1
|
Zeng Y, Luo Y, Zhao K, Liu S, Wu K, Wu Y, Du K, Pan W, Dai Y, Liu Y, Ren M, Tian F, Zhou L, Gu C. m6A-Mediated Induction of 7-Dehydrocholesterol Reductase Stimulates Cholesterol Synthesis and cAMP Signaling to Promote Bladder Cancer Metastasis. Cancer Res 2024; 84:3402-3418. [PMID: 39047230 DOI: 10.1158/0008-5472.can-23-3703] [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: 11/22/2023] [Revised: 03/29/2024] [Accepted: 07/16/2024] [Indexed: 07/27/2024]
Abstract
Dysregulation of cholesterol homeostasis occurs in multiple types of tumors and promotes cancer progression. Investigating the specific processes that induce abnormal cholesterol metabolism could identify therapeutic targets to improve cancer treatment. In this investigation, we observed upregulation of 7-dehydrocholesterol reductase (DHCR7), a vital enzyme involved in the synthesis of cholesterol, within bladder cancer tissues in comparison to normal tissues, which was correlated with increased bladder cancer metastasis. Increased expression of DHCR7 in bladder cancer was attributed to decreased mRNA degradation mediated by YTHDF2. Loss or inhibition of DHCR7 reduced bladder cancer cell invasion in vitro and metastasis in vivo. Mechanistically, DHCR7 promoted bladder cancer metastasis by activating the cAMP/protein kinase A/FAK pathway. Specifically, DHCR7 increased cAMP levels by elevating cholesterol content in lipid rafts, thereby facilitating the transduction of signaling pathways mediated by cAMP receptors. DHCR7 additionally enhanced the cAMP signaling pathway by reducing the concentration of 7-dehydrocholesterol and promoting the transcription of the G protein-coupled receptor, namely gastric inhibitory polypeptide receptor. Overall, these findings demonstrate that DHCR7 plays an important role in bladder cancer invasion and metastasis by modulating cholesterol synthesis and cAMP signaling. Furthermore, inhibition of DHCR7 shows promise as a viable therapeutic strategy for suppressing bladder cancer invasion and metastasis. Significance: Inhibiting DHCR7 induces cholesterol metabolism reprogramming and lipid raft remodeling to inactivate the cAMP/protein kinase A/FAK axis and suppress bladder cancer metastasis, indicating the therapeutic potential of targeting DHCR7.
Collapse
Affiliation(s)
- Youmiao Zeng
- Department of Urology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Urology, Henan Institute of Urology and Zhengzhou Key Laboratory for Molecular Biology of Urological Tumor Research, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Unit of Day Surgery Center, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yongbo Luo
- Department of Urology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Urology, Henan Institute of Urology and Zhengzhou Key Laboratory for Molecular Biology of Urological Tumor Research, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Unit of Day Surgery Center, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Keyuan Zhao
- Department of Urology, Shaoxing People's Hospital, Shaoxing, China
| | - Sheng Liu
- Department of General Surgery, The First Affiliated Hospital of Xinxiang Medical University, Weihui, China
| | - Kaiwen Wu
- Shenyang Medical College, Shenyang, China
| | - Yudong Wu
- Department of Urology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Kaixuan Du
- Department of Urology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Urology, Henan Institute of Urology and Zhengzhou Key Laboratory for Molecular Biology of Urological Tumor Research, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Unit of Day Surgery Center, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wenbang Pan
- Department of Urology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Urology, Henan Institute of Urology and Zhengzhou Key Laboratory for Molecular Biology of Urological Tumor Research, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Unit of Day Surgery Center, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yiheng Dai
- Department of Urology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Urology, Henan Institute of Urology and Zhengzhou Key Laboratory for Molecular Biology of Urological Tumor Research, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Unit of Day Surgery Center, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yuanhao Liu
- Department of Urology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Urology, Henan Institute of Urology and Zhengzhou Key Laboratory for Molecular Biology of Urological Tumor Research, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Unit of Day Surgery Center, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Mengda Ren
- Department of Urology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Urology, Henan Institute of Urology and Zhengzhou Key Laboratory for Molecular Biology of Urological Tumor Research, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Unit of Day Surgery Center, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Fengyan Tian
- Department of Pediatrics, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lijie Zhou
- Department of Urology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Urology, Henan Institute of Urology and Zhengzhou Key Laboratory for Molecular Biology of Urological Tumor Research, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Unit of Day Surgery Center, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chaohui Gu
- Department of Urology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Urology, Henan Institute of Urology and Zhengzhou Key Laboratory for Molecular Biology of Urological Tumor Research, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Unit of Day Surgery Center, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
2
|
Li J, Wang X, Zhang H, Hu X, Peng X, Jiang W, Zhuo L, Peng Y, Zeng G, Wang Z. Fenamates: Forgotten treasure for cancer treatment and prevention: Mechanisms of action, structural modification, and bright future. Med Res Rev 2024. [PMID: 39171404 DOI: 10.1002/med.22079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 08/03/2024] [Accepted: 08/08/2024] [Indexed: 08/23/2024]
Abstract
Fenamates as classical nonsteroidal anti-inflammatory agents are widely used for relieving pain. Preclinical studies and epidemiological data highlight their chemo-preventive and chemotherapeutic potential for cancer. However, comprehensive reviews of fenamates in cancer are limited. To accelerate the repurposing of fenamates, this review summarizes the results of fenamates alone or in combination with existing chemotherapeutic agents. This paper also explores targets of fenamates in cancer therapy, including COX, AKR family, AR, gap junction, FTO, TEAD, DHODH, TAS2R14, ion channels, and DNA. Besides, this paper discusses other mechanisms, such as regulating Wnt/β-catenin, TGF-β, p38 MAPK, and NF-κB pathway, and the regulation of the expressions of Sp, EGR-1, NAG-1, ATF-3, ErbB2, AR, as well as the modulation of the tumor immune microenvironment. Furthermore, this paper outlined the structural modifications of fenamates, highlighting their potential as promising leads for anticancer drugs.
Collapse
Affiliation(s)
- Junfang Li
- School of Pharmacy, Lanzhou University, Lanzhou, Gansu, China
| | - Xiaodong Wang
- School of Pharmacy, Lanzhou University, Lanzhou, Gansu, China
| | - Honghua Zhang
- School of Pharmacy, Lanzhou University, Lanzhou, Gansu, China
| | - Xiaoling Hu
- School of Pharmacy, Lanzhou University, Lanzhou, Gansu, China
| | - Xue Peng
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Weifan Jiang
- Postdoctoral Station for Basic Medicine, School of Basic Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Linsheng Zhuo
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Postdoctoral Station for Basic Medicine, School of Basic Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Yan Peng
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Guo Zeng
- Postdoctoral Station for Basic Medicine, School of Basic Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Zhen Wang
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Postdoctoral Station for Basic Medicine, School of Basic Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| |
Collapse
|
3
|
Han D, Xiong B, Zhang X, Chen C, Yao Z, Wu H, Cao J, Li J, Li P, Wang Z, Tian J. Knockdown of AMIGO2 suppresses proliferation and migration through regulating PPAR-γ in bladder cancer. Hereditas 2024; 161:21. [PMID: 38978149 PMCID: PMC11229346 DOI: 10.1186/s41065-024-00325-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 06/14/2024] [Indexed: 07/10/2024] Open
Abstract
PURPOSE This study aims to reveal the relationship between AMIGO2 and proliferation, migration and tumorigenicity of bladder cancer, and explore the potential molecular mechanisms. METHODS The expression level of AMIGO2 is measured by qRT-PCR and immunohistochemistry (IHC). Stable AMIGO2 knockdown cell lines T24 and 5637 were established by lentivirus transfection. Cell Counting Kit (CCK-8 assay) was produced to determine cell proliferation, flow cytometry analysis was utilized to detect cell cycle, and wound healing assay was proceeded to test migration ability of bladder cancer cells. Xenograft mouse model was established for investigating the effect of AMIGO2 on tumor formation in vivo. The RNA Sequencing technology was applied to explore the underlying mechanisms. The expression level of PPAR-γ was measured by Western Blot. RESULTS AMIGO2 was upregulated in bladder cancer cells and tissues. Inhibited expression of AMIGO2 suppresses cell proliferation and migration. Low AMIGO2 expression inhibited tumorigenicity of 5637 in nude mice. According to RNA-Seq and bioinformatics analysis, 917 DEGs were identified. The DEGs were mainly enriched in cell-cell adhesion, peroxisome proliferators-activated receptors (PPARs) signaling pathway and some other pathways. PPAR-γ is highly expressed in bladder cancer cell lines T24 and 5637, but when AMIGO2 is knocked down in T24 and 5637, the expression level of PPAR-γ is also decreased, and overexpression of PPAR-γ could reverse the suppression effect of cell proliferation and migration caused by the inhibition of AMIGO2. CONCLUSION AMIGO2 is overexpressed in bladder cancer cells and tissues. Knockdown of AMIGO2 suppresses bladder cancer cell proliferation and migration. These processes might be regulated by PPAR-γ signaling pathway.
