1
|
Wilczak M, Surman M, Przybyło M. The Role of Intracellular and Extracellular Vesicles in the Development of Therapy Resistance in Cancer. Curr Pharm Des 2024; 30:2765-2784. [PMID: 39113303 DOI: 10.2174/0113816128326325240723051625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 06/19/2024] [Indexed: 10/22/2024]
Abstract
Cancer is the second leading cause of global mortality and claims approximately 10 million lives annually. Despite advances in treatments such as surgery, chemotherapy, and immunotherapy, resistance to these methods has emerged. Multidrug resistance (MDR), where cancer cells resist diverse treatments, undermines therapy effectiveness, escalating mortality rates. MDR mechanisms include, among others, drug inactivation, reduced drug uptake, enhanced DNA repair, and activation of survival pathways such as autophagy. Moreover, MDR mechanisms can confer resistance to other therapies like radiotherapy. Understanding these mechanisms is crucial for improving treatment efficacy and identifying new therapeutic targets. Extracellular vesicles (EVs) have gathered attention for their role in cancer progression, including MDR development through protein transfer and genetic reprogramming. Autophagy, a process balancing cellular resources, also influences MDR. The intersection of EVs and autophagy further complicates the understanding of MDR. Both extracellular (exosomes, microvesicles) and intracellular (autophagic) vesicles contribute to therapy resistance by regulating the tumor microenvironment, facilitating cell communication, and modulating drug processing. While much is known about these pathways, there is still a need to explore their potential for predicting treatment responses and understanding tumor heterogeneity.
Collapse
Affiliation(s)
- Magdalena Wilczak
- Department of Glycoconjugate Biochemistry, Faculty of Biology, Institute of Zoology and Biomedical Research, Jagiellonian University, Krakow, Poland
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, Krakow, Poland
| | - Magdalena Surman
- Department of Glycoconjugate Biochemistry, Faculty of Biology, Institute of Zoology and Biomedical Research, Jagiellonian University, Krakow, Poland
| | - Małgorzata Przybyło
- Department of Glycoconjugate Biochemistry, Faculty of Biology, Institute of Zoology and Biomedical Research, Jagiellonian University, Krakow, Poland
| |
Collapse
|
2
|
Banaszek N, Kurpiewska D, Kozak K, Rutkowski P, Sobczuk P. Hedgehog pathway in sarcoma: from preclinical mechanism to clinical application. J Cancer Res Clin Oncol 2023; 149:17635-17649. [PMID: 37815662 PMCID: PMC10657326 DOI: 10.1007/s00432-023-05441-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 09/20/2023] [Indexed: 10/11/2023]
Abstract
Sarcomas are a diverse group of malignant neoplasms of mesenchymal origin. They develop rarely, but due to poor prognosis, they are a challenging and significant clinical problem. Currently, available therapeutic options have very limited activity. A better understating of sarcomas' pathogenesis may help develop more effective therapies in the future. The Sonic hedgehog (Shh) signaling pathway is involved in both embryonic development and mature tissue repair and carcinogenesis. Shh pathway inhibitors are presently used in the treatment of basal cell carcinoma. Its increased activity has been demonstrated in many sarcomas, including osteosarcoma, Ewing sarcoma, chondrosarcoma, rhabdomyosarcoma, leiomyosarcoma, and malignant rhabdoid tumor. In vitro studies have demonstrated the effectiveness of inhibitors of the Hedgehog pathway in inhibiting proliferation in those sarcomas in which the components of the pathway are overexpressed. These results were confirmed by in vivo studies, which additionally proved the influence of Shh pathway inhibitors on limiting the metastatic potential of sarcoma cells. However, until now, the efficacy of sarcomas treatment with Shh pathway inhibitors has not been established in clinical trials. The reason for that may be the non-canonical activation of the pathway or interactions with other signaling pathways, such as Wnt or Notch. In this review, we present the Shh signaling pathway's role in the pathogenesis of sarcomas, including both canonical and non-canonical signaling. We also propose how this knowledge could be potentially translated into clinics.
Collapse
Affiliation(s)
- Natalia Banaszek
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Skłodowska-Curie National Research Institute of Oncology in Warsaw, Warsaw, Poland
- Faculty of Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Dominika Kurpiewska
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Skłodowska-Curie National Research Institute of Oncology in Warsaw, Warsaw, Poland
- Faculty of Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Katarzyna Kozak
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Skłodowska-Curie National Research Institute of Oncology in Warsaw, Warsaw, Poland
| | - Piotr Rutkowski
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Skłodowska-Curie National Research Institute of Oncology in Warsaw, Warsaw, Poland
| | - Paweł Sobczuk
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Skłodowska-Curie National Research Institute of Oncology in Warsaw, Warsaw, Poland.
| |
Collapse
|
3
|
Virk RK, Garla R, Kaushal N, Bansal MP, Garg ML, Mohanty BP. The relevance of arsenic speciation analysis in health & medicine. CHEMOSPHERE 2023; 316:137735. [PMID: 36603678 DOI: 10.1016/j.chemosphere.2023.137735] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 12/24/2022] [Accepted: 12/31/2022] [Indexed: 06/17/2023]
Abstract
Long term exposure to arsenic through consumption of contaminated groundwater has been a global issue since the last five decades; while from an alternate standpoint, arsenic compounds have emerged as unparallel chemotherapeutic drugs. This review highlights the contribution from arsenic speciation studies that have played a pivotal role in the progression of our understanding of the biological behaviour of arsenic in humans. We also discuss the limitations of the speciation studies and their association with the interpretation of arsenic metabolism. Chromatographic separation followed by spectroscopic detection as well as the utilization of biotinylated pull-down assays, protein microarray and radiolabelled arsenic have been instrumental in identifying hundreds of metabolic arsenic conjugates, while, computational modelling has predicted thousands of them. However, these species exhibit a variegated pattern, which supports more than one hypothesis for the metabolic pathway of arsenic. Thus, the arsenic species are yet to be integrated into a coherent mechanistic pathway depicting its chemicobiological fate. Novel biorelevant arsenic species have been identified due to significant evolution in experimental methodologies. However, these methods are specific for the identification of only a group of arsenicals sharing similar physiochemical properties; and may not be applicable to other constituents of the vast spectrum of arsenic species. Consequently, the identity of arsenic binding partners in vivo and the sequence of events in arsenic metabolism are still elusive. This resonates the need for additional focus on the extraction and characterization of both low and high molecular weight arsenicals in a combinative manner.
