Sciences, Chengdu 611730, China
| | - Quan Gong
- The Fishery Institute of the Sichuan Academy of Agricultural Sciences, Chengdu 611730, China.
| |
Collapse
|
28
|
Silic-Benussi M, Sharova E, Corradin A, Urso L, Raimondi V, Cavallari I, Buldini B, Francescato S, Minuzzo SA, D’Agostino DM, Ciminale V. Repurposing Verapamil to Enhance Killing of T-ALL Cells by the mTOR Inhibitor Everolimus. Antioxidants (Basel) 2023; 12:antiox12030625. [PMID: 36978873 PMCID: PMC10045900 DOI: 10.3390/antiox12030625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 02/27/2023] [Accepted: 03/01/2023] [Indexed: 03/06/2023] Open
Abstract
New therapies are needed for patients with T-cell lymphoblastic leukemia (T-ALL) who do not respond to standard chemotherapy. Our previous studies showed that the mTORC1 inhibitor everolimus increases reactive oxygen species (ROS) levels, decreases the levels of NADPH and glucose-6-phosphate dehydrogenase (G6PD), the rate-limiting enzyme of the pentose phosphate pathway (PPP), and induces apoptosis in T-ALL cells. Studies in T-ALL-xenografted NOD/SCID mice demonstrated that everolimus improved their response to the glucocorticoid (GC) dexamethasone. Here we show that verapamil, a calcium antagonist used in the treatment of supraventricular tachyarrhythmias, enhanced the effects of everolimus on ROS and cell death in T-ALL cell lines. The death-enhancing effect was synergistic and was confirmed in assays on a panel of therapy-resistant patient-derived xenografts (PDX) and primary samples from T-ALL patients. The verapamil-everolimus combination produced a dramatic reduction in the levels of G6PD and induction of p38 MAPK phosphorylation. Studies of NOD/SCID mice inoculated with refractory T-ALL PDX cells demonstrated that the addition of verapamil to everolimus plus dexamethasone significantly reduced tumor growth in vivo. Taken together, our results provide a rationale for repurposing verapamil in association with mTORC inhibitors and GC to treat refractory T-ALL.
Collapse
Affiliation(s)
- Micol Silic-Benussi
- Veneto Institute of Oncology IOV—IRCCS, 35128 Padova, Italy
- Correspondence: (M.S.-B.); (V.C.)
| | | | | | - Loredana Urso
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35128 Padova, Italy
| | - Vittoria Raimondi
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35128 Padova, Italy
| | | | - Barbara Buldini
- Pediatric Hemato Oncology, Maternal and Child Health Department, University of Padova, 35128 Padova, Italy
| | - Samuela Francescato
- Pediatric Hemato Oncology, Maternal and Child Health Department, University of Padova, 35128 Padova, Italy
| | - Sonia A. Minuzzo
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35128 Padova, Italy
| | - Donna M. D’Agostino
- Veneto Institute of Oncology IOV—IRCCS, 35128 Padova, Italy
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy
| | - Vincenzo Ciminale
- Veneto Institute of Oncology IOV—IRCCS, 35128 Padova, Italy
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35128 Padova, Italy
- Correspondence: (M.S.-B.); (V.C.)
| |
Collapse
|
29
|
Han H, Alsayed AMM, Wang Y, Yan Q, Shen A, Zhang J, Ye Y, Liu Z, Wang K, Zheng X. Discovery of β-cyclocitral-derived mono-carbonyl curcumin analogs as anti-hepatocellular carcinoma agents via suppression of MAPK signaling pathway. Bioorg Chem 2023; 132:106358. [PMID: 36642021 DOI: 10.1016/j.bioorg.2023.106358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 11/22/2022] [Accepted: 01/08/2023] [Indexed: 01/12/2023]
Abstract
Hepatocellular carcinoma (HCC) is one of the most common malignant tumors with a high recurrence and mortality rate. In this study, a series of β-cyclocitral-derived mono-carbonyl curcumin analogs were synthesized and their anticancer properties were evaluated. Among the series, A19 exhibited the strongest cytotoxic activity by inhibiting cell viability and colony formation, inducing cell cycle G2/M phase arrest and cell apoptosis of HCC HepG2 and Huh-7 cells, while having almost no cytotoxicity on normal liver MIHA cells. Mechanistically, our results demonstrated that A19 triggered intense DNA damage via suppression of the ERK/JNK/p38 MAPK signaling pathway. Additionally, a combination of A19 with sorafenib significantly induced synergistic cytotoxicity in HCC cells. Overall, our results indicate the potential of A19 as a novel chemotherapeutic drug administered either separately or in combined therapy for HCC treatment.
