1
|
Yan X, Wang M, Ji L, Li X, Gao B. Machine learning and molecular subtyping reveal the impact of diverse patterns of cell death on the prognosis and treatment of hepatocellular carcinoma. Comput Biol Chem 2025; 115:108360. [PMID: 39874853 DOI: 10.1016/j.compbiolchem.2025.108360] [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: 09/23/2024] [Revised: 12/27/2024] [Accepted: 01/19/2025] [Indexed: 01/30/2025]
Abstract
Programmed cell death (PCD) is a significant factor in the progression of hepatocellular carcinoma (HCC) and might serve as a crucial marker for predicting HCC prognosis and therapy response. However, the classification of HCC based on diverse PCD patterns requires further investigation. This study identified a novel molecular classification named PCD subtype (C1, C2, and C3) based on the genes associated with 19 PCD patterns, distinguished by clinical, biological functional pathways, mutations, immune characteristics, and drug sensitivity. Validated in 4 independent datasets, diverse cell death pathways were enriched in the C3 subtype, including apoptosis, pyroptosis, and autophagy, it also exhibited a highly infiltrative immunosuppressive microenvironment and demonstrated higher sensitivity to compounds such as Paclitaxel, Bortezomib, and YK-4-279, while C1 subtype was significantly enriched in cuproptosis and metabolism-related pathways, suggesting that it may be more suitable for cuproptosis-inducing agent therapy. Subsequently, utilizing the machine learning algorithms, we constructed a cell death-related index (CDRI) with 22 gene features and constructed prognostic nomograms with high predictive performance by combining CDRI with clinical features. Notably, we found that CDRI effectively predicted the response of HCC patients to therapeutic strategies, where patients with high CDRI were more suitable for sorafenib drug therapy and patients with low CDRI were more ideal for transarterial chemoembolization (TACE). In conclusion, the PCD subtype and CDRI demonstrate significant efficacy in predicting the prognosis and therapeutic outcomes for patients with HCC. These findings offer valuable insights for the development of precise, individualized treatment strategies.
Collapse
Affiliation(s)
- Xinyue Yan
- College of Chemistry and Life Science of Beijing University of Technology, Beijing 100124, China
| | - Meng Wang
- College of Chemistry and Life Science of Beijing University of Technology, Beijing 100124, China
| | - Lurao Ji
- College of Chemistry and Life Science of Beijing University of Technology, Beijing 100124, China
| | - Xiaoqin Li
- College of Chemistry and Life Science of Beijing University of Technology, Beijing 100124, China.
| | - Bin Gao
- College of Chemistry and Life Science of Beijing University of Technology, Beijing 100124, China
| |
Collapse
|
2
|
Dos Santos Oliveira M, de C Griebeler M, Henz B, Dos Santos FF, Guardia GDA, Conceição HB, Galante PAF, Minussi DC, Oliveira MM, Lenz G. Population dynamics is a cancer driver. Carcinogenesis 2024; 45:893-902. [PMID: 38842162 DOI: 10.1093/carcin/bgae038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 05/16/2024] [Accepted: 06/04/2024] [Indexed: 06/07/2024] Open
Abstract
Most tissues are continuously renovated through the division of stem cells and the death of old or damaged cells, which is known as the cell turnover rate (CTOR). Despite being in a steady state, tissues have different population dynamics thus producing diverse clonality levels. Here, we propose and test that cell population dynamics can be a cancer driver. We employed the evolutionary software esiCancer to show that CTOR, within a range comparable to what is observed in human tissues, can amplify the risk of a mutation due to ancestral selection (ANSEL). In a high CTOR tissue, a mutated ancestral cell is likely to be selected and persist over generations, which leads to a scenario of elevated ANSEL profile, characterized by few niches of large clones, which does not occur in low CTOR. We found that CTOR is significantly associated with the risk of developing cancer, even when correcting for mutation load, indicating that population dynamics per se is a cancer driver. This concept is central to understanding cancer risk and for the design of new therapeutic interventions that minimizes the contribution of ANSEL in cancer growth.
Collapse
Affiliation(s)
- Mariana Dos Santos Oliveira
- Departamento de Biofísica, Universidade Federal do Rio Grande do Sul (UFRGS), Av. Bento Gonçalves, 9500, 91501-970, Porto Alegre, RS, Brazil
- Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul (UFRGS), Av. Bento Gonçalves, 9500, 91501-970, Porto Alegre, RS, Brazil
| | - Marcelo de C Griebeler
- Departamento de Economia e Relações Internacionais, Universidade Federal do Rio Grande do Sul (UFRGS), Av. João Pessoa, 52, 90040-000, Porto Alegre, RS, Brazil
| | - Bernardo Henz
- Instituto de Informática, Universidade Federal do Rio Grande do Sul (UFRGS), Av. Bento Gonçalves, 9500, 91501-970, Porto Alegre, RS, Brazil
- Instituto Federal Farroupilha, Campus Alegrete, Rodovia RS-377, s/n, 97555-000 Alegrete, RS, Brazil
| | - Filipe Ferreira Dos Santos
- Centro de Oncologia Molecular, Hospital Sirio-Libanes, Prof Daher Cutait, 69, 013080-60, São Paulo, SP, Brazil
- Departamento de Bioquimica, Instituto de Quimica, Universidade de São Paulo, Av. Prof. Lineu Prestes, 748, 05508-090, São Paulo, SP, Brazil
| | - Gabriela D A Guardia
- Centro de Oncologia Molecular, Hospital Sirio-Libanes, Prof Daher Cutait, 69, 013080-60, São Paulo, SP, Brazil
| | - Helena B Conceição
- Centro de Oncologia Molecular, Hospital Sirio-Libanes, Prof Daher Cutait, 69, 013080-60, São Paulo, SP, Brazil
- Interunidades em Bioinformática, Universidade de São Paulo, R. do Matão, 1010, 05508-090, São Paulo, SP, Brazil
| | - Pedro A F Galante
- Centro de Oncologia Molecular, Hospital Sirio-Libanes, Prof Daher Cutait, 69, 013080-60, São Paulo, SP, Brazil
- Departamento de Bioquimica, Instituto de Quimica, Universidade de São Paulo, Av. Prof. Lineu Prestes, 748, 05508-090, São Paulo, SP, Brazil
| | - Darlan C Minussi
- Departamento de Biofísica, Universidade Federal do Rio Grande do Sul (UFRGS), Av. Bento Gonçalves, 9500, 91501-970, Porto Alegre, RS, Brazil
- Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul (UFRGS), Av. Bento Gonçalves, 9500, 91501-970, Porto Alegre, RS, Brazil
| | - Manuel M Oliveira
- Instituto de Informática, Universidade Federal do Rio Grande do Sul (UFRGS), Av. Bento Gonçalves, 9500, 91501-970, Porto Alegre, RS, Brazil
| | - Guido Lenz
- Departamento de Biofísica, Universidade Federal do Rio Grande do Sul (UFRGS), Av. Bento Gonçalves, 9500, 91501-970, Porto Alegre, RS, Brazil
- Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul (UFRGS), Av. Bento Gonçalves, 9500, 91501-970, Porto Alegre, RS, Brazil
| |
Collapse
|
3
|
Leak L, Dixon SJ. Surveying the landscape of emerging and understudied cell death mechanisms. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2023; 1870:119432. [PMID: 36690038 PMCID: PMC9969746 DOI: 10.1016/j.bbamcr.2023.119432] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 01/09/2023] [Accepted: 01/15/2023] [Indexed: 01/22/2023]
Abstract
Cell death can be a highly regulated process. A large and growing number of mammalian cell death mechanisms have been described over the past few decades. Major pathways with established roles in normal or disease biology include apoptosis, necroptosis, pyroptosis and ferroptosis. However, additional non-apoptotic cell death mechanisms with unique morphological, genetic, and biochemical features have also been described. These mechanisms may play highly specialized physiological roles or only become activated in response to specific lethal stimuli or conditions. Understanding the nature of these emerging and understudied mechanisms may provide new insight into cell death biology and suggest new treatments for diseases such as cancer and neurodegeneration.