Collapse
Affiliation(s)
- Dali Han
- Department of Urology, Lanzhou University Second Hospital, Key Laboratory of Gansu Province for Urological Diseases, Clinical Center of Gansu Province for Nephro-Urology, Lanzhou University, Lanzhou, Gansu Province, China
| | - Bin Xiong
- Department of Oncology, Lanzhou University Second Hospital, Lanzhou, Gansu Province, China
| | - Xiangxiang Zhang
- Department of Urology, Gansu Provincial Hospital, Lanzhou, Gansu Province, China
| | - Chaohu Chen
- Department of Urology, Lanzhou University Second Hospital, Key Laboratory of Gansu Province for Urological Diseases, Clinical Center of Gansu Province for Nephro-Urology, Lanzhou University, Lanzhou, Gansu Province, China
| | - Zhiqiang Yao
- Department of Urology, Lanzhou University Second Hospital, Key Laboratory of Gansu Province for Urological Diseases, Clinical Center of Gansu Province for Nephro-Urology, Lanzhou University, Lanzhou, Gansu Province, China
| | - Hao Wu
- Department of Urology, Lanzhou University Second Hospital, Key Laboratory of Gansu Province for Urological Diseases, Clinical Center of Gansu Province for Nephro-Urology, Lanzhou University, Lanzhou, Gansu Province, China
| | - Jinlong Cao
- Department of Urology, Lanzhou University Second Hospital, Key Laboratory of Gansu Province for Urological Diseases, Clinical Center of Gansu Province for Nephro-Urology, Lanzhou University, Lanzhou, Gansu Province, China
| | - Jianpeng Li
- Department of Urology, Lanzhou University Second Hospital, Key Laboratory of Gansu Province for Urological Diseases, Clinical Center of Gansu Province for Nephro-Urology, Lanzhou University, Lanzhou, Gansu Province, China
| | - Pan Li
- Department of Urology, Lanzhou University Second Hospital, Key Laboratory of Gansu Province for Urological Diseases, Clinical Center of Gansu Province for Nephro-Urology, Lanzhou University, Lanzhou, Gansu Province, China
| | - Zhiping Wang
- Department of Urology, Lanzhou University Second Hospital, Key Laboratory of Gansu Province for Urological Diseases, Clinical Center of Gansu Province for Nephro-Urology, Lanzhou University, Lanzhou, Gansu Province, China
| | - Junqiang Tian
- Department of Urology, Lanzhou University Second Hospital, Key Laboratory of Gansu Province for Urological Diseases, Clinical Center of Gansu Province for Nephro-Urology, Lanzhou University, Lanzhou, Gansu Province, China.
| |
Collapse
|
4
|
DeTomaso A, Kim H, Shauh J, Adulla A, Zigo S, Ghoul M, Presicce P, Kallapur SG, Goodman W, Tilburgs T, Way SS, Hackney D, Moore J, Mesiano S. Progesterone inactivation in decidual stromal cells: A mechanism for inflammation-induced parturition. Proc Natl Acad Sci U S A 2024; 121:e2400601121. [PMID: 38861608 PMCID: PMC11194587 DOI: 10.1073/pnas.2400601121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 04/25/2024] [Indexed: 06/13/2024] Open
Abstract
The process of human parturition involves inflammation at the interface where fetal chorion trophoblast cells interact with maternal decidual stromal (DS) cells and maternal immune cells in the decidua (endometrium of pregnancy). This study tested the hypothesis that inflammation at the chorion-decidua interface (CDI) induces labor by negating the capacity for progesterone (P4) to block labor and that this is mediated by inactivation of P4 in DS cells by aldo-keto reductase family 1 member C1 (AKR1C1). In human, Rhesus macaque, and mouse CDI, AKR1C1 expression increased in association with term and preterm labor. In a human DS cell line and in explant cultures of term human fetal membranes containing the CDI, the prolabor inflammatory cytokine, interleukin-1ß (IL-1ß), and media conditioned by LPS-stimulated macrophages increased AKR1C1 expression and coordinately reduced nuclear P4 levels and P4 responsiveness. Loss of P4 responsiveness was overcome by inhibition of AKR1C1 activity, inhibition of AKR1C1 expression, and bypassing AKR1C1 activity with a P4 analog that is not metabolized by AKR1C1. Increased P4 activity in response to AKR1C1 inhibition was prevented by the P4 receptor antagonist RU486. Pharmacologic inhibition of AKR1C1 activity prevented parturition in a mouse model of inflammation-induced preterm parturition. The data suggest that inflammatory stimuli at the CDI drive labor by inducing AKR1C1-mediated P4 inactivation in DS cells and that inhibiting and/or bypassing of AKR1C1-mediated P4 inactivation is a plausible therapeutic strategy to mitigate the risk of inflammation-associated preterm birth.