Collapse
Affiliation(s)
- Haoyi Han
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, 1210 University Town, Wenzhou, Zhejiang 325035, China
| | - Ali Mohammed Mohammed Alsayed
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, 1210 University Town, Wenzhou, Zhejiang 325035, China
| | - Yi Wang
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, 1210 University Town, Wenzhou, Zhejiang 325035, China
| | - Qi Yan
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, 1210 University Town, Wenzhou, Zhejiang 325035, China
| | - Ancheng Shen
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, 1210 University Town, Wenzhou, Zhejiang 325035, China
| | - Jianxia Zhang
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, 1210 University Town, Wenzhou, Zhejiang 325035, China
| | - Yanfei Ye
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, 1210 University Town, Wenzhou, Zhejiang 325035, China
| | - Zhiguo Liu
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, 1210 University Town, Wenzhou, Zhejiang 325035, China.
| | - Kun Wang
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, 1210 University Town, Wenzhou, Zhejiang 325035, China.
| | - Xiaohui Zheng
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, 1210 University Town, Wenzhou, Zhejiang 325035, China.
| |
Collapse
|
30
|
Signaling pathways in rheumatoid arthritis: implications for targeted therapy. Signal Transduct Target Ther 2023; 8:68. [PMID: 36797236 PMCID: PMC9935929 DOI: 10.1038/s41392-023-01331-9] [Citation(s) in RCA: 97] [Impact Index Per Article: 97.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 12/16/2022] [Accepted: 01/18/2023] [Indexed: 02/18/2023] Open
Abstract
Rheumatoid arthritis (RA) is an incurable systemic autoimmune disease. Disease progression leads to joint deformity and associated loss of function, which significantly impacts the quality of life for sufferers and adds to losses in the labor force. In the past few decades, RA has attracted increased attention from researchers, the abnormal signaling pathways in RA are a very important research field in the diagnosis and treatment of RA, which provides important evidence for understanding this complex disease and developing novel RA-linked intervention targets. The current review intends to provide a comprehensive overview of RA, including a general introduction to the disease, historical events, epidemiology, risk factors, and pathological process, highlight the primary research progress of the disease and various signaling pathways and molecular mechanisms, including genetic factors, epigenetic factors, summarize the most recent developments in identifying novel signaling pathways in RA and new inhibitors for treating RA. therapeutic interventions including approved drugs, clinical drugs, pre-clinical drugs, and cutting-edge therapeutic technologies. These developments will hopefully drive progress in new strategically targeted therapies and hope to provide novel ideas for RA treatment options in the future.
Collapse
|
31
|
García-Flores N, Jiménez-Suárez J, Garnés-García C, Fernández-Aroca DM, Sabater S, Andrés I, Fernández-Aramburo A, Ruiz-Hidalgo MJ, Belandia B, Sanchez-Prieto R, Cimas FJ. P38 MAPK and Radiotherapy: Foes or Friends? Cancers (Basel) 2023; 15:861. [PMID: 36765819 PMCID: PMC9913882 DOI: 10.3390/cancers15030861] [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: 12/03/2022] [Revised: 01/16/2023] [Accepted: 01/24/2023] [Indexed: 01/31/2023] Open
Abstract
Over the last 30 years, the study of the cellular response to ionizing radiation (IR) has increased exponentially. Among the various signaling pathways affected by IR, p38 MAPK has been shown to be activated both in vitro and in vivo, with involvement in key processes triggered by IR-mediated genotoxic insult, such as the cell cycle, apoptosis or senescence. However, we do not yet have a definitive clue about the role of p38 MAPK in terms of radioresistance/sensitivity and its potential use to improve current radiotherapy. In this review, we summarize the current knowledge on this family of MAPKs in response to IR as well as in different aspects related to radiotherapy, such as their role in the control of REDOX, fibrosis, and in the radiosensitizing effect of several compounds.
Collapse
Affiliation(s)
- Natalia García-Flores
- Laboratorio de Oncología Molecular, Unidad de Medicina Molecular, Centro Regional de Investigaciones Biomédicas, Unidad Asociada de Biomedicina UCLM, Unidad Asociada al CSIC, Universidad de Castilla-La Mancha, 02008 Albacete, Spain
| | - Jaime Jiménez-Suárez
- Laboratorio de Oncología Molecular, Unidad de Medicina Molecular, Centro Regional de Investigaciones Biomédicas, Unidad Asociada de Biomedicina UCLM, Unidad Asociada al CSIC, Universidad de Castilla-La Mancha, 02008 Albacete, Spain
| | - Cristina Garnés-García
- Laboratorio de Oncología Molecular, Unidad de Medicina Molecular, Centro Regional de Investigaciones Biomédicas, Unidad Asociada de Biomedicina UCLM, Unidad Asociada al CSIC, Universidad de Castilla-La Mancha, 02008 Albacete, Spain