Collapse
Affiliation(s)
- Logan Leak
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Scott J Dixon
- Department of Biology, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
4
|
Canonical and Noncanonical ER Stress-Mediated Autophagy Is a Bite the Bullet in View of Cancer Therapy. Cells 2022; 11:cells11233773. [PMID: 36497032 PMCID: PMC9738281 DOI: 10.3390/cells11233773] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 11/20/2022] [Accepted: 11/22/2022] [Indexed: 11/29/2022] Open
Abstract
Cancer cells adapt multiple mechanisms to counter intense stress on their way to growth. Tumor microenvironment stress leads to canonical and noncanonical endoplasmic stress (ER) responses, which mediate autophagy and are engaged during proteotoxic challenges to clear unfolded or misfolded proteins and damaged organelles to mitigate stress. In these conditions, autophagy functions as a cytoprotective mechanism in which malignant tumor cells reuse degraded materials to generate energy under adverse growing conditions. However, cellular protection by autophagy is thought to be complicated, contentious, and context-dependent; the stress response to autophagy is suggested to support tumorigenesis and drug resistance, which must be adequately addressed. This review describes significant findings that suggest accelerated autophagy in cancer, a novel obstacle for anticancer therapy, and discusses the UPR components that have been suggested to be untreatable. Thus, addressing the UPR or noncanonical ER stress components is the most effective approach to suppressing cytoprotective autophagy for better and more effective cancer treatment.
Collapse
|
5
|
Poddar NK, Agarwal D, Agrawal Y, Wijayasinghe YS, Mukherjee A, Khan S. Deciphering the enigmatic crosstalk between prostate cancer and Alzheimer's disease: A current update on molecular mechanisms and combination therapy. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166524. [PMID: 35985445 DOI: 10.1016/j.bbadis.2022.166524] [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: 06/02/2022] [Revised: 08/05/2022] [Accepted: 08/11/2022] [Indexed: 11/26/2022]
Abstract
Alzheimer's disease (AD) and prostate cancer (PCa) are considered the leading causes of death in elderly people worldwide. Although both these diseases have striking differences in their pathologies, a few underlying mechanisms are similar when cell survival is considered. In the current study, we employed an in-silico approach to decipher the possible role of bacterial proteins in the initiation and progression of AD and PCa. We further analyzed the molecular connections between these two life-threatening diseases. The androgen deprivation therapy used against PCa has been shown to promote castrate resistant PCa as well as AD. In addition, cell signaling pathways, such as Akt, IGF, and Wnt contribute to the progression of both AD and PCa. Besides, various proteins and genes are also common in disease progression. One such similarity is mTOR signaling. mTOR is the common downstream target for many signaling pathways and plays a vital role in both PCa and AD. Targeting mTOR can be a favorable line of treatment for both AD and PCa. However, drug resistance is one of the challenges in effective drug therapy. A few drugs that target mTOR have now become ineffective due to the development of resistance. In that regard, phytochemicals can be a rich source of novel drug candidates as they can act via multiple mechanisms. This review also presents mTOR targeting phytochemicals with promising anti-PCa, anti-AD activities, and approaches to overcome the issues associated with phytochemical-based therapies in clinical trials.
Collapse
Affiliation(s)
- Nitesh Kumar Poddar
- Department of Biosciences, Manipal University Jaipur, Dehmi Kalan, Jaipur-Ajmer Expressway, Jaipur, Rajasthan 303007, India.
| | - Disha Agarwal
- Department of Biosciences, Manipal University Jaipur, Dehmi Kalan, Jaipur-Ajmer Expressway, Jaipur, Rajasthan 303007, India
| | - Yamini Agrawal
- Department of Biosciences, Manipal University Jaipur, Dehmi Kalan, Jaipur-Ajmer Expressway, Jaipur, Rajasthan 303007, India
| | | | - Arunima Mukherjee
- Department of Biosciences, Manipal University Jaipur, Dehmi Kalan, Jaipur-Ajmer Expressway, Jaipur, Rajasthan 303007, India
| | - Shahanavaj Khan
- Department of Health Sciences, Novel Global Community Educational Foundation, NSW, Australia; Department of Pharmaceutics, College of Pharmacy, PO Box 2457, King Saud University, Riyadh 11451, Saudi Arabia; Department of Medical Lab Technology, Indian Institute of health and Technology (IIHT), Deoband, 247554 Saharanpur, UP, India.
| |
Collapse
|
6
|
Sung JY, Cheong JH. Pan-Cancer Analysis of Clinical Relevance via Telomere Maintenance Mechanism. Int J Mol Sci 2021; 22:ijms222011101. [PMID: 34681758 PMCID: PMC8538844 DOI: 10.3390/ijms222011101] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 10/04/2021] [Accepted: 10/13/2021] [Indexed: 12/24/2022] Open
Abstract
Understanding the telomere maintenance mechanism (TMM) in immortal cancer cells is vital for TMM-targeted therapies in clinical settings. In this study, we classified four telomere maintenance mechanisms into telomerase, ALT, telomerase + ALT, and non-defined telomere maintenance mechanism (NDTMM) across 31 cancer types using 10,704 transcriptomic datasets from The Cancer Genome Atlas. Our results demonstrated that approximately 50% of the total cohort displayed ALT activity with high telomerase activity in most cancer types. We confirmed significant patient prognoses according to distinct TMMs in six cancer types: adrenocortical carcinoma (ACC), PAAD, HNSC, SARC, GBM, and metastatic cancer. Patients with metastasis had a poor prognosis in the ALT group (p < 0.006) subjected to RAS protein signal transduction. Glioblastoma patients had poor prognosis in NDTMM (p < 0.0043) and showed high levels of myeloid leukocyte activation. Pancreatic adenocarcinoma (p < 0.04) and head and neck squamous cell carcinoma (p < 0.046) patients had a good prognosis in the ALT group with high immune cell activation. Furthermore, we showed that master transcriptional regulators might affect the selection of the TMM pathway and explained why different telomere maintenance mechanisms exist. Furthermore, they can be used to segregate patients and predict responders to different TMM-targeted therapeutics.