Collapse
Affiliation(s)
- Angela DeTomaso
- Department of Pathology, Case Western Reserve University, Cleveland, OH44106
- Department of Reproductive Biology, Case Western Reserve University, Cleveland, OH44106
| | - Hyeyon Kim
- Department of Reproductive Biology, Case Western Reserve University, Cleveland, OH44106
| | - Jacqueline Shauh
- Department of Reproductive Biology, Case Western Reserve University, Cleveland, OH44106
| | - Anika Adulla
- Department of Reproductive Biology, Case Western Reserve University, Cleveland, OH44106
| | - Sarah Zigo
- Department of Reproductive Biology, Case Western Reserve University, Cleveland, OH44106
| | - Maya Ghoul
- Department of Reproductive Biology, Case Western Reserve University, Cleveland, OH44106
| | - Pietro Presicce
- Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, CA90095
| | - Suhas G. Kallapur
- Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, CA90095
| | - Wendy Goodman
- Department of Pathology, Case Western Reserve University, Cleveland, OH44106
| | - Tamara Tilburgs
- Cincinnati Children’s Hospital, Center for Inflammation and Tolerance, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH45229
| | - Sing-Sing Way
- Cincinnati Children’s Hospital, Center for Inflammation and Tolerance, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH45229
| | - David Hackney
- Department of Reproductive Biology, Case Western Reserve University, Cleveland, OH44106
- Department of Obstetrics and Gynecology, University Hospitals, Cleveland, OH44106
| | - John Moore
- Department of Reproductive Biology, Case Western Reserve University, Cleveland, OH44106
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH44106
| | - Sam Mesiano
- Department of Reproductive Biology, Case Western Reserve University, Cleveland, OH44106
- Department of Obstetrics and Gynecology, University Hospitals, Cleveland, OH44106
| |
Collapse
|
5
|
Silva IMD, Vacario BGL, Okuyama NCM, Barcelos GRM, Fuganti PE, Guembarovski RL, Cólus IMDS, Serpeloni JM. Polymorphisms in drug-metabolizing genes and urinary bladder cancer susceptibility and prognosis: Possible impacts and future management. Gene 2024; 907:148252. [PMID: 38350514 DOI: 10.1016/j.gene.2024.148252] [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: 10/09/2023] [Revised: 01/22/2024] [Accepted: 02/05/2024] [Indexed: 02/15/2024]
Abstract
Epidemiological studies have shown the association of genetic variants with risks of occupational and environmentally induced cancers, including bladder (BC). The current review summarizes the effects of variants in genes encoding phase I and II enzymes in well-designed studies to highlight their contribution to BC susceptibility and prognosis. Polymorphisms in genes codifying drug-metabolizing proteins are of particular interest because of their involvement in the metabolism of exogenous genotoxic compounds, such as tobacco and agrochemicals. The prognosis between muscle-invasive and non-muscle-invasive diseases is very different, and it is difficult to predict which will progress worse. Web of Science, PubMed, and Medline were searched to identify studies published between January 1, 2010, and February 2023. We included 73 eligible studies, more than 300 polymorphisms, and 46 genes/loci. The most studied candidate genes/loci of phase I metabolism were CYP1B1, CYP1A1, CYP1A2, CYP3A4, CYP2D6, CYP2A6, CYP3E1, and ALDH2, and those in phase II were GSTM1, GSTT1, NAT2, GSTP1, GSTA1, GSTO1, and UGT1A1. We used the 46 genes to construct a network of proteins and to evaluate their biological functions based on the Reactome and KEGG databases. Lastly, we assessed their expression in different tissues, including normal bladder and BC samples. The drug-metabolizing pathway plays a relevant role in BC, and our review discusses a list of genes that could provide clues for further exploration of susceptibility and prognostic biomarkers.
Collapse
Affiliation(s)
- Isabely Mayara da Silva
- Department of General Biology, Center of Biological Sciences, State University of Londrina (UEL), Londrina 86057-970, Brazil.
| | - Beatriz Geovana Leite Vacario
- Department of General Biology, Center of Biological Sciences, State University of Londrina (UEL), Londrina 86057-970, Brazil; Center of Health Sciences, State University of West Paraná (UNIOESTE), Francisco Beltrão-Paraná, 85605-010, Brazil.
| | - Nádia Calvo Martins Okuyama
- Department of General Biology, Center of Biological Sciences, State University of Londrina (UEL), Londrina 86057-970, Brazil.
| | - Gustavo Rafael Mazzaron Barcelos
- Department of Biosciences, Institute for Health and Society, Federal University of São Paulo (UNIFESP), Santos 11.060-001, Brazil.
| | | | - Roberta Losi Guembarovski
- Department of General Biology, Center of Biological Sciences, State University of Londrina (UEL), Londrina 86057-970, Brazil.
| | - Ilce Mara de Syllos Cólus
- Department of General Biology, Center of Biological Sciences, State University of Londrina (UEL), Londrina 86057-970, Brazil.
| | - Juliana Mara Serpeloni
- Department of General Biology, Center of Biological Sciences, State University of Londrina (UEL), Londrina 86057-970, Brazil.
| |
Collapse
|
6
|
Park J, Ngo TH, Paudel SB, Kil YS, Ryoo GH, Jin CH, Choi HI, Jung CH, Na M, Nam JW, Han AR. Angular dihydropyranocoumarins from the flowers of Peucedanum japonicum and their aldo-keto reductase inhibitory activities. PHYTOCHEMISTRY 2024; 219:113974. [PMID: 38211847 DOI: 10.1016/j.phytochem.2024.113974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 01/04/2024] [Accepted: 01/06/2024] [Indexed: 01/13/2024]
Abstract
Twenty-one angular dihydropyranocoumarins and a linear furanocoumarin, including four previously undescribed compounds (1-4), were isolated from the flowers of Peucedanum japonicum (Umbelliferae). The structures of 1-4, along with their absolute stereochemistry, were determined to be (3'S,4'S)-3'-O-propanoyl-4'-O-(3‴-methyl-2‴-butenoyl)khellactone (1), (3'S,4'S)-3'-O-propanoyl-4'-O-(2‴-methyl-2‴Z-butenoyl)khellactone (2), (3'S,4'S)-3'-O-propanoyl-4'-O-(2‴-methylbutanoyl)khellactone (3), and (3'S,4'S)-3'-O-(2″-methylpropanoyl)-4'-O-(3‴-methyl-2‴-butenoyl)khellactone (4) using one- and two-dimensional nuclear magnetic resonance, high-resolution electrospray ionization mass spectroscopy, and electronic circular dichroism spectroscopy. In addition, the absolute configuration of the three angular dihydropyranocoumarins (5-7) was determined for the first time in this study. Among the previously reported compounds isolated in this study, 8 and 9 were isolated for the first time from the genus Peucedanum, whereas 10 and 11 were previously unreported and had not been isolated from P. japonicum to date. Furthermore, all isolated compounds were evaluated for their aldo-keto reductase 1C1 inhibitory activities on A549 human non-small-cell lung cancer cells. Compounds 10 and 12 exhibited substantial AKR1C1 inhibitory activities with IC50 values of 35.8 ± 0.9 and 44.2 ± 1.5 μM, respectively.
Collapse
Affiliation(s)
- Jisu Park
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup-si, Jeollabuk-do, 56212, Republic of Korea; College of Pharmacy, Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Trung Huy Ngo
- College of Pharmacy, Yeungnam University, Gyeongsan-si, Gyeongsangbuk-do, 38541, Republic of Korea
| | - Sunil Babu Paudel
- College of Pharmacy, Yeungnam University, Gyeongsan-si, Gyeongsangbuk-do, 38541, Republic of Korea
| | - Yun-Seo Kil
- College of Pharmacy, Yeungnam University, Gyeongsan-si, Gyeongsangbuk-do, 38541, Republic of Korea
| | - Ga-Hee Ryoo
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup-si, Jeollabuk-do, 56212, Republic of Korea
| | - Chang Hyun Jin
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup-si, Jeollabuk-do, 56212, Republic of Korea
| | - Hong-Il Choi
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup-si, Jeollabuk-do, 56212, Republic of Korea
| | - Chan-Hun Jung
- Jeonju AgroBio-Materials Institute, Jeonju-si, Jeollabuk-do, 54810, Republic of Korea
| | - MinKyun Na
- College of Pharmacy, Chungnam National University, Daejeon, 34134, Republic of Korea.
| | - Joo-Won Nam
- College of Pharmacy, Yeungnam University, Gyeongsan-si, Gyeongsangbuk-do, 38541, Republic of Korea.
| | - Ah-Reum Han
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup-si, Jeollabuk-do, 56212, Republic of Korea.