| | - Diego M. Fernández-Aroca
- Laboratorio de Oncología Molecular, Unidad de Medicina Molecular, Centro Regional de Investigaciones Biomédicas, Unidad Asociada de Biomedicina UCLM, Unidad Asociada al CSIC, Universidad de Castilla-La Mancha, 02008 Albacete, Spain
| | - Sebastia Sabater
- Laboratorio de Oncología Molecular, Unidad de Medicina Molecular, Centro Regional de Investigaciones Biomédicas, Unidad Asociada de Biomedicina UCLM, Unidad Asociada al CSIC, Universidad de Castilla-La Mancha, 02008 Albacete, Spain
- Servicio de Oncología Radioterápica, Complejo Hospitalario Universitario de Albacete, 02006 Albacete, Spain
| | - Ignacio Andrés
- Laboratorio de Oncología Molecular, Unidad de Medicina Molecular, Centro Regional de Investigaciones Biomédicas, Unidad Asociada de Biomedicina UCLM, Unidad Asociada al CSIC, Universidad de Castilla-La Mancha, 02008 Albacete, Spain
- Servicio de Oncología Radioterápica, Complejo Hospitalario Universitario de Albacete, 02006 Albacete, Spain
| | - Antonio Fernández-Aramburo
- Laboratorio de Oncología Molecular, Unidad de Medicina Molecular, Centro Regional de Investigaciones Biomédicas, Unidad Asociada de Biomedicina UCLM, Unidad Asociada al CSIC, Universidad de Castilla-La Mancha, 02008 Albacete, Spain
- Servicio de Oncología Médica, Complejo Hospitalario Universitario de Albacete, 02006 Albacete, Spain
| | - María José Ruiz-Hidalgo
- Laboratorio de Oncología Molecular, Unidad de Medicina Molecular, Centro Regional de Investigaciones Biomédicas, Unidad Asociada de Biomedicina UCLM, Unidad Asociada al CSIC, Universidad de Castilla-La Mancha, 02008 Albacete, Spain
- Departamento de Química Inorgánica, Orgánica y Bioquímica, Área de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad de Castilla-La Mancha, 02008 Albacete, Spain
| | - Borja Belandia
- Departamento de Biología del Cáncer, Instituto de Investigaciones Biomédicas ‘Alberto Sols’ (CSIC-UAM), Unidad Asociada de Biomedicina UCLM, Unidad Asociada al CSIC, 28029 Madrid, Spain
| | - Ricardo Sanchez-Prieto
- Laboratorio de Oncología Molecular, Unidad de Medicina Molecular, Centro Regional de Investigaciones Biomédicas, Unidad Asociada de Biomedicina UCLM, Unidad Asociada al CSIC, Universidad de Castilla-La Mancha, 02008 Albacete, Spain
- Departamento de Biología del Cáncer, Instituto de Investigaciones Biomédicas ‘Alberto Sols’ (CSIC-UAM), Unidad Asociada de Biomedicina UCLM, Unidad Asociada al CSIC, 28029 Madrid, Spain
- Departamento de Ciencias Médicas, Facultad de Medicina, Universidad de Castilla-La Mancha, 02008 Albacete, Spain
| | - Francisco J. Cimas
- Laboratorio de Oncología Molecular, Unidad de Medicina Molecular, Centro Regional de Investigaciones Biomédicas, Unidad Asociada de Biomedicina UCLM, Unidad Asociada al CSIC, Universidad de Castilla-La Mancha, 02008 Albacete, Spain
- Departamento de Química Inorgánica, Orgánica y Bioquímica, Área de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad de Castilla-La Mancha, 02008 Albacete, Spain
| |
Collapse
|
32
|
Dayanidhi DL, Somarelli JA, Mantyh JB, Rupprecht G, Roghani RS, Vincoff S, Shin I, Zhao Y, Kim SY, McCall S, Hong J, Hsu DS. Psymberin, a marine-derived natural product, induces cancer cell growth arrest and protein translation inhibition. Front Med (Lausanne) 2022; 9:999004. [PMID: 36743670 PMCID: PMC9894252 DOI: 10.3389/fmed.2022.999004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 10/18/2022] [Indexed: 01/20/2023] Open
Abstract
Colorectal cancer (CRC) is the third most prevalent form of cancer in the United States and results in over 50,000 deaths per year. Treatments for metastatic CRC are limited, and therefore there is an unmet clinical need for more effective therapies. In our prior work, we coupled high-throughput chemical screens with patient-derived models of cancer to identify new potential therapeutic targets for CRC. However, this pipeline is limited by (1) the use of cell lines that do not appropriately recapitulate the tumor microenvironment, and (2) the use of patient-derived xenografts (PDXs), which are time-consuming and costly for validation of drug efficacy. To overcome these limitations, we have turned to patient-derived organoids. Organoids are increasingly being accepted as a "standard" preclinical model that recapitulates tumor microenvironment cross-talk in a rapid, cost-effective platform. In the present work, we employed a library of natural products, intermediates, and drug-like compounds for which full synthesis has been demonstrated. Using this compound library, we performed a high-throughput screen on multiple low-passage cancer cell lines to identify potential treatments. The top candidate, psymberin, was further validated, with a focus on CRC cell lines and organoids. Mechanistic and genomics analyses pinpointed protein translation inhibition as a mechanism of action of psymberin. These findings suggest the potential of psymberin as a novel therapy for the treatment of CRC.