Collapse
Affiliation(s)
- Ji-Yong Sung
- Department of Biomedical Systems Informatics, Yonsei University College of Medicine, Seoul 03722, Korea;
- Department of Laboratory Medicine, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Jae-Ho Cheong
- Department of Biomedical Systems Informatics, Yonsei University College of Medicine, Seoul 03722, Korea;
- Department of Surgery, Yonsei University College of Medicine, Seoul 03722, Korea
- Yonsei Biomedical Research Institute, Yonsei University College of Medicine, Seoul 03722, Korea
- Department of Biochemistry & Molecular Biology, Yonsei University College of Medicine, Seoul 03722, Korea
- Department of Research & Development, VeraVerse Inc., Seoul 03722, Korea
- Correspondence:
| |
Collapse
|
7
|
Proteome profile of telencephalon associates attenuated neurogenesis with chronic stress induced mood disorder phenotypes in zebrafish model. Pharmacol Biochem Behav 2021; 204:173170. [DOI: 10.1016/j.pbb.2021.173170] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 02/28/2021] [Accepted: 02/28/2021] [Indexed: 02/07/2023]
|
8
|
Li S, Li P, Liu W, Shang J, Qiu S, Li X, Liu W, Shi H, Zhou M, Liu H. Danhong Injection Alleviates Cardiac Fibrosis via Preventing the Hypermethylation of Rasal1 and Rassf1 in TAC Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:3158108. [PMID: 33456666 PMCID: PMC7787771 DOI: 10.1155/2020/3158108] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 12/02/2020] [Accepted: 12/06/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND/AIM Danhong injection (DHI) is a Chinese patent drug used for relieving cardiovascular diseases. Recent studies have suggested that DNA methylation plays a pivotal role in the maintenance of cardiac fibrosis (CF) in cardiovascular diseases. This study was aimed at identifying the effect and the underlying mechanism of DHI on CF, especially the DNA methylation. METHODS A CF murine model was established by thoracic aortic constriction (TAC). A 28-day daily treatment with or without DHI via intraperitoneal injection was carried out immediately following TAC surgery. The changes in cardiac function, pathology, and fibrosis following TAC were measured by echocardiography and immunostaining. We used methyl-seq analysis to assess the DNA methylation changes in whole genes and identified the methylation changes of two Ras signaling-related genes in TAC mice, including Ras protein activator like-1 (Rasal1) and Ras-association domain family 1 (Rassf1). Next, the methylation status and expression levels of Rasal1 and Rassf1 genes were consolidated by bisulfite sequencing, quantitative reverse transcription polymerase chain reaction (RT-qPCR), and Western blotting, respectively. To determine the underlying molecular mechanism, the expressions of DNA methyltransferases (DNMTs), Tet methylcytosine dioxygenase 3 (TET3), fibrosis-related genes, and the activity of Ras/ERK were measured by RT-qPCR and Western blotting. RESULTS DHI treatment alleviated CF and significantly improved cardiac function on day 28 of TAC. The methyl-seq analysis identified 42,606 differential methylated sites (DMSs), including 19,618 hypermethylated DMSs and 22,988 hypomethylated DMSs between TAC and sham-operated mice. The enrichment analysis of these DMSs suggested that the methylated regulation of Ras signal transduction and focal adhesion-related genes would be involved in the TAC-induced CF development. The results of bisulfite sequencing revealed that the TAC-induced methylation affected the CpG site in both of Rasal1 and Rassf1 genes, and DHI treatment remarkably downregulated the promoter methylation of Rasal1 and Rassf1 in CF hearts. Furthermore, DHI treatment upregulated the expressions of Rasal1 and Rassf1, inhibited the hyperactivity of Ras/ERK, and decreased the expressions of fibrosis-related genes. Notably, we found that DHI treatment markedly downregulated the expression of DNMT3B in CF hearts, while it did not affect the expressions of DNMT1, DNMT3A, and TET3. CONCLUSION Aberrant DNA methylation of Rasal1 and Rassf1 genes was involved in the CF development. DHI treatment alleviated CF, prevented the hypermethylation of Rasal1 and Rassf1, and downregulated DNMT3B expression in CF hearts.
Collapse
Affiliation(s)
- Sinai Li
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China
- Beijing Institute of Traditional Chinese Medicine, Beijing 100010, China
| | - Ping Li
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China
- Beijing Institute of Traditional Chinese Medicine, Beijing 100010, China
| | - Weihong Liu
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China
- Beijing Institute of Traditional Chinese Medicine, Beijing 100010, China
| | - Juju Shang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China
| | - Shenglei Qiu
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China
| | - Xiang Li
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China
| | - Wei Liu
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China
| | - Haoyue Shi
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China
| | - Mingxue Zhou
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China
- Beijing Institute of Traditional Chinese Medicine, Beijing 100010, China
| | - Hongxu Liu
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China
| |
Collapse
|
9
|
Liang Q, Ou M, Li Z, Ren Y, Wei W, Qiao X, Hu R, Wu X, Liu Y, Wang W. Functional analysis target of rapamycin (TOR) on the Penaeus vannamei in response to acute low temperature stress. FISH & SHELLFISH IMMUNOLOGY 2020; 96:53-61. [PMID: 31801694 DOI: 10.1016/j.fsi.2019.11.070] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 11/18/2019] [Accepted: 11/30/2019] [Indexed: 06/10/2023]
Abstract
Target of rapamycin (TOR) is an atypical of Ser/Thr protein kinase that plays an important role in many aspects such as cell growth, reproduction, differentiation, cell cycle regulation, autophagy and apoptosis. However, little information is known about the enzyme in crustaceans. Here, a novel TOR was identified from shrimp Penaeus vannamei (PvTOR) and its biological functions were investigated in response low temperature stress. The PvTOR gene encoded a polypeptide of 2464 amino acids with an estimated molecular mass of 279.4 kD and a predicted isoelectronic point (pI) of 7.30. Phylogenetic analysis revealed that PvTOR shared high similarity with other known species. PvTOR mRNA was detected in all the tested tissues and highest transcription in muscle and hepatopancreas. PvTOR transcriptional level was up-regulated significantly at 1.5 h and 3 h, and down-regulated at 12 h and 24 h after low temperature stress. TEM and autophagy indicator system GFP-PvLC3 suggested that low temperature induced autophagy generation. ROS, Ca2+ concentration and apoptosis rate were increased significantly in TOR-knockdown shrimp after low temperature stress. The autophagy associated gene ATG8II/I, PvBeclin-1, PvATG14, apoptosis gene PvPARP, Pvcasp-3, PvBAX and Pvp53 transcripts, and casp-3/8 activity in hemocyte were increased significantly in TOR-knockdown group shrimp at 3 h after low temperature stress. Additionally, THC counts of TOR-knockdown group were significantly higher than the dsGFP group. In summary, these results suggested that PvTOR plays an important role in the adaptation mechanisms of shrimp at low temperature by regulating autophagy and apoptosis.
Collapse
Affiliation(s)
- QingJian Liang
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Key Laboratory of Ecology and Environmental Science in Guangdong Higher Education, College of Life Science, South China Normal University, Guangzhou, 510631, PR China
| | - Mufei Ou
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Key Laboratory of Ecology and Environmental Science in Guangdong Higher Education, College of Life Science, South China Normal University, Guangzhou, 510631, PR China
| | - ZhongHua Li
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Key Laboratory of Ecology and Environmental Science in Guangdong Higher Education, College of Life Science, South China Normal University, Guangzhou, 510631, PR China
| | - YingHao Ren
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Key Laboratory of Ecology and Environmental Science in Guangdong Higher Education, College of Life Science, South China Normal University, Guangzhou, 510631, PR China
| | - Wei Wei
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Key Laboratory of Ecology and Environmental Science in Guangdong Higher Education, College of Life Science, South China Normal University, Guangzhou, 510631, PR China
| | - Xueli Qiao
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Key Laboratory of Ecology and Environmental Science in Guangdong Higher Education, College of Life Science, South China Normal University, Guangzhou, 510631, PR China
| | - Rui Hu
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Key Laboratory of Ecology and Environmental Science in Guangdong Higher Education, College of Life Science, South China Normal University, Guangzhou, 510631, PR China
| | - XuJian Wu
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Key Laboratory of Ecology and Environmental Science in Guangdong Higher Education, College of Life Science, South China Normal University, Guangzhou, 510631, PR China
| | - Yuan Liu
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Key Laboratory of Ecology and Environmental Science in Guangdong Higher Education, College of Life Science, South China Normal University, Guangzhou, 510631, PR China
| | - Weina Wang
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Key Laboratory of Ecology and Environmental Science in Guangdong Higher Education, College of Life Science, South China Normal University, Guangzhou, 510631, PR China.