Collapse
Affiliation(s)
- Rajbinder K Virk
- Department of Biophysics, Panjab University, Chandigarh, 160014, India.
| | - Roobee Garla
- Department of Biophysics, Panjab University, Chandigarh, 160014, India.
| | - Naveen Kaushal
- Department of Biophysics, Panjab University, Chandigarh, 160014, India.
| | - Mohinder P Bansal
- Department of Biophysics, Panjab University, Chandigarh, 160014, India.
| | - Mohan L Garg
- Department of Biophysics, Panjab University, Chandigarh, 160014, India.
| | - Biraja P Mohanty
- Department of Biophysics, Panjab University, Chandigarh, 160014, India.
| |
Collapse
|
4
|
Nguyen J, Saffari P, Pollack A, Vennam S, Gong X, West R, Pollack J. New Ameloblastoma Cell Lines Enable Preclinical Study of Targeted Therapies. J Dent Res 2022; 101:1517-1525. [PMID: 35689405 PMCID: PMC9608093 DOI: 10.1177/00220345221100773] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Ameloblastoma (AB) is an odontogenic tumor that arises from ameloblast-lineage cells. Although relatively uncommon and rarely metastatic, AB tumors are locally invasive and destructive to the jawbone and surrounding structures. Standard-of-care surgical resection often leads to disfigurement, and many tumors will locally recur, necessitating increasingly challenging surgeries. Recent genomic studies of AB have uncovered oncogenic driver mutations, including in the mitogen-activated protein kinase (MAPK) and Hedgehog signaling pathways. Medical therapies targeting those drivers would be a highly desirable alternative or addition to surgery; however, a paucity of existing AB cell lines has stymied clinical translation. To bridge this gap, here we report the establishment of 6 new AB cell lines-generated by "conditional reprogramming"-and their genomic characterization that reveals driver mutations in FGFR2, KRAS, NRAS, BRAF, PIK3CA, and SMO. Furthermore, in proof-of-principle studies, we use the new cell lines to investigate AB oncogene dependency and drug sensitivity. Among our findings, AB cells with KRAS or NRAS mutation (MAPK pathway) are exquisitely sensitive to MEK inhibition, which propels ameloblast differentiation. AB cells with activating SMO-L412F mutation (Hedgehog pathway) are insensitive to vismodegib; however, a distinct small-molecule SMO inhibitor, BMS-833923, significantly reduces both downstream Hedgehog signaling and tumor cell viability. The novel cell line resource enables preclinical studies and promises to speed the translation of new molecularly targeted therapies for the management of ameloblastoma and related odontogenic neoplasms.
Collapse
Affiliation(s)
- J. Nguyen
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - P.S. Saffari
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - A.S. Pollack
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - S. Vennam
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - X. Gong
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - R.B. West
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - J.R. Pollack
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
5
|
Zhang M, Gao L, Ye Y, Li X. Advances in glioma-associated oncogene (GLI) inhibitors for cancer therapy. Invest New Drugs 2022; 40:370-388. [PMID: 34837604 DOI: 10.1007/s10637-021-01187-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 09/22/2021] [Indexed: 10/19/2022]
Abstract
The Hedgehog/Glioma-associated oncogene homolog (HH/GLI) signaling pathway regulates self-renewal of rare and highly malignant cancer stem cells, which have been shown to account for the initiation and maintenance of tumor growth as well as for drug resistance, metastatic spread and relapse. As an important component of the Hh signaling pathway, glioma-associated oncogene (GLI) acts as a key signal transmission hub for various signaling pathways in many tumors. Here, we review direct and indirect inhibitors of GLI; summarize the abundant active structurally diverse natural GLI inhibitors; and discuss how to better develop and utilize GLI inhibitors to solve the problem of drug resistance in tumors of interest. In summary, GLI inhibitors will be promising candidates for various cancer treatments.
Collapse
Affiliation(s)
- Meng Zhang
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, China
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Lijuan Gao
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, China
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yiping Ye
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, China
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Xiaoyu Li
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, China.
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, Zhejiang, China.
| |
Collapse
|
6
|
Pu Y, Wang J, Wang S. Role of autophagy in drug resistance and regulation of osteosarcoma (Review). Mol Clin Oncol 2022; 16:72. [PMID: 35251623 DOI: 10.3892/mco.2022.2505] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 11/17/2021] [Indexed: 11/06/2022] Open
Affiliation(s)
- Yanchuan Pu
- Department of Orthopedics, Wuwei People's Hospital, Wuwei, Gansu 733000, P.R. China
| | - Jin Wang
- Department of Orthopedics, Wuwei People's Hospital, Wuwei, Gansu 733000, P.R. China
| | - Shizhong Wang
- Department of Orthopedics, Wuwei People's Hospital, Wuwei, Gansu 733000, P.R. China
| |
Collapse
|
7
|
Song ZL, Zhang J, Xu Q, Shi D, Yao X, Fang J. Structural Modification of Aminophenylarsenoxides Generates Candidates for Leukemia Treatment via Thioredoxin Reductase Inhibition. J Med Chem 2021; 64:16132-16146. [PMID: 34704769 DOI: 10.1021/acs.jmedchem.1c01441] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Upregulation of the selenoprotein thioredoxin reductase (TrxR) is of pathological significance in maintaining tumor phenotypes. Thus, TrxR inhibitors are promising cancer therapeutic agents. We prepared different amino-substituted phenylarsine oxides and evaluated their cytotoxicity and inhibition of TrxR. Compared with our reported p-substituted molecule (8), the o-substituted molecule (10) shows improved efficacy (nearly a fourfold increase) to kill leukemia HL-60 cells. Although the compounds 8 and 10 display similar potency to inhibit the purified TrxR, the o-substitution 10 exhibits higher potency than the p-substitution 8 to inhibit the cellular TrxR activity. Molecular docking results demonstrate the favorable weak interactions of the o-amino group with the TrxR C-terminal active site. Efficient inhibition of TrxR consequently induces the oxidative stress-mediated apoptosis of cancer cells. Silence of the TrxR expression sensitizes the cells to the arsenic compound treatment, further supporting the critical involvement of TrxR in the cellular actions of compound 10.
Collapse
Affiliation(s)
- Zi-Long Song
- State Key Laboratory of Applied Organic Chemistry, School of Pharmacy, and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China.,Botanical Agrochemicals Research & Development Center, Lanzhou Jiaotong University, Lanzhou 730070, China
| | - Junmin Zhang
- State Key Laboratory of Applied Organic Chemistry, School of Pharmacy, and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China.,State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau 999078, China
| | - Qianhe Xu
- State Key Laboratory of Applied Organic Chemistry, School of Pharmacy, and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Danfeng Shi
- State Key Laboratory of Applied Organic Chemistry, School of Pharmacy, and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Xiaojun Yao
- State Key Laboratory of Applied Organic Chemistry, School of Pharmacy, and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China.,State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau 999078, China
| | - Jianguo Fang
- State Key Laboratory of Applied Organic Chemistry, School of Pharmacy, and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| |
Collapse
|
8
|
Li Y, Feng C, Chen Y, Huang K, Li C, Xiong X, Li P, Zhou D, Peng X, Weng W, Deng X, Wu Y, Fang J. Improved Outcomes with Induction Chemotherapy Combined with Arsenic Trioxide in Stage 4 Neuroblastoma: A Case Series. Technol Cancer Res Treat 2021; 20:15330338211041454. [PMID: 34569870 PMCID: PMC8485563 DOI: 10.1177/15330338211041454] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Objective: The apoptotic and cytotoxic effects of arsenic trioxide (ATO) makes it a potentially suitable agent for the treatment of patients with neuroblastoma with poor prognosis; therefore, we try to evaluate the effectiveness and safety of ATO combined with reinduction/induction chemotherapy in children with recurrent/refractory or newly diagnosed stage 4 neuroblastoma. Methods: Retrospective analysis was performed on seven pediatric patients with recurrent /refractory or newly diagnosed stage 4 neuroblastoma treated with traditional reinduction/induction chemotherapy combined with ATO. Results: A total of 7 patients were treated synchronously with ATO and chemotherapy for up to nine courses; all patients received conventional chemotherapy plus a 0.16 mg/kg/day dose of intravenous ATO during reinduction/induction chemotherapy. Treatment was effective in five patients and ineffective in the other two patients. The overall response rate was 71.43% (5 of 7). The side effects of the ATO combination were minor, whereby only treatment in one patient was terminated at the sixth course due to a prolonged QT interval (0.51 s), which returned to normal after symptomatic treatment. Conclusions: ATO can be safely and effectively combined with chemotherapy drugs as a potential alternative means of treatment for high-risk stage 4 neuroblastoma, and we have observed that ATO can restore the sensitivity of chemotherapy in some patients who were resistant to previous chemotherapy. Further investigations and clinical data are required to confirm these observations.