Collapse
Affiliation(s)
- Divya L. Dayanidhi
- Division of Medical Oncology, Department of Medicine, Duke University Medical Center, Durham, NC, United States
- Center for Genomics and Computational Biology, Duke University, Durham, NC, United States
| | - Jason A. Somarelli
- Division of Medical Oncology, Department of Medicine, Duke University Medical Center, Durham, NC, United States
| | - John B. Mantyh
- Division of Medical Oncology, Department of Medicine, Duke University Medical Center, Durham, NC, United States
- Center for Genomics and Computational Biology, Duke University, Durham, NC, United States
| | - Gabrielle Rupprecht
- Division of Medical Oncology, Department of Medicine, Duke University Medical Center, Durham, NC, United States
- Center for Genomics and Computational Biology, Duke University, Durham, NC, United States
| | - Roham Salman Roghani
- Division of Medical Oncology, Department of Medicine, Duke University Medical Center, Durham, NC, United States
- Center for Genomics and Computational Biology, Duke University, Durham, NC, United States
| | - Sophia Vincoff
- Division of Medical Oncology, Department of Medicine, Duke University Medical Center, Durham, NC, United States
| | - Iljin Shin
- Department of Chemistry, Duke University, Durham, NC, United States
| | - Yiquan Zhao
- Department of Chemistry, Duke University, Durham, NC, United States
| | - So Young Kim
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, United States
| | - Shannon McCall
- Department of Pathology, Duke University, Durham, NC, United States
| | - Jiyong Hong
- Department of Chemistry, Duke University, Durham, NC, United States
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, United States
| | - David S. Hsu
- Division of Medical Oncology, Department of Medicine, Duke University Medical Center, Durham, NC, United States
- Center for Genomics and Computational Biology, Duke University, Durham, NC, United States
| |
Collapse
|
33
|
Qian J, Hu Y, Zhang X, Chi M, Xu S, Wang H, Zhang X. Mycobacterium tuberculosis PE_PGRS19 Induces Pyroptosis through a Non-Classical Caspase-11/GSDMD Pathway in Macrophages. Microorganisms 2022; 10:microorganisms10122473. [PMID: 36557726 PMCID: PMC9785159 DOI: 10.3390/microorganisms10122473] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 12/10/2022] [Indexed: 12/23/2022] Open
Abstract
The PE/PPE protein family commonly exists in pathogenic species, such as Mycobacterium tuberculosis, suggesting a role in virulence and its maintenance. However, the exact role of most PE/PPE proteins in host-pathogen interactions remains unknown. Here, we constructed a recombinant Mycobacterium smegmatis expressing M. tuberculosis PE_PGRS19 (Ms_PE_PGRS19) and found that PE_PGRS19 overexpression resulted in accelerated bacterial growth in vitro, increased bacterial survival in macrophages, and enhanced cell damage capacity. Ms_PE_PGRS19 also induced the expression of pro-inflammatory cytokines, such as IL-6, TNF-α, IL-1β, and IL-18. Furthermore, we demonstrated that Ms_PE_PGRS19 induced cell pyroptosis by cleaving caspase-11 via a non-classical pathway rather than caspase-1 activation and further inducing the cleavage of gasdermin D, which led to the release of IL-1β and IL-18. To the best of our current knowledge, this is the first report of a PE/PPE family protein activating cell pyroptosis via a non-classical pathway, which expands the knowledge on PE/PPE protein functions, and these pathogenic factors involved in bacterial survival and spread could be potential drug targets for anti-tuberculosis therapy.
Collapse
|
34
|
Wang Z, An J, Zhu D, Chen H, Lin A, Kang J, Liu W, Kang X. Periostin: an emerging activator of multiple signaling pathways. J Cell Commun Signal 2022; 16:515-530. [PMID: 35412260 PMCID: PMC9733775 DOI: 10.1007/s12079-022-00674-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 03/07/2022] [Indexed: 12/13/2022] Open
Abstract
Matricellular proteins are responsible for regulating the microenvironment, the behaviors of surrounding cells, and the homeostasis of tissues. Periostin (POSTN), a non-structural matricellular protein, can bind to many extracellular matrix proteins through its different domains. POSTN usually presents at low levels in most adult tissues but is highly expressed in pathological sites such as in tumors and inflamed organs. POSTN can bind to diverse integrins to interact with multiple signaling pathways within cells, which is one of its core biological functions. Increasing evidence shows that POSTN can activate the TGF-β, the PI3K/Akt, the Wnt, the RhoA/ROCK, the NF-κB, the MAPK and the JAK pathways to promote the occurrence and development of many diseases, especially cancer and inflammatory diseases. Furthermore, POSTN can interact with some pathways in an upstream and downstream relationship, forming complicated crosstalk. This article focuses on the interactions between POSTN and different signaling pathways in diverse diseases, attempting to explain the mechanisms of interaction and provide novel guidelines for the development of targeted therapies.