| |
Collapse
|
7
|
Fang J, Orobator ON, Olelewe C, Passeri G, Singh K, Awuah SG, Suntharalingam K. A Breast Cancer Stem Active Cobalt(III)-Cyclam Complex Containing Flufenamic Acid with Immunogenic Potential. Angew Chem Int Ed Engl 2024; 63:e202317940. [PMID: 38063406 PMCID: PMC10952489 DOI: 10.1002/anie.202317940] [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: 11/23/2023] [Indexed: 12/31/2023]
Abstract
The cytotoxic and immunogenic-activating properties of a cobalt(III)-cyclam complex bearing the non-steroidal anti-inflammatory drug, flufenamic acid is reported within the context of anti-cancer stem cell (CSC) drug discovery. The cobalt(III)-cyclam complex 1 displays sub-micromolar potency towards breast CSCs grown in monolayers, 24-fold and 31-fold greater than salinomycin (an established anti-breast CSC agent) and cisplatin (an anticancer metallopharmaceutical), respectively. Strikingly, the cobalt(III)-cyclam complex 1 is 69-fold and 50-fold more potent than salinomycin and cisplatin towards three-dimensionally cultured breast CSC mammospheres. Mechanistic studies reveal that 1 induces DNA damage, inhibits cyclooxygenase-2 expression, and prompts caspase-dependent apoptosis. Breast CSCs treated with 1 exhibit damage-associated molecular patterns characteristic of immunogenic cell death and are phagocytosed by macrophages. As far as we are aware, 1 is the first cobalt complex of any oxidation state or geometry to display both cytotoxic and immunogenic-activating effects on breast CSCs.
Collapse
Affiliation(s)
- Jiaxin Fang
- School of ChemistryUniversity of LeicesterLeicesterUK
| | | | | | | | - Kuldip Singh
- School of ChemistryUniversity of LeicesterLeicesterUK
| | - Samuel G. Awuah
- Department of ChemistryUniversity of KentuckyLexingtonKYUSA
- Department of Pharmaceutical SciencesUniversity of KentuckyLexingtonKYUSA
| | | |
Collapse
|
8
|
Kikuchi H, Maishi N, Yu L, Jia Z, Li C, Sato M, Takeda R, Ishizuka K, Hida Y, Shinohara N, Hida K. Low-dose metronomic cisplatin as an antiangiogenic and anti-inflammatory strategy for cancer. Br J Cancer 2024; 130:336-345. [PMID: 38036665 PMCID: PMC10803316 DOI: 10.1038/s41416-023-02498-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 10/28/2023] [Accepted: 11/06/2023] [Indexed: 12/02/2023] Open
Abstract
BACKGROUND Conventional chemotherapy is based on the maximum tolerated dose (MTD) and requires treatment-free intervals to restore normal host cells. MTD chemotherapy may induce angiogenesis or immunosuppressive cell infiltration during treatment-free intervals. Low-dose metronomic (LDM) chemotherapy is defined as frequent administration at lower doses and causes less inflammatory change, whereas MTD chemotherapy induces an inflammatory change. Although several LDM regimens have been applied, LDM cisplatin (CDDP) has been rarely reported. This study addressed the efficacy of LDM CDDP on tumour endothelial cell phenotypic alteration compared to MTD CDDP. METHODS Tumour growth and metastasis were assessed in bladder cancer-bearing mice treated with LDM or MTD gemcitabine (GEM) and CDDP. To elucidate the therapeutic effects of LDM CDDP, the change of tumour vasculature, tumour-infiltrating immune cells and inflammatory changes were evaluated by histological analysis and mRNA expression in tumour tissues. RESULTS Tumour growth and bone metastasis were more suppressed by LDM CDDP + MTD GEM treatment than MTD CDDP + MTD GEM. Myeloid-derived suppressor cell accumulation was reduced by LDM CDDP, whereas inflammatory change was induced in the tumour microenvironment by MTD CDDP. CONCLUSION LDM CDDP does not cause inflammatory change unlike MTD CDDP, suggesting that it is a promising strategy in chemotherapy.
Collapse
Affiliation(s)
- Hiroshi Kikuchi
- Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
- Department of Renal and Genitourinary Surgery, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Nako Maishi
- Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
- Department of Vascular Biology and Molecular Pathology, Hokkaido University Faculty of Dental Medicine, Sapporo, Japan
| | - Li Yu
- Department of Vascular Biology and Molecular Pathology, Hokkaido University Faculty of Dental Medicine, Sapporo, Japan
| | - Zi Jia
- Department of Vascular Biology and Molecular Pathology, Hokkaido University Faculty of Dental Medicine, Sapporo, Japan
| | - Cong Li
- Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
- Department of Vascular Biology and Molecular Pathology, Hokkaido University Faculty of Dental Medicine, Sapporo, Japan
| | - Masumi Sato
- Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Ryo Takeda
- Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
- Department of Vascular Biology and Molecular Pathology, Hokkaido University Faculty of Dental Medicine, Sapporo, Japan
| | - Keita Ishizuka
- Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Yasuhiro Hida
- Department of Cardiovascular and Thoracic Surgery, Faculty of Medicine, Hokkaido University, Sapporo, Japan
- Advanced Robotic and Endoscopic Surgery, School of Medicine, Fujita Health University, Toyoake, Japan
| | - Nobuo Shinohara
- Department of Renal and Genitourinary Surgery, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Kyoko Hida
- Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan.
- Department of Vascular Biology and Molecular Pathology, Hokkaido University Faculty of Dental Medicine, Sapporo, Japan.
| |
Collapse
|
9
|
Mehmandar-Oskuie A, Tohidfar M, Hajikhani B, Karimi F. Anticancer effects of cell-free culture supernatant of Escherichia coli in bladder cancer cell line: New insight into the regulation of inflammation. Gene 2023; 889:147795. [PMID: 37708921 DOI: 10.1016/j.gene.2023.147795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 08/23/2023] [Accepted: 09/11/2023] [Indexed: 09/16/2023]
Abstract
Bladder cancer (BC) is the 10th most common malignancy in worldwide, with substantial mortality and morbidity if not treated effectively. According to various research, inflammatory circumstances majorly impact the microenvironment of bladder cancer, and the chronic presence of cytokines and chemokines promotes tumor progression. In this investigation, we explored the impact of cell-free culture supernatant ofEscherichia colistrain 536 on inflammatory cytokines and chemokines in bladder cancer model microarray data (GSE162251). Then we examined in silico outcomes on human bladder cancer cell line 5637 to verify and extrapolate findings. This investigation revealed for the first time that this compound has potent suppressor effects on interleukin 1 beta (IL-1β), C-C motif chemokine ligand 2 (CCL2), and C-X3-C motif chemokine ligand 1 (CX3CL1) gene expression as well as increased NAD(P)H quinone dehydrogenase 1 (NQO1), as an anti-oxidant agent, gene expression in 4, 8, and 24 h. Moreover, we confirmed that c-MYC, a member of the MYC proto-oncogene family, gene expression reduced in 5637 cells in 4 h and then followed up its expression in 8 and 24 h. In addition, our investigation demonstrated that the supernatant raised the BCL2-Associated X Protein/B-cell lymphoma 2 (BAX/BCL2) ratio, and subsequent flow cytometry analysis demonstrated that the supernatant induction apoptosis and necrosis. In conclusion, our findings demonstrate that this compound is a potential candidate for the suppression of bladder cancer progression.