Collapse
Affiliation(s)
- Yang Li
- 56713Sun Yet-Sen Memorial Hospital, Sun Yet-Sen University, Guangzhou, China
| | - Chuchu Feng
- 56713Sun Yet-Sen Memorial Hospital, Sun Yet-Sen University, Guangzhou, China
| | - Yantao Chen
- 56713Sun Yet-Sen Memorial Hospital, Sun Yet-Sen University, Guangzhou, China
| | - Ke Huang
- 56713Sun Yet-Sen Memorial Hospital, Sun Yet-Sen University, Guangzhou, China
| | - Chunmou Li
- 56713Sun Yet-Sen Memorial Hospital, Sun Yet-Sen University, Guangzhou, China
| | - Xilin Xiong
- 56713Sun Yet-Sen Memorial Hospital, Sun Yet-Sen University, Guangzhou, China
| | - Peng Li
- 302944South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Dunhua Zhou
- 56713Sun Yet-Sen Memorial Hospital, Sun Yet-Sen University, Guangzhou, China
| | - Xiaomin Peng
- 56713Sun Yet-Sen Memorial Hospital, Sun Yet-Sen University, Guangzhou, China
| | - Wenjun Weng
- 56713Sun Yet-Sen Memorial Hospital, Sun Yet-Sen University, Guangzhou, China
| | - Xiaogeng Deng
- 56713Sun Yet-Sen Memorial Hospital, Sun Yet-Sen University, Guangzhou, China
| | - Yaohao Wu
- 56713Sun Yet-Sen Memorial Hospital, Sun Yet-Sen University, Guangzhou, China
| | - Jianpei Fang
- 56713Sun Yet-Sen Memorial Hospital, Sun Yet-Sen University, Guangzhou, China
| |
Collapse
|
9
|
Tang R, Zhu J, Liu Y, Wu N, Han J. Formulation Comprising Arsenic Trioxide and Dimercaprol Enhances Radiosensitivity of Pancreatic Cancer Xenografts. Technol Cancer Res Treat 2021; 20:15330338211036324. [PMID: 34433326 PMCID: PMC8404670 DOI: 10.1177/15330338211036324] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
OBJECTIVE To investigate the efficacy of a formula comprising arsenic trioxide and dimercaprol (BAL-ATO) as a radiosensitizing agent in model mice with pancreatic cancer xenografts. METHODS Female BALB/c nude mice bearing SW1990 human pancreatic cancer xenografts were divided into four treatment arms, including control, radiotherapy (RT), BAL-ATO, and RT + BAL-ATO groups. Survival and tumor volume were analyzed. We also assessed apoptosis in tumor samples by live imaging and detected hypoxia by confocal laser microscope observation. We further investigated the mechanisms of BAL-ATO action in RT by detecting affected proteins by western blot and immunohistochemistry assays. RESULTS Median survival was significantly longer in the RT + BAL-ATO group (64.5 days) compared with the control (49.5 days), RT (39 days), and BAL-ATO (48 days) groups (P < 0.001). RT + BAL-ATO inhibited the growth of tumors in mice by 73% compared with the control group, which was significantly higher than the rate of inhibition following RT alone (59%) (P < 0.01). Further analysis showed an improved microenvironment in terms of hypoxia in tumors treated with BAL-ATO alone or RT + BAL-ATO. Expression of signaling molecules associated with pancreatic cancer stem cells, including CD24, CD44, ALDH1A1, Gli-1, and Nestin, was detected in tumors treated with BAL-ATO alone or in combination with RT. CONCLUSION These data suggest that BAL-ATO function as a radiosensitizer in mice with pancreatic cancer xenografts, via mechanisms involving hypoxia reduction and inhibition of signaling pathways associated with pancreatic cancer stem cells. BAL-ATO may thus be a promising radiosensitizing agent in patients with pancreatic cancer.
Collapse
Affiliation(s)
- Renyan Tang
- Department of Traditional Chinese Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jianmin Zhu
- Shanghai Clinical Center, Chinese Academy of Sciences/Xuhui Central Hospital, Shanghai, China
| | - Ying Liu
- Department of Oncology, Yunnan Traditional Chinese Medicine Hospital, Kunming, China
| | - Ning Wu
- Department of Oncology, Shanghai Pudong New Area Gongli Hospital, Shanghai, China
| | - Jinbin Han
- Department of Traditional Chinese Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
10
|
Murad R, Avanes A, Ma X, Geng S, Mortazavi A, Momand J. Transcriptome and chromatin landscape changes associated with trastuzumab resistance in HER2+ breast cancer cells. Gene 2021; 799:145808. [PMID: 34224831 DOI: 10.1016/j.gene.2021.145808] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 05/29/2021] [Accepted: 06/30/2021] [Indexed: 12/09/2022]
Abstract
We set out to uncover transcriptome and chromatin landscape changes that occur in HER2 + breast cancer (BC) cells upon acquiring resistance to trastuzumab. RNA-seq analysis was applied to two independently-derived BC cell lines with acquired resistance to trastuzumab (SKBr3.HerR and BT-474HerR) and their parental drug-sensitive cell lines (SKBr3 and BT-474). Chromatin landscape analysis indicated that the most significant increase in accessibility in resistant cells occurs in PPP1R1B within a segment spanning introns 1b through intron 3. Footprint analysis of this segment revealed that FoxJ3 (within intron 2) and Pou5A1/Sox2 (within inton 3) transcription factor motifs are protected in resistant cells. Overall, 344 shared genes were upregulated in both resistant cell lines relative to their parental counterparts and 453 shared genes were downregulated in both resistant cell lines relative to their parental counterparts. In resistant cells, genes associated with autophagy and mitochondria organization are upregulated and genes associated with ribosome assembly and cell cycle are downregulated relative to parental cells. The five top upregulated genes in drug-resistant breast cancer cells are APOD, AZGP1, ETV5, ALPP, and PPP1R1B. This is the first report of increased chromatin accessibility within PPP1R1B associated with its t-Darpp transcript increase, and points to a possible mechanism for its activation in trastuzumab-resistant cells.