Collapse
Affiliation(s)
- Zhaoheng Wang
- grid.411294.b0000 0004 1798 9345Lanzhou University Second Hospital, 82, Cuiyingmen, Lanzhou, 730030 People’s Republic of China ,Orthopaedics Key Laboratory of Gansu Province, Lanzhou, 730030 People’s Republic of China
| | - Jiangdong An
- grid.411294.b0000 0004 1798 9345Lanzhou University Second Hospital, 82, Cuiyingmen, Lanzhou, 730030 People’s Republic of China
| | - Daxue Zhu
- grid.411294.b0000 0004 1798 9345Lanzhou University Second Hospital, 82, Cuiyingmen, Lanzhou, 730030 People’s Republic of China ,Orthopaedics Key Laboratory of Gansu Province, Lanzhou, 730030 People’s Republic of China
| | - Haiwei Chen
- grid.411294.b0000 0004 1798 9345Lanzhou University Second Hospital, 82, Cuiyingmen, Lanzhou, 730030 People’s Republic of China ,Orthopaedics Key Laboratory of Gansu Province, Lanzhou, 730030 People’s Republic of China
| | - Aixin Lin
- grid.411294.b0000 0004 1798 9345Lanzhou University Second Hospital, 82, Cuiyingmen, Lanzhou, 730030 People’s Republic of China ,Orthopaedics Key Laboratory of Gansu Province, Lanzhou, 730030 People’s Republic of China
| | - Jihe Kang
- grid.411294.b0000 0004 1798 9345Lanzhou University Second Hospital, 82, Cuiyingmen, Lanzhou, 730030 People’s Republic of China ,Orthopaedics Key Laboratory of Gansu Province, Lanzhou, 730030 People’s Republic of China
| | - Wenzhao Liu
- grid.411294.b0000 0004 1798 9345Lanzhou University Second Hospital, 82, Cuiyingmen, Lanzhou, 730030 People’s Republic of China ,Orthopaedics Key Laboratory of Gansu Province, Lanzhou, 730030 People’s Republic of China
| | - Xuewen Kang
- grid.411294.b0000 0004 1798 9345Lanzhou University Second Hospital, 82, Cuiyingmen, Lanzhou, 730030 People’s Republic of China ,Orthopaedics Key Laboratory of Gansu Province, Lanzhou, 730030 People’s Republic of China
| |
Collapse
|
35
|
Radnaa E, Richardson L, Goldman B, Burks J, Baljinnyam T, Vora N, Zhang HJ, Bonney E, Han A, Menon R. Stress signaler p38 mitogen-activated kinase activation: a cause for concern? Clin Sci (Lond) 2022; 136:1591-1614. [PMID: 36250628 PMCID: PMC9664350 DOI: 10.1042/cs20220491] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 10/14/2022] [Accepted: 10/17/2022] [Indexed: 11/17/2022]
Abstract
Oxidative stress (OS) induced activation of p38 mitogen-activated kinase (MAPK) and cell fate from p38 signaling was tested using the human fetal membrane's amnion epithelial cells (AEC). We created p38 KO AEC using the CRISPR/Cas9 approach and tested cell fate in response to OS on an AEC-free fetal membrane extracellular matrix (ECM). Screening using image CyTOF indicated OS causing epithelial-mesenchymal transition (EMT). Further testing revealed p38 deficiency prevented AEC senescence, EMT, cell migration, and inflammation. To functionally validate in vitro findings, fetal membrane-specific conditional KO (cKO) mice were developed by injecting Cre-recombinase encoded exosomes intra-amniotically into p38αloxP/loxP mice. Amnion membranes from p38 cKO mice had reduced senescence, EMT, and increased anti-inflammatory IL-10 compared with WT animals. Our study suggested that overwhelming activation of p38 in response to OS inducing risk exposures can have an adverse impact on cells, cause cell invasion, inflammation, and ECM degradation detrimental to tissue homeostasis.
Collapse
Affiliation(s)
- Enkhtuya Radnaa
- Division of Basic and Translational Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas, U.S.A
| | - Lauren Richardson
- Division of Basic and Translational Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas, U.S.A
| | - Brett Goldman
- Division of Basic and Translational Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas, U.S.A
| | - Jared K. Burks
- Flow Cytometry and Cellular Imaging Core Facility, Department of Leukemia, M.D. Anderson Cancer Center, Texas, U.S.A. 77030
| | - Tuvshintugs Baljinnyam
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch at Galveston, Galveston, Texas, U.S.A. 77555
| | - Natasha Vora
- Division of Basic and Translational Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas, U.S.A
| | - Hui-juan Zhang
- Department of Pathology, The International Peace Maternity and Child Health Hospital, University School of Medicine, Shanghai, China. 200030
| | - Elizabeth A. Bonney
- Department of Obstetrics and Gynecology, The University of Vermont, Burlington, VT, U.S.A. 05405ghout all figures, the following notations were
| | - Arum Han
- Department of Electrical and Computer Engineering, Department of Biomedical Engineering, Texas A&M University, College Station, Texas, U.S.A. 77843
| | - Ramkumar Menon
- Division of Basic and Translational Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas, U.S.A
| |
Collapse
|
36
|
Li T, Bai H, Yang L, Hao W, Wei S, Yan P. Low temperature exposure inhibits proliferation and induces apoptosis of bovine subcutaneous preadipocytes via p38 MAPK/JNK activation. Comp Biochem Physiol B Biochem Mol Biol 2022; 264:110813. [DOI: 10.1016/j.cbpb.2022.110813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 11/04/2022] [Accepted: 11/14/2022] [Indexed: 11/18/2022]
|
37
|
Su X, Wu W, Zhu Z, Lin X, Zeng Y. The effects of epithelial-mesenchymal transitions in COPD induced by cigarette smoke: an update. Respir Res 2022; 23:225. [PMID: 36045410 PMCID: PMC9429334 DOI: 10.1186/s12931-022-02153-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 08/25/2022] [Indexed: 12/15/2022] Open
Abstract
Cigarette smoke is a complex aerosol containing a large number of compounds with a variety of toxicity and carcinogenicity. Long-term exposure to cigarette smoke significantly increases the risk of a variety of diseases, including chronic obstructive pulmonary disease (COPD) and lung cancer. Epithelial–mesenchymal transition (EMT) is a unique biological process, that refers to epithelial cells losing their polarity and transforming into mobile mesenchymal cells, playing a crucial role in organ development, fibrosis, and cancer progression. Numerous recent studies have shown that EMT is an important pathophysiological process involved in airway fibrosis, airway remodeling, and malignant transformation of COPD. In this review, we summarized the effects of cigarette smoke on the development and progression of COPD and focus on the specific changes and underlying mechanisms of EMT in COPD induced by cigarette smoke. We spotlighted the signaling pathways involved in EMT induced by cigarette smoke and summarize the current research and treatment approaches for EMT in COPD, aiming to provide ideas for potential new treatment and research directions.