Collapse
Affiliation(s)
- Amirreza Mehmandar-Oskuie
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Masoud Tohidfar
- Department of Cell & Molecular Biology, Faculty of Life Sciences & Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Bahareh Hajikhani
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Forouzan Karimi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
10
|
Bazargan S, Bunch B, Ojwang‘ AME, Blauvelt J, Landin A, Ali J, Abrahams D, Cox C, Hall AM, Beatty MS, Poch M, Rejniak KA, Pilon-Thomas S. Targeting myeloid-derived suppressor cells with gemcitabine to enhance efficacy of adoptive cell therapy in bladder cancer. Front Immunol 2023; 14:1275375. [PMID: 37901214 PMCID: PMC10602731 DOI: 10.3389/fimmu.2023.1275375] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 09/28/2023] [Indexed: 10/31/2023] Open
Abstract
Background New therapeutics in development for bladder cancer need to address the recalcitrant nature of the disease. Intravesical adoptive cell therapy (ACT) with tumor infiltrating lymphocytes (TIL) can potentially induce durable responses in bladder cancer while maximizing T cells at the tumor site. T cells infused into the bladder directly encounter immunosuppressive populations, such as myeloid derived suppressor cells (MDSCs), that can attenuate T cell responses. Intravesical instillation of gemcitabine can be used as a lymphodepleting agent to precondition the bladder microenvironment for infused T cell products. Methods Urine samples from bladder cancer patients and healthy donors were analyzed by flow cytometry and cytometric bead array for immune profiling and cytokine quantification. MDSCs were isolated from the urine and cocultured with stimulated T cells to assess effects on proliferation. An orthotopic murine model of bladder cancer was established using the MB49-OVA cell line and immune profiling was performed. MDSCs from tumor-bearing mice were cocultured with OT-I splenocytes to assess T cell proliferation. Mice received intravesical instillation of gemcitabine and depletion of immune cells was measured via flow cytometry. Bladder tumor growth of mice treated with intravesical gemcitabine, OT-I transgenic T cells, or combination was monitored via ultrasound measurement. Results In comparison to healthy donors, urine specimen from bladder cancer patients show high levels of MDSCs and cytokines associated with myeloid chemotaxis, T cell chemotaxis, and inflammation. T cells isolated from healthy donors were less proliferative when cocultured with MDSCs from the urine. Orthotopic murine bladder tumors also presented with high levels of MDSCs along with enrichment of cytokines found in the patient urine samples. MDSCs isolated from spleens of tumor-bearing mice exerted suppressive effects on the proliferation of OT-I T cells. Intravesical instillation of gemcitabine reduced overall immune cells, MDSCs, and T cells in orthotopic bladder tumors. Combination treatment with gemcitabine and OT-I T cells resulted in sustained anti-tumor responses in comparison to monotherapy treatments. Conclusion MDSCs are enriched within the microenvironment of bladder tumors and are suppressive to T cells. Gemcitabine can be used to lymphodeplete bladder tumors and precondition the microenvironment for intravesical ACT.
Collapse
Affiliation(s)
- Sarah Bazargan
- Department of Immunology, Moffitt Cancer Center, Tampa, FL, United States
| | - Brittany Bunch
- Department of Immunology, Moffitt Cancer Center, Tampa, FL, United States
| | | | - Jamie Blauvelt
- Department of Immunology, Moffitt Cancer Center, Tampa, FL, United States
| | - Annick Landin
- Department of Immunology, Moffitt Cancer Center, Tampa, FL, United States
| | - Johannes Ali
- Department of Immunology, Moffitt Cancer Center, Tampa, FL, United States
| | - Dominique Abrahams
- Comparative Medicine, University of South Florida, Tampa, FL, United States
| | - Cheryl Cox
- Cell Therapy Facility, Moffitt Cancer Center, Tampa, FL, United States
| | - Amy M. Hall
- Department of Immunology, Moffitt Cancer Center, Tampa, FL, United States
| | - Matthew S. Beatty
- Department of Immunology, Moffitt Cancer Center, Tampa, FL, United States
| | - Michael Poch
- Department of Genitourinary Oncology, Moffitt Cancer Center, Tampa, FL, United States
| | - Katarzyna A. Rejniak
- Department of Integrated Mathematical Oncology, Moffitt Cancer Center, Tampa, FL, United States
| | - Shari Pilon-Thomas
- Department of Immunology, Moffitt Cancer Center, Tampa, FL, United States
- Department of Genitourinary Oncology, Moffitt Cancer Center, Tampa, FL, United States
| |
Collapse
|
11
|
Cataldo ML, De Placido P, Esposito D, Formisano L, Arpino G, Giuliano M, Bianco R, De Angelis C, Veneziani BM. The effect of the alpha-specific PI3K inhibitor alpelisib combined with anti-HER2 therapy in HER2+/PIK3CA mutant breast cancer. Front Oncol 2023; 13:1108242. [PMID: 37469415 PMCID: PMC10353540 DOI: 10.3389/fonc.2023.1108242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 06/15/2023] [Indexed: 07/21/2023] Open
Abstract
Background HER2 is amplified or overexpressed in around 20% of breast cancers (BC). HER2-targeted therapies have significantly improved the prognosis of patients with HER2+ BC, however, de novo and acquired resistance to anti-HER2 treatment is common. Activating mutations in the PIK3CA gene are reported in ∼30% of HER2+ BC and are associated with resistance to anti-HER2 therapies and a poor prognosis. Here, we investigated the in vitro and in vivo antitumor efficacy of the alpha-specific PI3K inhibitor alpelisib alone or in combination with anti-HER2 therapy using a panel of HER2+ BC cell lines. We also generated models of acquired resistance to alpelisib to investigate the mechanisms underlying resistance to alpha-specific PI3K inhibition. Materials and methods PIK3CA mutant (HCC1954, KPL4 and JMT1) and wild-type (BT474 and SKBR3) HER2+ BC cell lines were used. The HCC1954 and KPL4 cells were chronically exposed to increasing concentrations of alpelisib or to alpelisib + trastuzumab in order to generate derivatives with acquired resistance to alpelisib (AR) and to alpelisib + trastuzumab (ATR). The transcriptomic profiles of HCC1954, KPL4 and their AR and ATR derivatives were determined by RNA sequencing. Cell growth was assessed by MTT assay. Changes in the protein levels of key PI3K pathway components were assessed by Western blotting. Gene expression, cellular and patients' data from the Cancer Dependency Map (DepMap) and KMPlot datasets were interrogated. Results HER2+ BC cell lines harboring activating mutations in PIK3CA were less sensitive to single or dual anti-HER2 blockade compared to PIK3CA wild-type cells. Alpelisib treatment resulted in dose-dependent inhibition of the growth of cells with or without PIK3CA mutations and enhanced the antitumor efficacy of anti-HER2 therapies in vitro. In addition, alpelisib greatly delayed tumor growth of HCC1954 xenografts in vivo. Functional annotation of the significantly differentially expressed genes suggested the common activation of biological processes associated with oxidation reduction, cell proliferation, immune response and RNA synthesis in alpelisib-resistant models compared with native cells. Eight commonly upregulated genes (log2 fold-change >1, False Discovery Rate [FDR] <0.05) in models with acquired resistance to alpelisib or alpelisib + trastuzumab were identified. Among these, AKR1C1 was associated with alpelisib-resistance in vitro and with a poor prognosis in patients with HER2+ BC. Conclusions Our findings support the use of an alpha-selective PI3K inhibitor to overcome the therapeutic limitations associated with single or dual HER2 blockade in PIK3CA-mutant HER2+ breast cancer. Future studies are warranted to confirm the potential role of candidate genes/pathways in resistance to alpelisib.