Collapse
Affiliation(s)
- Rabi Murad
- Department of Developmental & Cell Biology, University of California Irvine, Irvine, CA 92617, USA
| | - Arabo Avanes
- Department of Chemistry and Biochemistry, California State University Los Angeles, Los Angeles, CA 90032, USA
| | - Xinyi Ma
- Department of Developmental & Cell Biology, University of California Irvine, Irvine, CA 92617, USA
| | - Shuhui Geng
- Department of Chemistry and Biochemistry, California State University Los Angeles, Los Angeles, CA 90032, USA
| | - Ali Mortazavi
- Department of Developmental & Cell Biology, University of California Irvine, Irvine, CA 92617, USA.
| | - Jamil Momand
- Department of Chemistry and Biochemistry, California State University Los Angeles, Los Angeles, CA 90032, USA.
| |
Collapse
|
11
|
Sonic Hedgehog Signature in Pediatric Primary Bone Tumors: Effects of the GLI Antagonist GANT61 on Ewing's Sarcoma Tumor Growth. Cancers (Basel) 2020; 12:cancers12113438. [PMID: 33228057 PMCID: PMC7699338 DOI: 10.3390/cancers12113438] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 11/16/2020] [Accepted: 11/17/2020] [Indexed: 01/07/2023] Open
Abstract
Simple Summary The poor clinical outcomes for Osteosarcoma (OS) and Ewing’s sarcoma (ES) patients underscore the urgency of developing novel therapeutic strategies for these pathologies. In this context, the emerging role of Sonic hedgehog (SHH) signaling in cancer has been critically evaluated, focusing on the potential for targeting SHH signaling as an anticancer strategy. The aims of this work were (1) to highlight and to compare a possible SHH/Gli signature between OS and ES, (2) to strengthen our knowledge concerning the role of EWS-FLI1 in the SHH signature in ES and (3) to evaluate the effect of the specific Gli inhibitor GANT61 in vivo on the growth of ES tumors using an orthotopic mice model. Our work identifies Gli1 as a promising therapeutic target in ES and demonstrates that GANT61, through inhibition of Gli1 transcriptional activity, may be a promising therapeutic strategy hindering ES tumor progression, and specifically primary tumor growth. Abstract Osteosarcoma (OS) and Ewing’s sarcoma (ES) are the most common malignant bone tumors in children and adolescents. In many cases, the prognosis remains very poor. The Sonic hedgehog (SHH) signaling pathway, strongly involved in the development of many cancers, regulate transcription via the transcriptional factors Gli1-3. In this context, RNAseq analysis of OS and ES cell lines reveals an increase of some major compounds of the SHH signaling cascade in ES cells, such as the transcriptional factor Gli1. This increase leads to an augmentation of the transcriptional response of Gli1 in ES cell lines, demonstrating a dysregulation of Gli1 signaling in ES cells and thus the rationale for targeting Gli1 in ES. The use of a preclinical model of ES demonstrates that GANT61, an inhibitor of the transcriptional factor Gli1, reduces ES primary tumor growth. In vitro experiments show that GANT61 decreases the viability of ES cell, mainly through its ability to induce caspase-3/7-dependent cell apoptosis. Taken together, these results demonstrates that GANT61 may be a promising therapeutic strategy for inhibiting the progression of primary ES tumors.
Collapse
|
12
|
Branco V, Pimentel J, Brito MA, Carvalho C. Thioredoxin, Glutathione and Related Molecules in Tumors of the Nervous System. Curr Med Chem 2020; 27:1878-1900. [PMID: 30706774 DOI: 10.2174/0929867326666190201113004] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 09/14/2018] [Accepted: 11/28/2018] [Indexed: 12/25/2022]
Abstract
BACKGROUND Central Nervous System (CNS) tumors have a poor survival prognosis due to their invasive and heterogeneous nature, in addition to the resistance to multiple treatments. OBJECTIVE In this paper, the main aspects of brain tumor biology and pathogenesis are reviewed both for primary tumors of the brain, (i.e., gliomas) and for metastasis from other malignant tumors, namely lung cancer, breast cancer and malignant melanoma which account for a high percentage of overall malignant brain tumors. We review the role of antioxidant systems, namely the thioredoxin and glutathione systems, in the genesis and/or progression of brain tumors. METHODS Although overexpression of Thioredoxin Reductase (TrxR) and Thioredoxin (Trx) is often linked to increased malignancy rate of brain tumors, and higher expression of Glutathione (GSH) and Glutathione S-Transferases (GST) are associated to resistance to therapy, several knowledge gaps still exist regarding for example, the role of Peroxiredoxins (Prx), and Glutaredoxins (Grx). CONCLUSION Due to their central role in redox homeostasis and ROS scavenging, redox systems are potential targets for new antitumorals and examples of innovative therapeutics aiming at improving success rates in brain tumor treatment are discussed.
Collapse
Affiliation(s)
- Vasco Branco
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - José Pimentel
- Laboratory of Neuropathology, Department of Neurology, Hospital de Santa Maria (CHLN), Av. Prof. Egas Moniz, 1649-036 Lisboa, Portugal.,Faculty of Medicine, Lisbon University, Av. Prof. Egas Moniz, 1649-036 Lisboa, Portugal
| | - Maria Alexandra Brito
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - Cristina Carvalho
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| |
Collapse
|
13
|
Ma W, Shen H, Li Q, Song H, Guo Y, Li F, Zhou X, Guo X, Shi J, Cui Q, Xing J, Deng J, Yu Y, Liu W, Zhao H. MARVELD1 attenuates arsenic trioxide-induced apoptosis in liver cancer cells by inhibiting reactive oxygen species production. ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:200. [PMID: 31205918 DOI: 10.21037/atm.2019.04.38] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Background Arsenic trioxide (As2O3) is widely used for the treatment of acute promyelocytic leukemia (APL), and more recently, has also been applied to solid tumors. However, there are a fraction of patients with solid tumors, such as liver cancer, who respond to As2O3 treatment poorly. The underlying mechanisms for this remain unclear. Methods We determined the suitable concentration of drugs by IC50. Cell Counting Kit-8 (CCK-8) and flow cytometry were used to analyze the apoptosis. Morphological changes of the cells were observed by laser scanning confocal microscopy. Furthermore, reactive oxygen species (ROS) and mitochondrial membrane potential (MMP) were detected by flow cytometry. Quantitative polymerase chain reaction (qPCR) and Western blot tests were conducted to detect the mRNA and protein levels in different groups. Finally, a xenograft tumor assay and histopathological analysis were performed to evaluate the MARVELD1 function in cell proliferation and apoptosis. Results Here, we show that MARVELD1 enhances the therapeutic effects of epirubicin, while inducing the strong resistance of liver cancer cells to As2O3 treatment. We further demonstrate that the As2O3-induced apoptosis was inhibited by MARVELD1 overexpression (24 h Vector vs. MARVELD1 =30.58% vs. 17.41%, P<0.01; 48 h Vector vs. MARVELD1 =46.50% vs. 21.02%, P<0.01), possibly through inhibiting ROS production by enhancing TRXR1 expression. In vivo, we found a significantly increased size (Vector vs. MARVELD1 =203.90±21.92 vs. 675.70±37.84 mm3, P<0.001) and weight (Vector vs. MARVELD1 =0.19±0.02 vs. 0.58±0.05 g, P<0.001) of tumors with high expression of MARVELD1 after As2O3 treatment. Consistently, a higher expression of MARVELD1 predicted a poor prognosis for liver cancer patients. Conclusions Our data identified a unique role of MARVELD1 in As2O3-induced apoptosis and As2O3 cancer therapy resistance.