Collapse
Affiliation(s)
- Xiaoshan Su
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Fujian Medical University, Respirology Medicine Centre of Fujian Province, Quanzhou, China
| | - Weijing Wu
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Fujian Medical University, Respirology Medicine Centre of Fujian Province, Quanzhou, China
| | - Zhixing Zhu
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Fujian Medical University, Respirology Medicine Centre of Fujian Province, Quanzhou, China
| | - Xiaoping Lin
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Fujian Medical University, Respirology Medicine Centre of Fujian Province, Quanzhou, China
| | - Yiming Zeng
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Fujian Medical University, Respirology Medicine Centre of Fujian Province, Quanzhou, China.
| |
Collapse
|
38
|
Liu X, Chipurupalli S, Jiang P, Tavasoli M, Yoo BH, McPhee M, Mazinani S, Francia G, Kerbel RS, Rosen KV. ErbB2/Her2-dependent downregulation of a cell death-promoting protein BLNK in breast cancer cells is required for 3D breast tumor growth. Cell Death Dis 2022; 13:687. [PMID: 35933456 PMCID: PMC9357009 DOI: 10.1038/s41419-022-05117-9] [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: 01/21/2022] [Revised: 07/15/2022] [Accepted: 07/19/2022] [Indexed: 01/21/2023]
Abstract
A significant proportion of breast cancers are driven by ErbB2/Her2 oncoprotein that they overexpress. These malignancies are typically treated with various ErbB2-targeted drugs, but many such cancers develop resistance to these agents and become incurable. Conceivably, treatment of ErbB2-positive cancers could be facilitated by use of agents blocking oncogenic signaling mechanisms downstream of ErbB2. However, current understanding of these mechanisms is limited. The ability of solid tumor cells to resist anoikis, cell death triggered by cell detachment from the extracellular matrix (ECM), is thought to be critical for 3D tumor growth. In an effort to understand the mechanisms of ErbB2-driven breast cancer cell anoikis resistance we found that detachment of non-malignant breast epithelial cells from the ECM upregulates a cell death-promoting tumor suppressor adapter protein BLNK and that ErbB2 blocks this upregulation by reducing tumor cell levels of transcription factor IRF6. We further observed that trastuzumab, a therapeutic anti-ErbB2 antibody, upregulates BLNK in human trastuzumab-sensitive but not trastuzumab-resistant ErbB2-positive breast cancer cells. Moreover, we established that BLNK promotes anoikis by activating p38 MAP kinase and that ErbB2-dependent BLNK downregulation blocks breast cancer cell anoikis. In search for pharmacological approaches allowing to upregulate BLNK in tumor cells we found that clinically approved proteasome inhibitor bortezomib upregulates IRF6 and BLNK in human breast cancer cells and inhibits their 3D growth in a BLNK-dependent manner. In addition, we found that BLNK upregulation in human ErbB2-positive breast cancer cells blocks their ability to form tumors in mice. Furthermore, we used publicly available data on mRNA levels in multiple breast cancers to demonstrate that increased BLNK mRNA levels correlate with increased relapse-free survival in a cohort of approximately 400 patients with ErbB2-positive breast cancer. In summary, we discovered a novel mechanism of ErbB2-driven 3D breast tumor growth mediated by ErbB2-dependent BLNK downregulation.