| |
Collapse
|
10
|
Wallner C, Drysch M, Hahn SA, Becerikli M, Puscz F, Wagner JM, Sacher M, Sogorski A, Dadras M, Lehnhardt M, Behr B. Alterations in pectoralis muscle cell characteristics after radiation of the human breast in situ. JOURNAL OF RADIATION RESEARCH 2019; 60:825-830. [PMID: 31665379 PMCID: PMC7357228 DOI: 10.1093/jrr/rrz067] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 08/09/2019] [Accepted: 08/26/2019] [Indexed: 06/10/2023]
Abstract
The life-time risk of being diagnosed with breast cancer is ~12%, hence breast cancer is by far the most common cancer among women. The multimodal treatment concept of breast cancer often intends radiation. The utilized ionizing radiation leads changes in the tissue resulting in tissue damage due to an alteration of molecular factors. The goal of this study was to identify the role of muscle-catabolic proteins after radiation of human pectoralis major muscles in situ. Tissue of the pectoralis major muscle was collected in 12 breast cancer patients after radiation (maximum 3 years after radiation) undergoing a deep inferior epigastric perforator free-flap breast reconstruction. At the same time, an intraindividual comparison to rectus abdominis muscle was carried out upon free-flap elevation. Immunological properties, cell proliferation, differentiation as well as the expression profile of the muscle tissue were investigated through immunohistological reactions, a DNA-microarray and histology. We found significantly increased neutrophil immigration in the radiated muscle tissue. At the same time, proteins responsible for muscular atrophy and apoptosis were significantly elevated in immunohistochemistry. A DNA microarray detected immunological upregulation and myo-differentiative disorders in radiated muscle tissue. This novel study investigating catabolism in radiated muscle in situ can serve as a basis for the treatment of radiation-accompanied muscle disorders.
Collapse
Affiliation(s)
- Christoph Wallner
- Department of Plastic Surgery, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bürkle-de-la-Camp Platz 1, 44789 Bochum, Germany
| | - Marius Drysch
- Department of Plastic Surgery, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bürkle-de-la-Camp Platz 1, 44789 Bochum, Germany
| | - Stephan A Hahn
- Department of Molecular Gastrointestinal Oncology, Ruhr-University Bochum, Universitätsstraße 150, 44780 Bochum, Germany
| | - Mustafa Becerikli
- Department of Plastic Surgery, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bürkle-de-la-Camp Platz 1, 44789 Bochum, Germany
| | - Fleming Puscz
- Department of Plastic Surgery, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bürkle-de-la-Camp Platz 1, 44789 Bochum, Germany
| | - Johannes Maximilian Wagner
- Department of Plastic Surgery, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bürkle-de-la-Camp Platz 1, 44789 Bochum, Germany
| | - Maxi Sacher
- Department of Plastic Surgery, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bürkle-de-la-Camp Platz 1, 44789 Bochum, Germany
| | - Alexander Sogorski
- Department of Plastic Surgery, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bürkle-de-la-Camp Platz 1, 44789 Bochum, Germany
| | - Mehran Dadras
- Department of Plastic Surgery, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bürkle-de-la-Camp Platz 1, 44789 Bochum, Germany
| | - Marcus Lehnhardt
- Department of Plastic Surgery, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bürkle-de-la-Camp Platz 1, 44789 Bochum, Germany
| | - Björn Behr
- Department of Plastic Surgery, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bürkle-de-la-Camp Platz 1, 44789 Bochum, Germany
| |
Collapse
|
11
|
Feng X, Zhang L, Nie S, Zhuang L, Wang W, Huang J, Yan X, Meng F. The Effect of Ras Homolog C/Rho-Associated Coiled-Protein Kinase (Rho/ROCK) Signaling Pathways on Proliferation and Apoptosis of Human Myeloma Cells. Med Sci Monit 2019; 25:7605-7616. [PMID: 31599230 PMCID: PMC6798802 DOI: 10.12659/msm.915998] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND The aim of this study was to explore the impact of Ras homolog C/Rho-associated coiled-protein kinase (Rho/ROCK) signaling pathways intervention on biological characteristics of the human multiple myeloma cell lines RPMI-8226 and U266 cells, and to investigate the expression of RhoC, ROCK1, and ROCK2 in RPMI-8226 and U266 cells. MATERIAL AND METHODS RPMI8226 and U266 cell lines were treated by 5-aza-2-deoxycytidine (5-Aza-Dc), trichostatin A (TSA), RhoA inhibitor CCG-1423, Rac1 inhibitor NSC23766, and ROCK inhibitor fasudil. Cell proliferation was examined by Cell Counting Kit-8 (CCK-8) assay and clone formation. Cell apoptosis was examined by flow cytometry and TUNEL assay. The mRNA and protein expressions of RhoC, ROCK1, and ROCK2 were detected by quantitative reverse transcription-polymerase chain reaction (qRT-PCR) and western blot, respectively. RESULTS CCG-1423, NSC23766, and fasudil could significantly inhibit the proliferation of RPMI8226 and U266 cells. The inhibitory effect was dose- and time-dependent within a certain concentration range (P<0.05). After treatment with CCG-1423, NSC23766, and fasudil for 24 hours, the apoptosis rates of RPMI8226 and U266 cells were significantly higher than those of the control group, which were dose-dependent (P<0.05). Compared with the control group, the mRNA and protein expressions of RhoC, ROCK1, and ROCK2 in RPMI8226 and U266 cells were significantly decreased with single 5-Aza-Dc or TSA treatment. However, the effects were obviously stronger after combined treatment of 5-Aza-CdR and TSA (P<0.05). CONCLUSIONS We found that 5-Aza-Dc and TSA can effectively decrease the mRNA and protein expressions of RhoC, ROCK1, and ROCK2. Furthermore, Rho and ROCK inhibitors significantly inhibit cell growth and induce cell apoptosis in the human multiple myeloma cell lines RPMI-8226 and U266.
Collapse
Affiliation(s)
- Xianqi Feng
- Department of Hematology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China (mainland)
| | - Ling Zhang
- Department of Pediatrics, Laiwu People's Hospital, Laiwu, Shandong, China (mainland)
| | - Shumin Nie
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China (mainland)
| | - Likun Zhuang
- Central Laboratory, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China (mainland)
| | - Wei Wang
- Department of Hematology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China (mainland)
| | - Junxia Huang
- Department of Hematology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China (mainland)
| | - Xueshen Yan
- Department of Hematology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China (mainland)
| | - Fanjun Meng
- Department of Hematology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China (mainland)
| |
Collapse
|
12
|
Perspectives of RAS and RHEB GTPase Signaling Pathways in Regenerating Brain Neurons. Int J Mol Sci 2018; 19:ijms19124052. [PMID: 30558189 PMCID: PMC6321366 DOI: 10.3390/ijms19124052] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 12/05/2018] [Accepted: 12/13/2018] [Indexed: 12/29/2022] Open
Abstract
Cellular activation of RAS GTPases into the GTP-binding “ON” state is a key switch for regulating brain functions. Molecular protein structural elements of rat sarcoma (RAS) and RAS homolog protein enriched in brain (RHEB) GTPases involved in this switch are discussed including their subcellular membrane localization for triggering specific signaling pathways resulting in regulation of synaptic connectivity, axonal growth, differentiation, migration, cytoskeletal dynamics, neural protection, and apoptosis. A beneficial role of neuronal H-RAS activity is suggested from cellular and animal models of neurodegenerative diseases. Recent experiments on optogenetic regulation offer insights into the spatiotemporal aspects controlling RAS/mitogen activated protein kinase (MAPK) or phosphoinositide-3 kinase (PI3K) pathways. As optogenetic manipulation of cellular signaling in deep brain regions critically requires penetration of light through large distances of absorbing tissue, we discuss magnetic guidance of re-growing axons as a complementary approach. In Parkinson’s disease, dopaminergic neuronal cell bodies degenerate in the substantia nigra. Current human trials of stem cell-derived dopaminergic neurons must take into account the inability of neuronal axons navigating over a large distance from the grafted site into striatal target regions. Grafting dopaminergic precursor neurons directly into the degenerating substantia nigra is discussed as a novel concept aiming to guide axonal growth by activating GTPase signaling through protein-functionalized intracellular magnetic nanoparticles responding to external magnets.