Collapse
Affiliation(s)
- Wenping Ma
- Department of Medical Genetics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Haiyang Shen
- Department of Intervention Therapy, The Fourth Medical Center of PLA General Hospital, Beijing 100048, China
| | - Qian Li
- Department of Intervention Therapy, The Fourth Medical Center of PLA General Hospital, Beijing 100048, China
| | - Hao Song
- Department of Intervention Therapy, The Fourth Medical Center of PLA General Hospital, Beijing 100048, China
| | - Yanyan Guo
- Department of Intervention Therapy, The Fourth Medical Center of PLA General Hospital, Beijing 100048, China
| | - Fangrong Li
- Department of Intervention Therapy, The Fourth Medical Center of PLA General Hospital, Beijing 100048, China
| | - Xingang Zhou
- Department of Pathology, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Xinwu Guo
- Sansure Biotech Inc., Changsha 410205, China
| | - Jingdong Shi
- Department of General Surgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| | - Qi Cui
- Department of Medical Genetics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Jinhao Xing
- Department of Medical Genetics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Jinhai Deng
- Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Youtao Yu
- Department of Intervention Therapy, The Fourth Medical Center of PLA General Hospital, Beijing 100048, China
| | - Wenjie Liu
- Department of Hepatobiliary Surgery, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Hongshan Zhao
- Department of Medical Genetics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| |
Collapse
|
14
|
Ghanbari A, Cheraghzadeh Z, Mahmoudi R, Zibara K, Hosseini E. GLI inhibitors overcome Erlotinib resistance in human pancreatic cancer cells by modulating E-cadherin. J Chemother 2019; 31:141-149. [PMID: 30983542 DOI: 10.1080/1120009x.2019.1584422] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Inhibition of hedgehog (Hh) signalling pathway, including its end effector GLI1, can reverse epithelial-to-mesenchymal transition (EMT) which plays an important role in drug resistance of pancreatic cancer cells to Erlotinib (ETB). This study investigated the effect of GLI inhibitors Forskolin (FSK), GANT-61 (GNT), and Arsenic trioxide (ATX) on suppressing the resistance of pancreatic cancer cells to ETB. The effect of GLI inhibitors was evaluated by measuring mRNA expression levels of EMT factors using quantitative RT-PCR. Immunocytochemistry and flow cytometry were used to assess E-cadherin (E-Cad) and GLI1 protein levels. MTT and apoptosis assays were used to evaluate the synergistic effects for the combination treatment of each GLI inhibitor with ETB. Pancreatic cancer cells PANC-1 treated by GNT showed the highest significant reduction in mRNA levels of GLI1 and other EMT pathway genes. Moreover, GNT was able to upregulate E-Cad and downregulate GLI1 proteins, more than FSK, while ATX had no effect. Apoptosis levels of PANC-1 cells following treatment with LD30 concentrations of FSK, GNT, or ATX, showed 57%, 62% and 67%, respectively, in comparison to ETB (∼48%). Importantly, combination treatments of ETB with either FSK, GNT, or ATX demonstrated a significant increase in apoptotic cells reaching 61% (ETB + FSK), 80% (ETB + GNT) or 88% (ETB + ATX). FSK did not have much effect on the drug resistance of PANC-1 cells to ETB. However, GNT, but more effectively ATX, were able to reduce the drug resistance of this cell line to ETB.
Collapse
Affiliation(s)
- Amir Ghanbari
- a Cellular and Molecular Research Center , Yasuj University of Medical Sciences , Yasuj , Iran
| | - Zeinab Cheraghzadeh
- a Cellular and Molecular Research Center , Yasuj University of Medical Sciences , Yasuj , Iran
| | - Reza Mahmoudi
- a Cellular and Molecular Research Center , Yasuj University of Medical Sciences , Yasuj , Iran
| | - Kazem Zibara
- b PRASE, Biology Department, Faculty of Sciences-I , Lebanese University , Beirut , Lebanon
| | - Ebrahim Hosseini
- a Cellular and Molecular Research Center , Yasuj University of Medical Sciences , Yasuj , Iran
| |
Collapse
|
15
|
Dawood M, Hamdoun S, Efferth T. Multifactorial Modes of Action of Arsenic Trioxide in Cancer Cells as Analyzed by Classical and Network Pharmacology. Front Pharmacol 2018; 9:143. [PMID: 29535630 PMCID: PMC5835320 DOI: 10.3389/fphar.2018.00143] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Accepted: 02/09/2018] [Indexed: 12/13/2022] Open
Abstract
Arsenic trioxide is a traditional remedy in Chinese Medicine since ages. Nowadays, it is clinically used to treat acute promyelocytic leukemia (APL) by targeting PML/RARA. However, the drug's activity is broader and the mechanisms of action in other tumor types remain unclear. In this study, we investigated molecular modes of action by classical and network pharmacological approaches. CEM/ADR5000 resistance leukemic cells were similar sensitive to As2O3 as their wild-type counterpart CCRF-CEM (resistance ratio: 1.88). Drug-resistant U87.MG ΔEGFR glioblastoma cells harboring mutated epidermal growth factor receptor were even more sensitive (collateral sensitive) than wild-type U87.MG cells (resistance ratio: 0.33). HCT-116 colon carcinoma p53-/- knockout cells were 7.16-fold resistant toward As2O3 compared to wild-type cells. Forty genes determining cellular responsiveness to As2O3 were identified by microarray and COMPARE analyses in 58 cell lines of the NCI panel. Hierarchical cluster analysis-based heat mapping revealed significant differences between As2O3 sensitive cell lines and resistant cell lines with p-value: 1.86 × 10-5. The genes were subjected to Galaxy Cistrome gene promoter transcription factor analysis to predict the binding of transcription factors. We have exemplarily chosen NF-kB and AP-1, and indeed As2O3 dose-dependently inhibited the promoter activity of these two transcription factors in reporter cell lines. Furthermore, the genes identified here and those published in the literature were assembled and subjected to Ingenuity Pathway Analysis for comprehensive network pharmacological approaches that included all known factors of resistance of tumor cells to As2O3. In addition to pathways related to the anticancer effects of As2O3, several neurological pathways were identified. As arsenic is well-known to exert neurotoxicity, these pathways might account for neurological side effects. In conclusion, the activity of As2O3 is not restricted to acute promyelocytic leukemia. In addition to PML/RARA, numerous other genes belonging to diverse functional classes may also contribute to its cytotoxicity. Network pharmacology is suited to unravel the multifactorial modes of action of As2O3.