Collapse
Affiliation(s)
- Maria Letizia Cataldo
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Pietro De Placido
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Daniela Esposito
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Luigi Formisano
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Grazia Arpino
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Mario Giuliano
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Roberto Bianco
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Carmine De Angelis
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Bianca Maria Veneziani
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| |
Collapse
|
12
|
Nie Z, Gao Y, Chen M, Peng Y, Guo N, Cao H, Huang D, Gao X, Zhang S. Genome-Wide Screening Identifies Gene AKR1C1 Critical for Resistance to Pirarubicin in Bladder Cancer. Cancers (Basel) 2023; 15:cancers15092487. [PMID: 37173953 PMCID: PMC10177075 DOI: 10.3390/cancers15092487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/21/2023] [Accepted: 04/24/2023] [Indexed: 05/15/2023] Open
Abstract
Non-muscle-invasive bladder cancer (NMIBC) is a common tumor of the urinary system. Given its high rates of recurrence, progression, and drug resistance, NMIBC seriously affects the quality of life and limits the survival time of patients. Pirarubicin (THP) is a bladder infusion chemotherapy drug recommended by the guidelines for NMIBC. Although the widespread use of THP reduces the recurrence rate of NMIBC, 10-50% of patients still suffer from tumor recurrence, which is closely related to tumor resistance to chemotherapy drugs. This study was performed to screen the critical genes causing THP resistance in bladder cancer cell lines by using the CRISPR/dCas9-SAM system. Thus, AKR1C1 was screened. Results showed that the high expression of AKR1C1 could enhance the drug resistance of bladder cancer to THP both in vivo and in vitro. This gene could reduce the levels of 4-hydroxynonenal and reactive oxygen species (ROS) and resist THP-induced apoptosis. However, AKR1C1 did not affect the proliferation, invasion, or migration of the bladder cancer cells. Aspirin, which is an AKR1C1 inhibitor, could help reduce the drug resistance caused by AKR1C1. After receiving THP treatment, the bladder cancer cell lines could upregulate the expression of the AKR1C1 gene through the ROS/KEAP1/NRF2 pathway, leading to resistance to THP treatment. Using tempol, which is an inhibitor of ROS, could prevent the upregulation of AKR1C1 expression.
Collapse
Affiliation(s)
- Zhenyu Nie
- Central Laboratory, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou 570208, China
| | - Yuanhui Gao
- Central Laboratory, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou 570208, China
| | - Mei Chen
- Central Laboratory, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou 570208, China
| | - Yanling Peng
- Central Laboratory, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou 570208, China
| | - Na Guo
- Central Laboratory, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou 570208, China
| | - Hui Cao
- Central Laboratory, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou 570208, China
| | - Denggao Huang
- Central Laboratory, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou 570208, China
| | - Xin Gao
- Graduate School of Chinese Academy of Medical Sciences & Peking Union Medical College, Tsinghua University, Beijing 100190, China
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Shufang Zhang
- Central Laboratory, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou 570208, China
| |
Collapse
|
13
|
Zhang Z, Li X, Wang Y, Wei Y, Wei X. Involvement of inflammasomes in tumor microenvironment and tumor therapies. J Hematol Oncol 2023; 16:24. [PMID: 36932407 PMCID: PMC10022228 DOI: 10.1186/s13045-023-01407-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 02/08/2023] [Indexed: 03/19/2023] Open
Abstract
Inflammasomes are macromolecular platforms formed in response to damage-associated molecular patterns (DAMPs) and pathogen-associated molecular patterns, whose formation would cause maturation of interleukin-1 (IL-1) family members and gasdermin D (GSDMD), leading to IL-1 secretion and pyroptosis respectively. Several kinds of inflammasomes detecting different types of dangers have been found. The activation of inflammasomes is regulated at both transcription and posttranscription levels, which is crucial in protecting the host from infections and sterile insults. Present findings have illustrated that inflammasomes are involved in not only infection but also the pathology of tumors implying an important link between inflammation and tumor development. Generally, inflammasomes participate in tumorigenesis, cell death, metastasis, immune evasion, chemotherapy, target therapy, and radiotherapy. Inflammasome components are upregulated in some tumors, and inflammasomes can be activated in cancer cells and other stromal cells by DAMPs, chemotherapy agents, and radiation. In some cases, inflammasomes inhibit tumor progression by initiating GSDMD-mediated pyroptosis in cancer cells and stimulating IL-1 signal-mediated anti-tumor immunity. However, IL-1 signal recruits immunosuppressive cell subsets in other cases. We discuss the conflicting results and propose some possible explanations. Additionally, we also summarize interventions targeting inflammasome pathways in both preclinical and clinical stages. Interventions targeting inflammasomes are promising for immunotherapy and combination therapy.
Collapse
Affiliation(s)
- Ziqi Zhang
- grid.13291.380000 0001 0807 1581Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041 Sichuan People’s Republic of China
| | - Xue Li
- grid.13291.380000 0001 0807 1581Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041 Sichuan People’s Republic of China
| | - Yang Wang
- grid.13291.380000 0001 0807 1581Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041 Sichuan People’s Republic of China
| | - Yuquan Wei
- grid.13291.380000 0001 0807 1581Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041 Sichuan People’s Republic of China
| | - Xiawei Wei
- grid.13291.380000 0001 0807 1581Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041 Sichuan People’s Republic of China
| |
Collapse
|
14
|
Synthesis, Characterization and Biological Investigation of the Platinum(IV) Tolfenamato Prodrug–Resolving Cisplatin-Resistance in Ovarian Carcinoma Cell Lines. Int J Mol Sci 2023; 24:ijms24065718. [PMID: 36982792 PMCID: PMC10056020 DOI: 10.3390/ijms24065718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/10/2023] [Accepted: 03/13/2023] [Indexed: 03/19/2023] Open
Abstract
The research on the anticancer potential of platinum(IV) complexes represents one strategy to circumvent the deficits of approved platinum(II) drugs. Regarding the role of inflammation during carcinogenesis, the effects of non-steroidal anti-inflammatory drug (NSAID) ligands on the cytotoxicity of platinum(IV) complexes is of special interest. The synthesis of cisplatin- and oxaliplatin-based platinum(IV) complexes with four different NSAID ligands is presented in this work. Nine platinum(IV) complexes were synthesized and characterized by use of nuclear magnetic resonance (NMR) spectroscopy (1H, 13C, 195Pt, 19F), high-resolution mass spectrometry, and elemental analysis. The cytotoxic activity of eight compounds was evaluated for two isogenic pairs of cisplatin-sensitive and -resistant ovarian carcinoma cell lines. Platinum(IV) fenamato complexes with a cisplatin core showed especially high in vitro cytotoxicity against the tested cell lines. The most promising complex, 7, was further analyzed for its stability in different buffer solutions and behavior in cell cycle and cell death experiments. Compound 7 induces a strong cytostatic effect and cell line-dependent early apoptotic or late necrotic cell death processes. Gene expression analysis suggests that compound 7 acts through a stress-response pathway integrating p21, CHOP, and ATF3.