Collapse
|
13
|
Kumaraswamy A, Mamidi A, Desai P, Sivagnanam A, Perumalsamy LR, Ramakrishnan C, Gromiha M, Rajalingam K, Mahalingam S. The non-enzymatic RAS effector RASSF7 inhibits oncogenic c-Myc function. J Biol Chem 2018; 293:15691-15705. [PMID: 30139745 DOI: 10.1074/jbc.ra118.004452] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 08/12/2018] [Indexed: 11/06/2022] Open
Abstract
c-Myc is a proto-oncogene controlling expression of multiple genes involved in cell growth and differentiation. Although the functional role of c-Myc as a transcriptional regulator has been intensively studied, targeting this protein in cancer remains a challenge. Here, we report a trimodal regulation of c-Myc function by the Ras effector, Ras-association domain family member 7 (RASSF7), a nonenzymatic protein modulating protein-protein interactions to regulate cell proliferation. Using HEK293T and HeLa cell lines, we provide evidence that RASSF7 destabilizes the c-Myc protein by promoting Cullin4B-mediated polyubiquitination and degradation. Furthermore, RASSF7 competed with MYC-associated factor X (MAX) in the formation of a heterodimeric complex with c-Myc and attenuated its occupancy on target gene promoters to regulate transcription. Consequently, RASSF7 inhibited c-Myc-mediated oncogenic transformation, and an inverse correlation between the expression levels of the RASSF7 and c-Myc genes was evident in human cancers. Furthermore, we found that RASSF7 interacts with c-Myc via its RA and leucine zipper (LZ) domains and LZ domain peptide is sufficient to inhibit c-Myc function, suggesting that this peptide might be used to target oncogenic c-Myc. These results unveil that RASSF7 and c-Myc are functionally linked in the control of tumorigenesis and open up potential therapeutic avenues for targeting the "undruggable" c-Myc protein in a subset of human cancers.
Collapse
Affiliation(s)
- Anbarasu Kumaraswamy
- From the National Cancer Tissue Biobank, Laboratory of Molecular Cell Biology and
| | - Anitha Mamidi
- From the National Cancer Tissue Biobank, Laboratory of Molecular Cell Biology and
| | - Pavitra Desai
- From the National Cancer Tissue Biobank, Laboratory of Molecular Cell Biology and
| | - Ananthi Sivagnanam
- From the National Cancer Tissue Biobank, Laboratory of Molecular Cell Biology and
| | | | - Chandrasekaran Ramakrishnan
- Protein Bioinformatics Laboratory, Bhupat and Jyoti Mehta School of Biosciences, Department of Biotechnology, Indian Institute of Technology-Madras, Chennai 600036, India and
| | - Michael Gromiha
- Protein Bioinformatics Laboratory, Bhupat and Jyoti Mehta School of Biosciences, Department of Biotechnology, Indian Institute of Technology-Madras, Chennai 600036, India and
| | - Krishnaraj Rajalingam
- the MSU-FZI, Institute of Immunology, University Medical Center Mainz, JGU, 55131 Mainz, Germany
| | | |
Collapse
|
14
|
Schöpel M, Shkura O, Seidel J, Kock K, Zhong X, Löffek S, Helfrich I, Bachmann HS, Scherkenbeck J, Herrmann C, Stoll R. Allosteric Activation of GDP-Bound Ras Isoforms by Bisphenol Derivative Plasticisers. Int J Mol Sci 2018; 19:ijms19041133. [PMID: 29642594 PMCID: PMC5979466 DOI: 10.3390/ijms19041133] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 04/04/2018] [Accepted: 04/05/2018] [Indexed: 01/13/2023] Open
Abstract
The protein family of small GTPases controls cellular processes by acting as a binary switch between an active and an inactive state. The most prominent family members are H-Ras, N-Ras, and K-Ras isoforms, which are highly related and frequently mutated in cancer. Bisphenols are widespread in modern life because of their industrial application as plasticisers. Bisphenol A (BPA) is the best-known member and has gained significant scientific as well as public attention as an endocrine disrupting chemical, a fact that eventually led to its replacement. However, compounds used to replace BPA still contain the molecular scaffold of bisphenols. BPA, BPAF, BPB, BPE, BPF, and an amine-substituted BPAF-derivate all interact with all GDP-bound Ras-Isoforms through binding to a common site on these proteins. NMR-, SOScat-, and GDI- assay-based data revealed a new bisphenol-induced, allosterically activated GDP-bound Ras conformation that define these plasticisers as Ras allosteric agonists.
Collapse
Affiliation(s)
- Miriam Schöpel
- Faculty of Chemistry and Biochemistry, Ruhr University of Bochum, Universitätsstr. 150, D-44780 Bochum, Germany.
| | - Oleksandr Shkura
- Faculty of Chemistry and Biochemistry, Ruhr University of Bochum, Universitätsstr. 150, D-44780 Bochum, Germany.
| | - Jana Seidel
- Faculty of Chemistry and Biochemistry, Ruhr University of Bochum, Universitätsstr. 150, D-44780 Bochum, Germany.
| | - Klaus Kock
- Faculty of Chemistry and Biochemistry, Ruhr University of Bochum, Universitätsstr. 150, D-44780 Bochum, Germany.
| | - Xueyin Zhong
- Faculty of Chemistry and Biochemistry, Ruhr University of Bochum, Universitätsstr. 150, D-44780 Bochum, Germany.
| | - Stefanie Löffek
- Skin Cancer Unit of the Dermatology Department, West German Cancer Center, University Hospital Essen, University Duisburg-Essen and the German Cancer Consortium (DKTK), D-45147 Essen, Germany.
| | - Iris Helfrich
- Skin Cancer Unit of the Dermatology Department, West German Cancer Center, University Hospital Essen, University Duisburg-Essen and the German Cancer Consortium (DKTK), D-45147 Essen, Germany.
| | - Hagen S Bachmann
- Institute of Pharmacology and Toxicology, Witten/Herdecke University, Stockumer Str. 10, D-58453 Witten, Germany.
| | - Jürgen Scherkenbeck
- Faculty of Mathematics and Natural Sciences, University of Wuppertal, Gaußstr. 20, D-42119 Wuppertal, Germany.
| | - Christian Herrmann
- Faculty of Chemistry and Biochemistry, Ruhr University of Bochum, Universitätsstr. 150, D-44780 Bochum, Germany.
| | - Raphael Stoll
- Faculty of Chemistry and Biochemistry, Ruhr University of Bochum, Universitätsstr. 150, D-44780 Bochum, Germany.