Collapse
Affiliation(s)
| | | | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmacy and Biochemistry, Johannes Gutenberg University, Mainz, Germany
| |
Collapse
|
16
|
D'Amico D, Canettieri G. Translating Hedgehog in Cancer: Controlling Protein Synthesis. Trends Mol Med 2016; 22:851-862. [PMID: 27591077 DOI: 10.1016/j.molmed.2016.08.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 08/04/2016] [Accepted: 08/05/2016] [Indexed: 11/17/2022]
Abstract
Developmental Hedgehog (Hh) signaling is found deregulated in a broad spectrum of human malignancies and, thus, is an attractive target for cancer therapy. Currently available Hh inhibitors have shown the rapid occurrence of drug resistance, due to altered signaling in collateral pathways. Emerging observations suggest that Hh signaling regulates protein translation in pathways that depend both on Cap- and IRES-mediated translation. In addition, translational regulators have been shown to modulate Hh function. In this opinion, we describe this novel Hh/translation crosstalk and argue that it plays a relevant role in Hh-mediated tumorigenesis and drug resistance. As such, we suggest that drugs targeting translation might be introduced in novel protocols aimed at treating malignancies driven by aberrant Hh signaling.
Collapse
Affiliation(s)
- Davide D'Amico
- Laboratory of Integrative and Systems Physiology, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | | |
Collapse
|
17
|
Xing G, Wang B, Lei Y, Liu C, Wang Z, Shi H, Yang R, Qin W, Jiang Y, Lv H. In vitro effect of sodium arsenite on Echinococcus granulosus protoscoleces. Mol Biochem Parasitol 2016; 207:49-55. [DOI: 10.1016/j.molbiopara.2016.05.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2015] [Revised: 05/03/2016] [Accepted: 05/21/2016] [Indexed: 01/26/2023]
|
18
|
Parrales A, Iwakuma T. Targeting Oncogenic Mutant p53 for Cancer Therapy. Front Oncol 2015; 5:288. [PMID: 26732534 PMCID: PMC4685147 DOI: 10.3389/fonc.2015.00288] [Citation(s) in RCA: 247] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 12/07/2015] [Indexed: 12/14/2022] Open
Abstract
Among genetic alterations in human cancers, mutations in the tumor suppressor p53 gene are the most common, occurring in over 50% of human cancers. The majority of p53 mutations are missense mutations and result in the accumulation of dysfunctional p53 protein in tumors. These mutants frequently have oncogenic gain-of-function activities and exacerbate malignant properties of cancer cells, such as metastasis and drug resistance. Increasing evidence reveals that stabilization of mutant p53 in tumors is crucial for its oncogenic activities, while depletion of mutant p53 attenuates malignant properties of cancer cells. Thus, mutant p53 is an attractive druggable target for cancer therapy. Different approaches have been taken to develop small-molecule compounds that specifically target mutant p53. These include compounds that restore wild-type conformation and transcriptional activity of mutant p53, induce depletion of mutant p53, inhibit downstream pathways of oncogenic mutant p53, and induce synthetic lethality to mutant p53. In this review article, we comprehensively discuss the current strategies targeting oncogenic mutant p53 in cancers, with special focus on compounds that restore wild-type p53 transcriptional activity of mutant p53 and those reducing mutant p53 levels.
Collapse
Affiliation(s)
- Alejandro Parrales
- Department of Cancer Biology, University of Kansas Medical Center , Kansas City, KS , USA
| | - Tomoo Iwakuma
- Department of Cancer Biology, University of Kansas Medical Center , Kansas City, KS , USA
| |
Collapse
|
19
|
Arsenic trioxide negatively affects Echinococcus granulosus. Antimicrob Agents Chemother 2015; 59:6946-51. [PMID: 26324279 DOI: 10.1128/aac.04340-14] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Accepted: 08/14/2015] [Indexed: 11/20/2022] Open
Abstract
Spillage of cyst contents during surgery is the major cause of recurrences of hydatidosis, also called cystic echinococcosis (CE). Currently, many scolicidal agents are used for inactivation of the cyst contents. However, due to complications in the use of those agents, new and more-effective treatment options are urgently needed. The aim of this study was to investigate the in vitro efficacy of arsenic trioxide (ATO) against Echinococcus granulosus protoscolices. Protoscolices of E. granulosus were incubated in vitro with 2, 4, 6, and 8 μmol/liter ATO; viability of protoscolices was assessed daily by microscopic observation of movements and 0.1% eosin staining. A small sample from each culture was processed for scanning and transmission electron microscopy. ATO demonstrated a potent ability to kill protoscolices, suggesting that ATO may represent a new strategy in treating hydatid cyst echinococcosis. However, the in vivo efficacy and possible side effects of ATO need to be explored.
Collapse
|
20
|
Kramann R, Fleig SV, Schneider RK, Fabian SL, DiRocco DP, Maarouf O, Wongboonsin J, Ikeda Y, Heckl D, Chang SL, Rennke HG, Waikar SS, Humphreys BD. Pharmacological GLI2 inhibition prevents myofibroblast cell-cycle progression and reduces kidney fibrosis. J Clin Invest 2015; 125:2935-51. [PMID: 26193634 PMCID: PMC4563736 DOI: 10.1172/jci74929] [Citation(s) in RCA: 132] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2013] [Accepted: 06/04/2015] [Indexed: 12/21/2022] Open
Abstract
Chronic kidney disease is characterized by interstitial fibrosis and proliferation of scar-secreting myofibroblasts, ultimately leading to end-stage renal disease. The hedgehog (Hh) pathway transcriptional effectors GLI1 and GLI2 are expressed in myofibroblast progenitors; however, the role of these effectors during fibrogenesis is poorly understood. Here, we demonstrated that GLI2, but not GLI1, drives myofibroblast cell-cycle progression in cultured mesenchymal stem cell-like progenitors. In animals exposed to unilateral ureteral obstruction, Hh pathway suppression by expression of the GLI3 repressor in GLI1+ myofibroblast progenitors limited kidney fibrosis. Myofibroblast-specific deletion of Gli2, but not Gli1, also limited kidney fibrosis, and induction of myofibroblast-specific cell-cycle arrest mediated this inhibition. Pharmacologic targeting of this pathway with darinaparsin, an arsenical in clinical trials, reduced fibrosis through reduction of GLI2 protein levels and subsequent cell-cycle arrest in myofibroblasts. GLI2 overexpression rescued the cell-cycle effect of darinaparsin in vitro. While darinaparsin ameliorated fibrosis in WT and Gli1-KO mice, it was not effective in conditional Gli2-KO mice, supporting GLI2 as a direct darinaparsin target. The GLI inhibitor GANT61 also reduced fibrosis in mice. Finally, GLI1 and GLI2 were upregulated in the kidneys of patients with high-grade fibrosis. Together, these data indicate that GLI inhibition has potential as a therapeutic strategy to limit myofibroblast proliferation in kidney fibrosis.