Collapse
Affiliation(s)
- Xiaoyang Liu
- grid.55602.340000 0004 1936 8200Departments of Pediatrics & Biochemistry and Molecular Biology, Dalhousie University, Halifax, NS Canada
| | - Sandhya Chipurupalli
- grid.55602.340000 0004 1936 8200Departments of Pediatrics & Biochemistry and Molecular Biology, Dalhousie University, Halifax, NS Canada
| | - Peijia Jiang
- grid.55602.340000 0004 1936 8200Departments of Pediatrics & Biochemistry and Molecular Biology, Dalhousie University, Halifax, NS Canada
| | - Mahtab Tavasoli
- grid.55602.340000 0004 1936 8200Department of Pharmacology, Department of Pediatrics, Dalhousie University, Halifax, NS Canada
| | - Byong Hoon Yoo
- grid.55602.340000 0004 1936 8200Departments of Pediatrics & Biochemistry and Molecular Biology, Dalhousie University, Halifax, NS Canada
| | - Michael McPhee
- grid.55602.340000 0004 1936 8200Departments of Pediatrics & Biochemistry and Molecular Biology, Dalhousie University, Halifax, NS Canada
| | - Sina Mazinani
- grid.55602.340000 0004 1936 8200Departments of Pediatrics & Biochemistry and Molecular Biology, Dalhousie University, Halifax, NS Canada
| | - Giulio Francia
- grid.267324.60000 0001 0668 0420Border Biomedical Research Center, University of Texas at El Paso (UTEP), El Paso, TX USA
| | - Robert S. Kerbel
- grid.17063.330000 0001 2157 2938Biological Sciences Platform, Sunnybrook Research Institute, Toronto, ON Canada ,grid.17063.330000 0001 2157 2938University of Toronto Department of Medical Biophysics, Toronto, ON Canada
| | - Kirill V. Rosen
- grid.55602.340000 0004 1936 8200Departments of Pediatrics & Biochemistry and Molecular Biology, Dalhousie University, Halifax, NS Canada
| |
Collapse
|
39
|
Zhao Y, Dong J, Liao Y, Wang H, Zhou D, Kang J, Chen X. Identification and validation of four photodynamic therapy related genes inhibiting MAPK and inducing cell cycle alteration in squamous cell carcinoma. Front Oncol 2022; 12:946493. [PMID: 35992777 PMCID: PMC9386316 DOI: 10.3389/fonc.2022.946493] [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: 05/17/2022] [Accepted: 07/13/2022] [Indexed: 11/13/2022] Open
Abstract
Introduction Cutaneous squamous cell carcinoma (cSCC) is the second most common skin cancer, and photodynamic therapy (PDT) is a promising modality against cSCC. This study investigated the impact of PDT on the MAPK pathway and cell cycle alternation of cSCC as well as the related molecular mechanisms. Method Expressing mRNA profile data sets GSE98767, GSE45216, and GSE84758 were acquired from the GEO database. The functions of differently expressed genes (DEGs) were enriched by Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis. Least absolute shrinkage and selection operator (Lasso) analysis were used to establish a diagnosis model based on GSE98767. A correlation analysis and a protein–protein interaction (PPI) network were used to evaluate the relationship between cSCC-PDT-related genes and the MAPK pathway. Single-sample gene set enrichment analysis (ssGSEA) was performed on GSE98767 to estimate MAPK activation and cell cycle activity. Finally, the effect of MAPK activation on the cell cycle was explored in vitro. Result Four cSCC-PDT-related genes, DUSP6, EFNB2, DNAJB1, and CCNL1, were identified as diagnostic markers of cSCC, which were upregulated in cSCC or LC50 PDT-protocol treatment and negatively correlated with the MAPK promoter. Despite having a smaller MAPK activation score, cSCC showed higher cell cycle activity. The PDT treatment suppressed the G1 to G2/M phase in JNK overexpressed A431 cells. Conclusion CCNL1, DNAJB1, DUSP6, and EFNB2 were identified as potential PDT target genes in cSCC treatment, whose potential therapeutic mechanism was inhibiting the MAPK pathway and inducing cell cycle alternation.
Collapse
Affiliation(s)
- Yingchao Zhao
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Jianxiang Dong
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Yuxuan Liao
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Hongyi Wang
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Dawei Zhou
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Jian Kang
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Jian Kang, ; Xiang Chen,
| | - Xiang Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Jian Kang, ; Xiang Chen,
| |
Collapse
|
40
|
Silva GF, da Silva JS, de Alencar AKN, de Moraes Carvalho da Silva M, Montagnoli TL, de Souza Rocha B, de Freitas RHCN, Sudo RT, Fraga CAM, Zapata-Sudo G. Novel p38 Mitogen-Activated Protein Kinase Inhibitor Reverses Hypoxia-Induced Pulmonary Arterial Hypertension in Rats. Pharmaceuticals (Basel) 2022; 15:ph15070900. [PMID: 35890198 PMCID: PMC9316801 DOI: 10.3390/ph15070900] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 07/05/2022] [Accepted: 07/16/2022] [Indexed: 11/22/2022] Open
Abstract
Mitogen-activated protein kinase (MAPK) signaling is strongly implicated in cardiovascular remodeling in pulmonary hypertension (PH) and right ventricle (RV) failure. The effects of a newly designed p38 inhibitor, LASSBio-1824, were investigated in experimentally induced PH. Male Wistar rats were exposed to hypoxia and SU5416 (SuHx), and normoxic rats were used as controls. Oral treatment was performed for 14 days with either vehicle or LASSBio-1824 (50 mg/kg). Pulmonary vascular resistance and RV structure and function were assessed by echocardiography and catheterization. Histological, immunohistochemical and Western blot analysis of lung and RV were performed to investigate cardiovascular remodeling and inflammation. Treatment with LASSBio-1824 normalized vascular resistance by attenuating vessel muscularization and endothelial dysfunction. In the heart, treatment decreased RV systolic pressure, hypertrophy and collagen content, improving cardiac function. Protein content of TNF-α, iNOS, phosphorylated p38 and caspase-3 were reduced both in lung vessels and RV tissues after treatment and a reduced activation of transcription factor c-fos was found in cardiomyocytes of treated SuHx rats. Therefore, LASSBio-1824 represents a potential candidate for remodeling-targeted treatment of PH.
Collapse
Affiliation(s)
- Grazielle Fernandes Silva
- Programa de Pesquisa em Desenvolvimento de Fármacos, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (G.F.S.); (J.S.d.S.); (A.K.N.d.A.); (M.d.M.C.d.S.); (T.L.M.); (B.d.S.R.); (R.H.C.N.d.F.); or (R.T.S.)