| |
Collapse
|
15
|
Serdar M, Herz J, Kempe K, Winterhager E, Jastrow H, Heumann R, Felderhoff-Müser U, Bendix I. Protection of Oligodendrocytes Through Neuronal Overexpression of the Small GTPase Ras in Hyperoxia-Induced Neonatal Brain Injury. Front Neurol 2018; 9:175. [PMID: 29619004 PMCID: PMC5871665 DOI: 10.3389/fneur.2018.00175] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 03/06/2018] [Indexed: 12/29/2022] Open
Abstract
Prematurely born infants are highly susceptible to various environmental factors, such as inflammation, drug exposure, and also high environmental oxygen concentrations. Hyperoxia induces perinatal brain injury affecting white and gray matter development. It is well known that mitogen-activated protein kinase signaling is involved in cell survival, proliferation, and differentiation. Therefore, we aim to elucidate cell-specific responses of neuronal overexpression of the small GTPase Ras on hyperoxia-mediated brain injury. Six-day-old (P6) synRas mice (neuronal Ras overexpression under the synapsin promoter) or wild-type littermates were kept under hyperoxia (80% oxygen) or room air (21% oxygen) for 24 h. Apoptosis was analyzed by Western blot of cleaved Caspase-3 and neuronal and oligodendrocyte degeneration via immunohistochemistry. Short-term differentiation capacity of oligodendrocytes was assessed by quantification of myelin basic protein expression at P11. Long-lasting changes of hyperoxia-induced alteration of myelin structures were evaluated via transmission electron microscopy in young adult animals (P42). Western blot analysis of active Caspase-3 demonstrates a significant upregulation in wild-type littermates exposed to hyperoxia whereas synRas mice did not show any marked alteration of cleaved Caspase-3 protein levels. Immunohistochemistry revealed a protective effect of neuronal Ras overexpression on neuron and oligodendrocyte survival. Hyperoxia-induced hypomyelination in wild-type littermates was restored in synRas mice. These short-term protective effects through promotion of neuronal survival translated into long-lasting improvement of ultrastructural alterations of myelin sheaths in mice with neuronal overexpression of Ras compared with hyperoxic wild-type mice. Our data suggest that transgenic increase of neuronal Ras activity in the immature brain results in secondary protection of oligodendrocytes from hyperoxia-induced white matter brain injury.
Collapse
Affiliation(s)
- Meray Serdar
- Department of Pediatrics I, Neonatology, University Hospital, University Duisburg-Essen, Essen, Germany
| | - Josephine Herz
- Department of Pediatrics I, Neonatology, University Hospital, University Duisburg-Essen, Essen, Germany
| | - Karina Kempe
- Department of Pediatrics I, Neonatology, University Hospital, University Duisburg-Essen, Essen, Germany
| | - Elke Winterhager
- Imaging Center Essen, EM Unit, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Holger Jastrow
- Imaging Center Essen, EM Unit, University Hospital Essen, University Duisburg-Essen, Essen, Germany.,Institute of Anatomy, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Rolf Heumann
- Biochemistry II, Molecular Neurobiochemistry, Faculty of Chemistry and Biochemistry, Ruhr University Bochum, Bochum, Germany
| | - Ursula Felderhoff-Müser
- Department of Pediatrics I, Neonatology, University Hospital, University Duisburg-Essen, Essen, Germany
| | - Ivo Bendix
- Department of Pediatrics I, Neonatology, University Hospital, University Duisburg-Essen, Essen, Germany
| |
Collapse
|
16
|
Nakhaei-Rad S, Haghighi F, Nouri P, Rezaei Adariani S, Lissy J, Kazemein Jasemi NS, Dvorsky R, Ahmadian MR. Structural fingerprints, interactions, and signaling networks of RAS family proteins beyond RAS isoforms. Crit Rev Biochem Mol Biol 2018; 53:130-156. [PMID: 29457927 DOI: 10.1080/10409238.2018.1431605] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Saeideh Nakhaei-Rad
- a Institute of Biochemistry and Molecular Biology II, Medical Faculty , Heinrich-Heine University , Düsseldorf , Germany
| | - Fereshteh Haghighi
- a Institute of Biochemistry and Molecular Biology II, Medical Faculty , Heinrich-Heine University , Düsseldorf , Germany
| | - Parivash Nouri
- a Institute of Biochemistry and Molecular Biology II, Medical Faculty , Heinrich-Heine University , Düsseldorf , Germany
| | - Soheila Rezaei Adariani
- a Institute of Biochemistry and Molecular Biology II, Medical Faculty , Heinrich-Heine University , Düsseldorf , Germany
| | - Jana Lissy
- a Institute of Biochemistry and Molecular Biology II, Medical Faculty , Heinrich-Heine University , Düsseldorf , Germany
| | - Neda S Kazemein Jasemi
- a Institute of Biochemistry and Molecular Biology II, Medical Faculty , Heinrich-Heine University , Düsseldorf , Germany
| | - Radovan Dvorsky
- a Institute of Biochemistry and Molecular Biology II, Medical Faculty , Heinrich-Heine University , Düsseldorf , Germany
| | - Mohammad Reza Ahmadian
- a Institute of Biochemistry and Molecular Biology II, Medical Faculty , Heinrich-Heine University , Düsseldorf , Germany
| |
Collapse
|
17
|
Nelson N, Clark GJ. Rheb may complex with RASSF1A to coordinate Hippo and TOR signaling. Oncotarget 2017; 7:33821-31. [PMID: 27034171 PMCID: PMC5085121 DOI: 10.18632/oncotarget.8447] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 03/23/2016] [Indexed: 12/19/2022] Open
Abstract
The TOR pathway is a vital component of cellular homeostasis that controls the synthesis of proteins, nucleic acids and lipids. Its core is the TOR kinase. Activation of the TOR pathway suppresses autophagy, which plays a vital but complex role in tumorigenesis. The TOR pathway is regulated by activation of the Ras-related protein Rheb, which can bind mTOR. The Hippo pathway is a major growth control module that regulates cell growth, differentiation and apoptosis. Its core consists of an MST/LATS kinase cascade that can be activated by the RASSF1A tumor suppressor. The TOR and Hippo pathways may be coordinately regulated to promote cellular homeostasis. However, the links between the pathways remain only partially understood. We now demonstrate that in addition to mTOR regulation, Rheb also impacts the Hippo pathway by forming a complex with RASSF1A. Using stable clones of two human lung tumor cell lines (NCI-H1792 and NCI-H1299) with shRNA-mediated silencing or ectopic overexpression of RASSF1A, we show that activated Rheb stimulates the Hippo pathway, but is suppressed in its ability to stimulate the TOR pathway. Moreover, by selectively labeling autophagic vacuoles we show that RASSF1A inhibits the ability of Rheb to suppress autophagy and enhance cell growth. Thus, we identify a new connection that impacts coordination of Hippo and TOR signaling. As RASSF1A expression is frequently lost in human tumors, the RASSF1A status of a tumor may impact not just its Hippo pathway status, but also its TOR pathway status.
Collapse
Affiliation(s)
- Nicholas Nelson
- Department of Biochemistry and Molecular Biology, University of Louisville, Louisville, KY, USA.,Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, USA
| | - Geoffrey J Clark
- Department of Biochemistry and Molecular Biology, University of Louisville, Louisville, KY, USA.,Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, USA
| |
Collapse
|
18
|
Potheraveedu VN, Schöpel M, Stoll R, Heumann R. Rheb in neuronal degeneration, regeneration, and connectivity. Biol Chem 2017; 398:589-606. [PMID: 28212107 DOI: 10.1515/hsz-2016-0312] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2016] [Accepted: 02/02/2017] [Indexed: 01/31/2023]
Abstract
The small GTPase Rheb was originally detected as an immediate early response protein whose expression was induced by NMDA-dependent synaptic activity in the brain. Rheb's activity is highly regulated by its GTPase activating protein (GAP), the tuberous sclerosis complex protein, which stimulates the conversion from the active, GTP-loaded into the inactive, GDP-loaded conformation. Rheb has been established as an evolutionarily conserved molecular switch protein regulating cellular growth, cell volume, cell cycle, autophagy, and amino acid uptake. The subcellular localization of Rheb and its interacting proteins critically regulate its activity and function. In stem cells, constitutive activation of Rheb enhances differentiation at the expense of self-renewal partially explaining the adverse effects of deregulated Rheb in the mammalian brain. In the context of various cellular stress conditions such as oxidative stress, ER-stress, death factor signaling, and cellular aging, Rheb activation surprisingly enhances rather than prevents cellular degeneration. This review addresses cell type- and cell state-specific function(s) of Rheb and mainly focuses on neurons and their surrounding glial cells. Mechanisms will be discussed in the context of therapy that interferes with Rheb's activity using the antibiotic rapamycin or low molecular weight compounds.