Collapse
Affiliation(s)
- Rafael Kramann
- Renal Division, Brigham and Women’s Hospital, Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
- Division of Nephrology and Clinical Immunology, RWTH Aachen University Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Susanne V. Fleig
- Renal Division, Brigham and Women’s Hospital, Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
- Division of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - Rebekka K. Schneider
- Division of Hematology, Brigham and Women’s Hospital, Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Steven L. Fabian
- Renal Division, Brigham and Women’s Hospital, Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Derek P. DiRocco
- Renal Division, Brigham and Women’s Hospital, Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Omar Maarouf
- Renal Division, Brigham and Women’s Hospital, Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Janewit Wongboonsin
- Renal Division, Brigham and Women’s Hospital, Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Yoichiro Ikeda
- Renal Division, Brigham and Women’s Hospital, Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Dirk Heckl
- Division of Hematology, Brigham and Women’s Hospital, Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | | | - Helmut G. Rennke
- Department of Pathology, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Sushrut S. Waikar
- Renal Division, Brigham and Women’s Hospital, Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Benjamin D. Humphreys
- Renal Division, Brigham and Women’s Hospital, Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
- Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
| |
Collapse
|
21
|
Subbarayan PR, Ardalan B. In the war against solid tumors arsenic trioxide needs partners. J Gastrointest Cancer 2015; 45:363-71. [PMID: 24825822 DOI: 10.1007/s12029-014-9617-8] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
In the past decade, the therapeutic potential of arsenic trioxide (ATO) in the treatment of acute promyelocytic leukemia (APL) was recognized. This encouraged other investigators to test the efficacy of ATO in the management of other hematological and solid tumor malignancies. Notably, as a single agent, arsenic trioxide did not benefit patients diagnosed with solid tumors. However, when it was combined with other agents, treatment benefit emerged. In this article, we have summarized the outcome of clinical trials that used arsenic trioxide as a single agent as well as in combination settings in patients diagnosed with solid tumors. We have also reviewed possible additional mechanisms by which ATO may be useful as a chemosensitizer in combination therapy. We hope that our review will encourage clinical investigators to rationally combine ATO with additional chemotherapeutic agents in treating patients diagnosed with solid tumors.
Collapse
Affiliation(s)
- Pochi R Subbarayan
- Department of Medicine, University of Miami Miller School of Medicine, 1550 NW 10th Avenue, FOX 431A, Miami, FL, 33136, USA
| | | |
Collapse
|
22
|
Abstract
The hedgehog (Hh) signaling pathway is well established as being evolutionarily conserved across vertebrates, and is involved in organogenesis, hematopoiesis, embryogenesis and homeostasis of adult tissues. At a microscopic level, the Hh signaling pathway controls the proliferation, apoptosis, cell-cycle and differentiation programs of stem and progenitor cells. Increasing evidence suggests that aberrant activation of the Hh signaling pathway is related to neoplasm, including solid tumors and hematologic malignancies. Currently the Hh signaling pathway has become one of the most studied potential therapeutic targets in hematological malignancies. In this review, we focus on findings related to Hh signaling in the initiation, maintenance, progression and chemoresistance of hematological malignancies, looking forward to better targeted treatment strategies.
Collapse
Affiliation(s)
- Lingyun Geng
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong University , Jinan, Shandong , P. R. China
| | | |
Collapse
|
23
|
Liu Y, Duan D, Yao J, Zhang B, Peng S, Ma H, Song Y, Fang J. Dithiaarsanes Induce Oxidative Stress-Mediated Apoptosis in HL-60 Cells by Selectively Targeting Thioredoxin Reductase. J Med Chem 2014; 57:5203-11. [DOI: 10.1021/jm500221p] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Yaping Liu
- State Key
Laboratory of Applied
Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu 730000, China
| | - Dongzhu Duan
- State Key
Laboratory of Applied
Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu 730000, China
| | - Juan Yao
- State Key
Laboratory of Applied
Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu 730000, China
| | - Baoxin Zhang
- State Key
Laboratory of Applied
Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu 730000, China
| | - Shoujiao Peng
- State Key
Laboratory of Applied
Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu 730000, China
| | - HuiLong Ma
- State Key
Laboratory of Applied
Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu 730000, China
| | - Yanlin Song
- State Key
Laboratory of Applied
Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu 730000, China
| | - Jianguo Fang
- State Key
Laboratory of Applied
Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu 730000, China
| |
Collapse
|
24
|
Srivastava RK, Li C, Chaudhary SC, Ballestas ME, Elmets CA, Robbins DJ, Matalon S, Deshane JS, Afaq F, Bickers DR, Athar M. Unfolded protein response (UPR) signaling regulates arsenic trioxide-mediated macrophage innate immune function disruption. Toxicol Appl Pharmacol 2013; 272:879-87. [PMID: 23954561 PMCID: PMC6028020 DOI: 10.1016/j.taap.2013.08.004] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Revised: 08/02/2013] [Accepted: 08/03/2013] [Indexed: 11/26/2022]
Abstract
Arsenic exposure is known to disrupt innate immune functions in humans and in experimental animals. In this study, we provide a mechanism by which arsenic trioxide (ATO) disrupts macrophage functions. ATO treatment of murine macrophage cells diminished internalization of FITC-labeled latex beads, impaired clearance of phagocytosed fluorescent bacteria and reduced secretion of pro-inflammatory cytokines. These impairments in macrophage functions are associated with ATO-induced unfolded protein response (UPR) signaling pathway characterized by the enhancement in proteins such as GRP78, p-PERK, p-eIF2α, ATF4 and CHOP. The expression of these proteins is altered both at transcriptional and translational levels. Pretreatment with chemical chaperon, 4-phenylbutyric acid (PBA) attenuated the ATO-induced activation in UPR signaling and afforded protection against ATO-induced disruption of macrophage functions. This treatment also reduced ATO-mediated reactive oxygen species (ROS) generation. Interestingly, treatment with antioxidant N-acetylcysteine (NAC) prior to ATO exposure, not only reduced ROS production and UPR signaling but also improved macrophage functions. These data demonstrate that UPR signaling and ROS generation are interdependent and are involved in the arsenic-induced pathobiology of macrophage. These data also provide a novel strategy to block the ATO-dependent impairment in innate immune responses.