- Programa de Pós-Graduação em Cardiologia, Instituto do Coração Edson Saad, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-913, RJ, Brazil
| | - Jaqueline Soares da Silva
- Programa de Pesquisa em Desenvolvimento de Fármacos, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (G.F.S.); (J.S.d.S.); (A.K.N.d.A.); (M.d.M.C.d.S.); (T.L.M.); (B.d.S.R.); (R.H.C.N.d.F.); or (R.T.S.)
- Programa de Pós-Graduação em Cardiologia, Instituto do Coração Edson Saad, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-913, RJ, Brazil
| | - Allan Kardec Nogueira de Alencar
- Programa de Pesquisa em Desenvolvimento de Fármacos, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (G.F.S.); (J.S.d.S.); (A.K.N.d.A.); (M.d.M.C.d.S.); (T.L.M.); (B.d.S.R.); (R.H.C.N.d.F.); or (R.T.S.)
| | - Marina de Moraes Carvalho da Silva
- Programa de Pesquisa em Desenvolvimento de Fármacos, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (G.F.S.); (J.S.d.S.); (A.K.N.d.A.); (M.d.M.C.d.S.); (T.L.M.); (B.d.S.R.); (R.H.C.N.d.F.); or (R.T.S.)
- Programa de Pós-Graduação em Farmacologia e Química Medicinal, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil
| | - Tadeu Lima Montagnoli
- Programa de Pesquisa em Desenvolvimento de Fármacos, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (G.F.S.); (J.S.d.S.); (A.K.N.d.A.); (M.d.M.C.d.S.); (T.L.M.); (B.d.S.R.); (R.H.C.N.d.F.); or (R.T.S.)
- Programa de Pós-Graduação em Farmacologia e Química Medicinal, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil
| | - Bruna de Souza Rocha
- Programa de Pesquisa em Desenvolvimento de Fármacos, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (G.F.S.); (J.S.d.S.); (A.K.N.d.A.); (M.d.M.C.d.S.); (T.L.M.); (B.d.S.R.); (R.H.C.N.d.F.); or (R.T.S.)
- Programa de Pós-Graduação em Cardiologia, Instituto do Coração Edson Saad, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-913, RJ, Brazil
| | - Rosana Helena Coimbra Nogueira de Freitas
- Programa de Pesquisa em Desenvolvimento de Fármacos, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (G.F.S.); (J.S.d.S.); (A.K.N.d.A.); (M.d.M.C.d.S.); (T.L.M.); (B.d.S.R.); (R.H.C.N.d.F.); or (R.T.S.)
| | - Roberto Takashi Sudo
- Programa de Pesquisa em Desenvolvimento de Fármacos, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (G.F.S.); (J.S.d.S.); (A.K.N.d.A.); (M.d.M.C.d.S.); (T.L.M.); (B.d.S.R.); (R.H.C.N.d.F.); or (R.T.S.)
- Programa de Pós-Graduação em Farmacologia e Química Medicinal, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil
| | - Carlos Alberto Manssour Fraga
- Programa de Pesquisa em Desenvolvimento de Fármacos, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (G.F.S.); (J.S.d.S.); (A.K.N.d.A.); (M.d.M.C.d.S.); (T.L.M.); (B.d.S.R.); (R.H.C.N.d.F.); or (R.T.S.)
- Programa de Pós-Graduação em Farmacologia e Química Medicinal, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil
- Correspondence: (C.A.M.F.); or (G.Z.-S.); Tel./Fax: +55-21-39386478 (C.A.M.F.); +55-21-39386505 (G.Z.-S.)
| | - Gisele Zapata-Sudo
- Programa de Pesquisa em Desenvolvimento de Fármacos, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (G.F.S.); (J.S.d.S.); (A.K.N.d.A.); (M.d.M.C.d.S.); (T.L.M.); (B.d.S.R.); (R.H.C.N.d.F.); or (R.T.S.)
- Programa de Pós-Graduação em Cardiologia, Instituto do Coração Edson Saad, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-913, RJ, Brazil
- Programa de Pós-Graduação em Farmacologia e Química Medicinal, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil
- Correspondence: (C.A.M.F.); or (G.Z.-S.); Tel./Fax: +55-21-39386478 (C.A.M.F.); +55-21-39386505 (G.Z.-S.)
| |
Collapse
|
41
|
Cip1 tunes cell cycle arrest duration upon calcineurin activation. Proc Natl Acad Sci U S A 2022; 119:e2202469119. [PMID: 35653562 DOI: 10.1073/pnas.2202469119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
SignificanceTo ensure their survival, cells arrest the cell division cycle when they are exposed to environmental stress. The duration of this arrest is dependent upon the time it takes a cell to adapt to a particular environment. How cells adjust the amount of time they remain arrested is not known. This study investigates the role of the phosphatase calcineurin in controlling cell cycle arrest duration in yeast. We show that calcineurin lengthens arrest by prolonging Hog1-dependent activation of the poorly characterized cyclin-dependent kinase inhibitor Cip1. Cip1 only impacts cell cycle arrest in response to stressors that robustly activate calcineurin, suggesting that Cip1 is a context-specific regulator that differentially adjusts the length of arrest depending on the particular stressor.
Collapse
|