Collapse
Affiliation(s)
- Veena Nambiar Potheraveedu
- Molecular Neurobiochemistry, Faculty of Chemistry and Biochemistry, Ruhr University of Bochum, Universitätstr. 150, D-44780 Bochum
| | - Miriam Schöpel
- Biomolecular NMR, Ruhr University of Bochum, D-44780 Bochum
| | - Raphael Stoll
- Biomolecular NMR, Ruhr University of Bochum, D-44780 Bochum
| | - Rolf Heumann
- Molecular Neurobiochemistry, Faculty of Chemistry and Biochemistry, Ruhr University of Bochum, Universitätstr. 150, D-44780 Bochum
| |
Collapse
|
19
|
Wang J, Yao X, Huang J. New tricks for human farnesyltransferase inhibitor: cancer and beyond. MEDCHEMCOMM 2017; 8:841-854. [PMID: 30108801 PMCID: PMC6072492 DOI: 10.1039/c7md00030h] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 02/15/2017] [Indexed: 12/18/2022]
Abstract
Human protein farnesyltransferase (FTase) catalyzes the addition of a C15-farnesyl lipid group to the cysteine residue located in the COOH-terminal tetrapeptide motif of a variety of important substrate proteins, including well-known Ras protein superfamily. The farnesylation of Ras protein is required both for its normal physiological function, and for the transforming capacity of its oncogenic mutants. Over the last several decades, FTase inhibitors (FTIs) were developed to disrupt the farnesylation of oncogenic Ras as anti-cancer agents, and some of them have entered cancer clinical investigation. On the other hand, some substrates of FTase were demonstrated to be related with other human diseases, including Hutchinson-Gilford progeria syndrome, chronic hepatitis D, and cardiovascular diseases. In this review, we summarize the roles of FTase in malignant transformation, proliferation, apoptosis, angiogenesis, and metastasis of tumor cells, and the recently anticancer clinical research advances of FTIs. The therapeutic prospect of FTIs on several other human diseases is also discussed.
Collapse
Affiliation(s)
- Jingyuan Wang
- Shanghai Key Laboratory of New Drug Design , School of Pharmacy , East China University of Science and Technology , 130 Mei Long Road , Shanghai 200237 , China . ; Tel: (+86)21 64253681
| | - Xue Yao
- Shanghai Key Laboratory of New Drug Design , School of Pharmacy , East China University of Science and Technology , 130 Mei Long Road , Shanghai 200237 , China . ; Tel: (+86)21 64253681
| | - Jin Huang
- Shanghai Key Laboratory of New Drug Design , School of Pharmacy , East China University of Science and Technology , 130 Mei Long Road , Shanghai 200237 , China . ; Tel: (+86)21 64253681
| |
Collapse
|
20
|
Schöpel M, Potheraveedu VN, Al-Harthy T, Abdel-Jalil R, Heumann R, Stoll R. The small GTPases Ras and Rheb studied by multidimensional NMR spectroscopy: structure and function. Biol Chem 2017; 398:577-588. [DOI: 10.1515/hsz-2016-0276] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 01/23/2017] [Indexed: 12/15/2022]
Abstract
Abstract
Ras GTPases are key players in cellular signalling because they act as binary switches. These states manifest through toggling between an active (GTP-loaded) and an inactive (GDP-loaded) form. The hydrolysis and replenishing of GTP is controlled by two additional protein classes: GAP (GTPase-activating)- and GEF (Guanine nucleotide exchange factors)-proteins. The complex interplay of the proteins is known as the GTPase-cycle. Several point mutations of the Ras protein deregulate this cycle. Mutations in Ras are associated with up to one-third of human cancers. The three isoforms of Ras (H, N, K) exhibit high sequence similarity and mainly differ in a region called HVR (hypervariable region). The HVR governs the differential action and cellular distribution of the three isoforms. Rheb is a Ras-like GTPase that is conserved from yeast to mammals. Rheb is mainly involved in activation of cell growth through stimulation of mTORC1 activity. In this review, we summarise multidimensional NMR studies on Rheb and Ras carried out to characterise their structure-function relationship and explain how the activity of these small GTPases can be modulated by low molecular weight compounds. These might help to design GTPase-selective antagonists for treatment of cancer and brain disease.
Collapse
|
21
|
Lamontagne J, Steel LF, Bouchard MJ. Hepatitis B virus and microRNAs: Complex interactions affecting hepatitis B virus replication and hepatitis B virus-associated diseases. World J Gastroenterol 2015; 21:7375-7399. [PMID: 26139985 PMCID: PMC4481434 DOI: 10.3748/wjg.v21.i24.7375] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Revised: 03/25/2015] [Accepted: 05/21/2015] [Indexed: 02/06/2023] Open
Abstract
Chronic infection with the hepatitis B virus (HBV) is the leading risk factor for the development of hepatocellular carcinoma (HCC). With nearly 750000 deaths yearly, hepatocellular carcinoma is the second highest cause of cancer-related death in the world. Unfortunately, the molecular mechanisms that contribute to the development of HBV-associated HCC remain incompletely understood. Recently, microRNAs (miRNAs), a family of small non-coding RNAs that play a role primarily in post-transcriptional gene regulation, have been recognized as important regulators of cellular homeostasis, and altered regulation of miRNA expression has been suggested to play a significant role in virus-associated diseases and the development of many cancers. With this in mind, many groups have begun to investigate the relationship between miRNAs and HBV replication and HBV-associated disease. Multiple findings suggest that some miRNAs, such as miR-122, and miR-125 and miR-199 family members, are playing a role in HBV replication and HBV-associated disease, including the development of HBV-associated HCC. In this review, we discuss the current state of our understanding of the relationship between HBV and miRNAs, including how HBV affects cellular miRNAs, how these miRNAs impact HBV replication, and the relationship between HBV-mediated miRNA regulation and HCC development. We also address the impact of challenges in studying HBV, such as the lack of an effective model system for infectivity and a reliance on transformed cell lines, on our understanding of the relationship between HBV and miRNAs, and propose potential applications of miRNA-related techniques that could enhance our understanding of the role miRNAs play in HBV replication and HBV-associated disease, ultimately leading to new therapeutic options and improved patient outcomes.