Collapse
Affiliation(s)
- Ritesh K. Srivastava
- Department of Dermatology and Skin Diseases Research Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Changzhao Li
- Department of Dermatology and Skin Diseases Research Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Sandeep C. Chaudhary
- Department of Dermatology and Skin Diseases Research Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Mary E. Ballestas
- Department of Pediatrics Infectious Disease, Children’s of Alabama, School of Medicine, University of Alabama at Birmingham, AL, USA
| | - Craig A. Elmets
- Department of Dermatology and Skin Diseases Research Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - David J. Robbins
- Department of Surgery, Molecular Oncology Program, Miller School of Medicine, University of Miami, Miami, USA
| | - Sadis Matalon
- Department of Anesthesiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jessy S. Deshane
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Farrukh Afaq
- Department of Dermatology and Skin Diseases Research Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - David R. Bickers
- Department of Dermatology, Columbia University Medical Center, New York, USA
| | - Mohammad Athar
- Department of Dermatology and Skin Diseases Research Center, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
25
|
Han JB, Sang F, Chang JJ, Hua YQ, Shi WD, Tang LH, Liu LM. Arsenic trioxide inhibits viability of pancreatic cancer stem cells in culture and in a xenograft model via binding to SHH-Gli. Onco Targets Ther 2013. [PMID: 23990729 DOI: 10.2147/ott.s49148.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
OBJECTIVE Overexpression of the sonic hedgehog (SHH) signaling pathway is an essential characteristic of pancreatic cancer stem cells (PCSCs) and arsenic trioxide (ATO) is described as a SHH inhibitor. This study evaluates whether ATO has the potential to inhibit viability of PCSCs via binding to SHH-Gli proteins. METHODS Cell counting kit-8 and flow cytometry were used for analyzing apoptosis in cells in vitro. The animal model was an athymic nude mouse model bearing subcutaneous xenografts of SW1990 pancreatic cancer cells. The terminal deoxynucleotidyl transferase dUTP nick end labeling assay and immunohistochemistry were used for tumor tissue analysis. The interaction between Gli1 and ATO was examined by a confocal system and an ultraviolet absorption spectrum assay. RESULTS ATO induced apoptosis in pancreatic cancer cells, especially CD24(+)CD44(+) cells in vitro. Combination treatment of ATO and low dose gemcitabine inhibited tumor growth by 60.9% (P = 0.004), and decreased the expression of CD24, CD44, and aldehyde dehydrogenase 1 family, member A1 significantly in vivo. ATO changed the structure of the recombinant Gli1 zinc finger peptides in a cell-free condition and the binding action of ATO to recombinant Gli1 was observed in cultured pancreatic cancer cells. CONCLUSION ATO may have the potential to inhibit viability of PCSCs via binding to SHH-Gli proteins in vitro and in vivo.
Collapse
Affiliation(s)
- Jin-Bin Han
- Department of integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China ; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
26
|
Han JB, Sang F, Chang JJ, Hua YQ, Shi WD, Tang LH, Liu LM. Arsenic trioxide inhibits viability of pancreatic cancer stem cells in culture and in a xenograft model via binding to SHH-Gli. Onco Targets Ther 2013; 6:1129-38. [PMID: 23990729 PMCID: PMC3753152 DOI: 10.2147/ott.s49148] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Objective Overexpression of the sonic hedgehog (SHH) signaling pathway is an essential characteristic of pancreatic cancer stem cells (PCSCs) and arsenic trioxide (ATO) is described as a SHH inhibitor. This study evaluates whether ATO has the potential to inhibit viability of PCSCs via binding to SHH-Gli proteins. Methods Cell counting kit-8 and flow cytometry were used for analyzing apoptosis in cells in vitro. The animal model was an athymic nude mouse model bearing subcutaneous xenografts of SW1990 pancreatic cancer cells. The terminal deoxynucleotidyl transferase dUTP nick end labeling assay and immunohistochemistry were used for tumor tissue analysis. The interaction between Gli1 and ATO was examined by a confocal system and an ultraviolet absorption spectrum assay. Results ATO induced apoptosis in pancreatic cancer cells, especially CD24+CD44+ cells in vitro. Combination treatment of ATO and low dose gemcitabine inhibited tumor growth by 60.9% (P = 0.004), and decreased the expression of CD24, CD44, and aldehyde dehydrogenase 1 family, member A1 significantly in vivo. ATO changed the structure of the recombinant Gli1 zinc finger peptides in a cell-free condition and the binding action of ATO to recombinant Gli1 was observed in cultured pancreatic cancer cells. Conclusion ATO may have the potential to inhibit viability of PCSCs via binding to SHH-Gli proteins in vitro and in vivo.
Collapse
Affiliation(s)
- Jin-Bin Han
- Department of integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China ; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
27
|
Barry NPE, Sadler PJ. Exploration of the medical periodic table: towards new targets. Chem Commun (Camb) 2013; 49:5106-5131. [PMID: 23636600 DOI: 10.1039/c3cc41143e] [Citation(s) in RCA: 573] [Impact Index Per Article: 47.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
Metallodrugs offer potential for unique mechanisms of drug action based on the choice of the metal, its oxidation state, the types and number of coordinated ligands and the coordination geometry. We discuss recent progress in identifying new target sites and elucidating the mechanisms of action of anti-cancer, anti-bacterial, anti-viral, anti-parasitic, anti-inflammatory, and anti-neurodegenerative agents, as well as in the design of metal-based diagnostic agents. Progress in identifying and defining target sites has been accelerated recently by advances in proteomics, genomics and metal speciation analysis. Examples of metal compounds and chelating agents (enzyme inhibitors) currently in clinical use, clinical trials or preclinical development are highlighted.
Collapse
Affiliation(s)
- Nicolas P E Barry
- University of Warwick, Department of Chemistry, Gibbet Hill Road, Warwick, UK.
| | | |
Collapse
|
28
|
Sapra A, Thorpe C. An arsenical-maleimide for the generation of new targeted biochemical reagents. J Am Chem Soc 2013; 135:2415-8. [PMID: 23384038 DOI: 10.1021/ja310553h] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The finding that arsenic trioxide is an effective treatment for acute promyelocytic leukemia has renewed interest in the pharmacological uses of inorganic and organic arsenicals. Here we synthesized and characterized the reactivity of an arsenical-maleimide (As-Mal) that can be efficiently conjugated to exposed cysteine residues in peptides and proteins with the ultimate goal of directing these As(III) species to vicinal thiols in susceptible targets within cells and tissues. As-Mal conjugated to a surface cysteine in thioredoxin provides a more potent inhibitor for Escherichia coli thioredoxin reductase than comparable simple inorganic or organic arsenicals. As-Mal can be coupled to all of the eight cysteine residues of reduced unfolded ribonuclease A or to site-specific locations using appropriate cysteine mutations. We observed particularly strong binding to the two CxxC motifs of protein disulfide isomerase using a mutant RNase in which As-Mal was specifically incorporated at residues 26 and 110. As-Mal will serve as a facile reagent for the incorporation of As(III) species into a wide range of thiol-containing proteins, biomaterials, and surfaces.
Collapse
Affiliation(s)
- Aparna Sapra
- Department of Chemistry and Biochemistry, University of Delaware, Newark, Delaware 19716, USA
| | | |
Collapse
|