Collapse
|
15
|
SUN TINGTING, SUN XUE, WANG XIN, GUO RUI, YU YUANHUA, GAO LE. Analysis of the mechanism of aldo-keto reductase dependent cis-platin resistance in HepG2 cells based on transcriptomic and NADH metabolic state. BIOCELL 2023. [DOI: 10.32604/biocell.2023.026229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/11/2023]
|
16
|
Sah DK, Khoi PN, Li S, Arjunan A, Jeong JU, Jung YD. (-)-Epigallocatechin-3-Gallate Prevents IL-1β-Induced uPAR Expression and Invasiveness via the Suppression of NF-κB and AP-1 in Human Bladder Cancer Cells. Int J Mol Sci 2022; 23:ijms232214008. [PMID: 36430487 PMCID: PMC9697952 DOI: 10.3390/ijms232214008] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 11/07/2022] [Accepted: 11/10/2022] [Indexed: 11/16/2022] Open
Abstract
(-)-Epigallocatechin-3-O-gallate (EGCG), a primary green tea polyphenol, has powerful iron scavengers, belongs to the family of flavonoids with antioxidant properties, and can be used to prevent cancer. Urokinase-type plasminogen activator receptors (uPARs) are glycosylphosphatidylinositol (GPI)-anchored cell membrane receptors that have crucial roles in cell invasion and metastasis of several cancers including bladder cancer. The mechanism of action of EGCG on uPAR expression has not been reported clearly yet. In this study, we investigated the effect of EGCG on interleukin (IL)-1β-induced cell invasion and uPAR activity in T24 human bladder cancer cells. Interestingly, nuclear factor (NF)-κB and activator protein (AP)-1 transcription factors were critically required for IL-1β-induced high uPAR expression, and EGCG suppressed the transcriptional activity of both the ERK1/2 and JNK signaling pathways with the AP-1 subunit c-Jun. EGCG blocked the IL-1β-stimulated reactive oxygen species (ROS) production, in turn suppressing NF-κB signaling and anti-invasion effects by inhibiting uPAR expression. These results suggest that EGCG may exert at least part of its anticancer effect by controlling uPAR expression through the suppression of ERK1/2, JNK, AP-1, and NF-κB.
Collapse
Affiliation(s)
- Dhiraj Kumar Sah
- Department of Biochemistry, Chonnam National University Medical School, Hwasun 58128, Republic of Korea
| | - Pham Ngoc Khoi
- Faculty of Basic Medical Sciences, Pham Ngoc Thach University of Medicine, Ho Chi Minh City 740500, Vietnam
| | - Shinan Li
- Department of Biochemistry, Chonnam National University Medical School, Hwasun 58128, Republic of Korea
| | - Archana Arjunan
- Department of Biochemistry, Chonnam National University Medical School, Hwasun 58128, Republic of Korea
| | - Jae-Uk Jeong
- Department of Radiation Oncology, Chonnam National University Medical School, Hwasun 58128, Republic of Korea
| | - Young Do Jung
- Department of Biochemistry, Chonnam National University Medical School, Hwasun 58128, Republic of Korea
- Correspondence:
| |
Collapse
|
17
|
Park H, Imoto S, Miyano S. PredictiveNetwork: predictive gene network estimation with application to gastric cancer drug response-predictive network analysis. BMC Bioinformatics 2022; 23:342. [PMID: 35974335 PMCID: PMC9380306 DOI: 10.1186/s12859-022-04871-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 08/02/2022] [Indexed: 11/22/2022] Open
Abstract
Background Gene regulatory networks have garnered a large amount of attention to understand disease mechanisms caused by complex molecular network interactions. These networks have been applied to predict specific clinical characteristics, e.g., cancer, pathogenicity, and anti-cancer drug sensitivity. However, in most previous studies using network-based prediction, the gene networks were estimated first, and predicted clinical characteristics based on pre-estimated networks. Thus, the estimated networks cannot describe clinical characteristic-specific gene regulatory systems. Furthermore, existing computational methods were developed from algorithmic and mathematics viewpoints, without considering network biology. Results To effectively predict clinical characteristics and estimate gene networks that provide critical insights into understanding the biological mechanisms involved in a clinical characteristic, we propose a novel strategy for predictive gene network estimation. The proposed strategy simultaneously performs gene network estimation and prediction of the clinical characteristic. In this strategy, the gene network is estimated with minimal network estimation and prediction errors. We incorporate network biology by assuming that neighboring genes in a network have similar biological functions, while hub genes play key roles in biological processes. Thus, the proposed method provides interpretable prediction results and enables us to uncover biologically reliable marker identification. Monte Carlo simulations shows the effectiveness of our method for feature selection in gene estimation and prediction with excellent prediction accuracy. We applied the proposed strategy to construct gastric cancer drug-responsive networks. Conclusion We identified gastric drug response predictive markers and drug sensitivity/resistance-specific markers, AKR1B10, AKR1C3, ANXA10, and ZNF165, based on GDSC data analysis. Our results for identifying drug sensitive and resistant specific molecular interplay are strongly supported by previous studies. We expect that the proposed strategy will be a useful tool for uncovering crucial molecular interactions involved a specific biological mechanism, such as cancer progression or acquired drug resistance. Supplementary Information The online version contains supplementary material available at 10.1186/s12859-022-04871-z.
Collapse
Affiliation(s)
- Heewon Park
- M&D Data Science Center, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, Japan.
| | - Seiya Imoto
- Human Genome Center, Institute of Medical Science, University of Tokyo, 4-6-1 Shirokane-dai, Minato-ku, Tokyo, Japan
| | - Satoru Miyano
- M&D Data Science Center, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, Japan.,Human Genome Center, Institute of Medical Science, University of Tokyo, 4-6-1 Shirokane-dai, Minato-ku, Tokyo, Japan
| |
Collapse
|
18
|
Kljun J, Pavlič R, Hafner E, Lipec T, Moreno-Da Silva S, Tič P, Turel I, Büdefeld T, Stojan J, Rižner TL. Ruthenium complexes show potent inhibition of AKR1C1, AKR1C2, and AKR1C3 enzymes and anti-proliferative action against chemoresistant ovarian cancer cell line. Front Pharmacol 2022; 13:920379. [PMID: 36034868 PMCID: PMC9403717 DOI: 10.3389/fphar.2022.920379] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 06/28/2022] [Indexed: 12/04/2022] Open
Abstract
In this study, we present the synthesis, kinetic studies of inhibitory activity toward aldo-keto reductase 1C (AKR1C) enzymes, and anticancer potential toward chemoresistant ovarian cancer of 10 organoruthenium compounds bearing diketonate (1–6) and hydroxyquinolinate (7–10) chelating ligands with the general formula [(η6-p-cymene)Ru(chel)(X)]n+ where chel represents the chelating ligand and X the chlorido or pta ligand. Our studies show that these compounds are potent inhibitors of the AKR enzymes with an uncommon inhibitory mechanism, where two inhibitor molecules bind to the enzyme in a first fast and reversible step and a second slower and irreversible step. The binding potency of each step is dependent on the chemical structure of the monodentate ligands in the metalloinhibitors with the chlorido complexes generally acting as reversible inhibitors and pta complexes as irreversible inhibitors. Our study also shows that compounds 1–9 have a moderate yet better anti-proliferative and anti-migration action on the chemoresistant ovarian cancer cell line COV362 compared to carboplatin and similar effects to cisplatin.