Collapse
MESH Headings
- Animals
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/therapy
- Carcinoma, Hepatocellular/virology
- Cell Transformation, Viral
- Gene Expression Regulation, Neoplastic
- Genetic Therapy
- Hepatitis B virus/genetics
- Hepatitis B virus/growth & development
- Hepatitis B virus/metabolism
- Hepatitis B, Chronic/complications
- Hepatitis B, Chronic/therapy
- Hepatitis B, Chronic/virology
- Host-Pathogen Interactions
- Humans
- Liver Neoplasms/genetics
- Liver Neoplasms/metabolism
- Liver Neoplasms/pathology
- Liver Neoplasms/therapy
- Liver Neoplasms/virology
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Virus Replication
Collapse
|
22
|
Gao JZ, DU JL, Wang YL, Li J, Wei LX, Guo MZ. Synergistic effects of curcumin and bevacizumab on cell signaling pathways in hepatocellular carcinoma. Oncol Lett 2014; 9:295-299. [PMID: 25435978 PMCID: PMC4246621 DOI: 10.3892/ol.2014.2694] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Accepted: 09/26/2014] [Indexed: 12/22/2022] Open
Abstract
The aim of the present study was to explore the effects of curcumin in combination with bevacizumab on the vascular endothelial growth factor (VEGF)/VEGF receptor (VEGFR)/K-ras pathway in hepatocellular carcinoma. A total of 30 Sprague Dawley (SD) rats were randomly divided into five groups: Control, model, curcumin, VEGF blocker, and curcumin + VEGF blocker groups. The mRNA levels of VEGF and VEGFR in all groups were subsequently measured by quantitative reverse transcriptase-polymerase chain reaction and the protein expression of K-ras was detected by western blot analysis. Compared with the control group, the mRNA levels of VEGF and VEGFR were revealed to be significantly increased in the model, curcumin and VEGF blocker groups. The VEGF mRNA levels in the curcumin, VEGF blocker and curcumin + VEGF blocker groups were all decreased when compared with the model group. In addition, the VEGF mRNA levels in the curcumin + VEGF blocker group were significantly lower compared with the curcumin group (P<0.05). The VEGF mRNA levels in the curcumin, VEGF blocker and curcumin + VEGF blocker groups were decreased when compared with the model group (P=0.0001). No significant differences in VEGF mRNA levels were identified between the VEGF blocker and curcumin groups (P=0.863), whereas the VEGF mRNA levels in the curcumin + VEGF blocker group were significantly lower than that of the curcumin group (P=0.025). Curcumin and the VEGF blocker are each capable of inhibiting hepatocellular carcinoma progression by regulating the VEGF/VEGFR/K-ras pathway. The combination of the two compounds has a synergistic effect on the inhibition of the effects of the VEGF signaling pathways in hepatocellular carcinoma progression.
Collapse
Affiliation(s)
- Jian-Zhi Gao
- Department of Pathology, General Hospital of the People's Liberation Army, Beijing 100853, P.R. China ; Basic Medical College of Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| | - Jing-Li DU
- Department of Gastroenterology and Hepatology, General Hospital of the People's Liberation Army, Beijing 100853, P.R. China ; Department of Gastroenterology, Armed Police Corps Hospital of Qinghai, Xining, Qinghai 810006, P.R. China
| | - Yong-Ling Wang
- Basic Medical College of Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| | - Jia Li
- Department of Pathology, General Hospital of the People's Liberation Army, Beijing 100853, P.R. China
| | - Li-Xin Wei
- Department of Pathology, General Hospital of the People's Liberation Army, Beijing 100853, P.R. China
| | - Ming-Zhou Guo
- Department of Gastroenterology and Hepatology, General Hospital of the People's Liberation Army, Beijing 100853, P.R. China
| |
Collapse
|
23
|
Zhou XH, Yang CQ, Zhang CL, Gao Y, Yuan HB, Wang C. RASSF5 inhibits growth and invasion and induces apoptosis in osteosarcoma cells through activation of MST1/LATS1 signaling. Oncol Rep 2014; 32:1505-12. [PMID: 25109282 DOI: 10.3892/or.2014.3387] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Accepted: 07/09/2014] [Indexed: 11/06/2022] Open
Abstract
Ras association (RalGDS/AF-6) domain family member RASSF5 has been implicated in a variety of key biological processes, including cell proliferation, cell cycle regulation and apoptosis. It is believed to play an important role in tumorigenesis as a tumor suppressor in a number of malignancies. Yet, little is known concerning the function and underlying mechanisms of RASSF5 in human osteosarcoma (OS). The expression of RASSF5 was examined by immunohistochemical assay using a tissue microarray in 45 cases of OS tissues. A gain-of-function approach was used to observe the effects of lentiviral vector-mediated overexpression of RASSF5 (Lv-RASSF5) on cell growth, invasion and apoptosis, respectively, as indicated by MTT, Transwell and flow cytometry assays, and the expression levels of mammalian sterile 20-like (MST1) kinase, large tumor suppressor 1 (LATS1), proliferating cell nuclear antigen (PCNA), matrix metallopeptidase-9 (MMP-9) and p53 were detected by real-time PCR and western blot assays in OS cells (MG-63 and U-2 OS). The results indicated that the expression of RASSF5 protein was significantly downregulated in OS tissues compared to that in adjacent non-cancerous tissues (ANCT) (40.0 vs. 73.3%, P=0.002), and had a negative correlation with distant metastasis of the tumor (P=0.01). Overexpression of RASSF5 markedly suppressed cell proliferation and invasion, and induced cell apoptosis in the OS cell lines with increased expression of MST1, LATS1 and p53 and decreased expression of PCNA and MMP-9. Taken together, our findings demonstrate that RASSF5 expression is negatively correlated with distant metastasis of OS, and RASSF5 may function as a tumor suppressor in OS cells through activation of the MST1/LATS1 pathway.
Collapse
Affiliation(s)
- Xu-Hui Zhou
- Department of Orthopedic Surgery, Changzheng Hospital, Second Military Medical University, Shanghai 200003, P.R. China
| | - Chao-Qun Yang
- Department of Orthopedic Surgery, Changzheng Hospital, Second Military Medical University, Shanghai 200003, P.R. China
| | - Cheng-Lin Zhang
- Department of Orthopedic Surgery, Changzheng Hospital, Second Military Medical University, Shanghai 200003, P.R. China
| | - Yang Gao
- Department of Orthopedic Surgery, Changzheng Hospital, Second Military Medical University, Shanghai 200003, P.R. China
| | - Hong-Bin Yuan
- Department of Anesthesiology, Changzheng Hospital, Second Military Medical University, Shanghai 200003, P.R. China
| | - Ce Wang
- Department of Orthopedic Surgery, Changzheng Hospital, Second Military Medical University, Shanghai 200003, P.R. China
| |
Collapse
|
24
|
Schöpel M, Jockers KFG, Düppe PM, Autzen J, Potheraveedu VN, Ince S, Yip KT, Heumann R, Herrmann C, Scherkenbeck J, Stoll R. Bisphenol A binds to Ras proteins and competes with guanine nucleotide exchange: implications for GTPase-selective antagonists. J Med Chem 2013; 56:9664-72. [PMID: 24266771 DOI: 10.1021/jm401291q] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
We show for the first time that bisphenol A (10) has the capacity to interact directly with K-Ras and that Rheb weakly binds to bisphenol A (10) and 4,4'-biphenol derivatives. We have characterized these interactions at atomic resolution suggesting that these compounds sterically interfere with the Sos-mediated nucleotide exchange in H- and K-Ras. We show that 4,4'-biphenol (5) selectively inhibits Rheb signaling and induces cell death suggesting that this compound might be a novel candidate for treatment of tuberous sclerosis-mediated tumor growth. Our results propose a new mode of action for bisphenol A (10) that advocates a reduced exposure to this compound in our environment. Our data may lay the foundation for the future design of GTPase-selective antagonists with higher affinity to benefit of the treatment of cancer because K-Ras inhibition is regarded to be a promising strategy with a potential therapeutic window for targeting Sos in Ras-driven tumors.
Collapse
Affiliation(s)
- Miriam Schöpel
- Faculty of Chemistry and Biochemistry, Ruhr University of Bochum , Universitätsstraße 150, D-44780 Bochum, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|