Collapse
Affiliation(s)
- Jakob Kljun
- Department of Chemistry and Biochemistry, Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
| | - Renata Pavlič
- Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Eva Hafner
- Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Tanja Lipec
- Department of Chemistry and Biochemistry, Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
| | - Sara Moreno-Da Silva
- Department of Chemistry and Biochemistry, Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
- Faculty of Chemical Sciences, Universidad Complutense de Madrid, Madrid, Spain
| | - Primož Tič
- Department of Chemistry and Biochemistry, Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
| | - Iztok Turel
- Department of Chemistry and Biochemistry, Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
- *Correspondence: Iztok Turel, ; Tea Lanišnik Rižner,
| | - Tomaž Büdefeld
- Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Jure Stojan
- Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Tea Lanišnik Rižner
- Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
- *Correspondence: Iztok Turel, ; Tea Lanišnik Rižner,
| |
Collapse
|
19
|
Zhao X, Cui L, Zhang Y, Guo C, Deng L, Wen Z, Lu Z, Shi X, Xing H, Liu Y, Zhang Y. Screening for Potential Therapeutic Agents for Non-Small Cell Lung Cancer by Targeting Ferroptosis. Front Mol Biosci 2022; 9:917602. [PMID: 36203872 PMCID: PMC9532010 DOI: 10.3389/fmolb.2022.917602] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 06/23/2022] [Indexed: 11/13/2022] Open
Abstract
Ferroptosis is a form of non-apoptotic and iron-dependent cell death originally identified in cancer cells. Recently, emerging evidence showed that ferroptosis-targeting therapy could be a novel promising anti-tumour treatment. However, systematic analyses of ferroptosis-related genes for the prognosis of non-small cell lung cancer (NSCLC) and the development of antitumor drugs exploiting the ferroptosis process remain rare. This study aimed to identify genes related to ferroptosis and NSCLC and to initially screen lead compounds that induce ferroptosis in tumor cells. We downloaded mRNA expression profiles and NSCLC clinical data from The Cancer Genome Atlas database to explore the prognostic role of ferroptosis-related genes. Four prognosis-associated ferroptosis-related genes were screened using univariate Cox regression analysis and the lasso Cox regression analysis, which could divide patients with NSCLC into high- and low-risk groups. Then, based on differentially expressed risk- and ferroptosis-related genes, the negatively correlated lead compound flufenamic acid (FFA) was screened through the Connective Map database. This project confirmed that FFA induced ferroptosis in A549 cells and inhibited growth and migration in a dose-dependent manner through CCK-8, scratch, and immunofluorescence assays. In conclusion, targeting ferroptosis might be a therapeutic alternative for NSCLC.
Collapse
Affiliation(s)
- Xin Zhao
- Department of Pharmacology, Shanxi Medical University, Taiyuan, China
| | - Lijuan Cui
- Department of Pharmacology, Shanxi Medical University, Taiyuan, China
| | - Yushan Zhang
- Department of Pharmacology, Shanxi Medical University, Taiyuan, China
| | - Chao Guo
- Department of Pharmacology, Shanxi Medical University, Taiyuan, China
| | - Lijiao Deng
- Department of Pharmacology, Shanxi Medical University, Taiyuan, China
| | - Zhitong Wen
- Department of Pharmacology, Shanxi Medical University, Taiyuan, China
| | - Zhihong Lu
- Department of Pharmacology, Shanxi Medical University, Taiyuan, China
- Department of Endocrinology, First Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan, China
| | - Xiaoyuan Shi
- Department of Pharmacology, Shanxi Medical University, Taiyuan, China
| | - Haojie Xing
- Department of Pharmacology, Shanxi Medical University, Taiyuan, China
| | - Yunfeng Liu
- Department of Endocrinology, First Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan, China
- *Correspondence: Yunfeng Liu, ; Yi Zhang, , orcid.org/0000-0003-0305-3127
| | - Yi Zhang
- Department of Pharmacology, Shanxi Medical University, Taiyuan, China
- *Correspondence: Yunfeng Liu, ; Yi Zhang, , orcid.org/0000-0003-0305-3127
| |
Collapse
|
20
|
Singh K, Rustagi Y, Abouhashem AS, Tabasum S, Verma P, Hernandez E, Pal D, Khona DK, Mohanty SK, Kumar M, Srivastava R, Guda PR, Verma SS, Mahajan S, Killian JA, Walker LA, Ghatak S, Mathew-Steiner SS, Wanczyk K, Liu S, Wan J, Yan P, Bundschuh R, Khanna S, Gordillo GM, Murphy MP, Roy S, Sen CK. Genome-wide DNA hypermethylation opposes healing in chronic wound patients by impairing epithelial-to-mesenchymal transition. J Clin Invest 2022; 132:157279. [PMID: 35819852 PMCID: PMC9433101 DOI: 10.1172/jci157279] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 07/07/2022] [Indexed: 12/15/2022] Open
Abstract
An extreme chronic wound tissue microenvironment causes epigenetic gene silencing. An unbiased whole-genome methylome was studied in the wound-edge tissue of patients with chronic wounds. A total of 4,689 differentially methylated regions (DMRs) were identified in chronic wound-edge skin compared with unwounded human skin. Hypermethylation was more frequently observed (3,661 DMRs) in the chronic wound-edge tissue compared with hypomethylation (1,028 DMRs). Twenty-six hypermethylated DMRs were involved in epithelial-mesenchymal transition (EMT). Bisulfite sequencing validated hypermethylation of a predicted specific upstream regulator TP53. RNA-Seq analysis was performed to qualify findings from methylome analysis. Analysis of the downregulated genes identified the TP53 signaling pathway as being significantly silenced. Direct comparison of hypermethylation and downregulated genes identified 4 genes, ADAM17, NOTCH, TWIST1, and SMURF1, that functionally represent the EMT pathway. Single-cell RNA-Seq studies revealed that these effects on gene expression were limited to the keratinocyte cell compartment. Experimental murine studies established that tissue ischemia potently induces wound-edge gene methylation and that 5′-azacytidine, inhibitor of methylation, improved wound closure. To specifically address the significance of TP53 methylation, keratinocyte-specific editing of TP53 methylation at the wound edge was achieved by a tissue nanotransfection-based CRISPR/dCas9 approach. This work identified that reversal of methylation-dependent keratinocyte gene silencing represents a productive therapeutic strategy to improve wound closure.
Collapse
Affiliation(s)
- Kanhaiya Singh
- Department of Surgery, Indiana University School of Medicine, Indianapolis, United States of America
| | - Yashika Rustagi
- Department of Surgery, Indiana University School of Medicine, Indianapolis, United States of America
| | - Ahmed S Abouhashem
- Department of Surgery, Indiana University School of Medicine, Indianapolis, United States of America
| | - Saba Tabasum
- Department of Surgery, Indiana University School of Medicine, Indianapolis, United States of America
| | - Priyanka Verma
- Department of Surgery, Indiana University School of Medicine, Indianapolis, United States of America
| | - Edward Hernandez
- Department of Surgery, Indiana University School of Medicine, Indianapolis, United States of America
| | - Durba Pal
- Department of Biomedical Engineering, Indian Institute of Technology Ropar, Ropar, India
| | - Dolly K Khona
- Department of Surgery, Indiana University School of Medicine, Indianapolis, United States of America
| | - Sujit K Mohanty
- Department of Surgery, Indiana University School of Medicine, Indianapolis, United States of America
| | - Manishekhar Kumar
- Department of Surgery, Indiana University School of Medicine, Indianapolis, United States of America
| | - Rajneesh Srivastava
- Department of Surgery, Indiana University School of Medicine, Indianapolis, United States of America
| | - Poornachander R Guda
- Department of Surgery, Indiana University School of Medicine, Indianapolis, United States of America
| | - Sumit S Verma
- Department of Surgery, Indiana University School of Medicine, Indianapolis, United States of America
| | - Sanskruti Mahajan
- Department of Surgery, Indiana University School of Medicine, Indianapolis, United States of America
| | - Jackson A Killian
- Department of Physics, Ohio State University, Columbus, United States of America
| | - Logan A Walker
- Department of Physics, Ohio State University, Columbus, United States of America
| | - Subhadip Ghatak
- Department of Surgery, Indiana University School of Medicine, Indianapolis, United States of America
| | - Shomita S Mathew-Steiner
- Department of Surgery, Indiana University School of Medicine, Indianapolis, United States of America
| | - Kristen Wanczyk
- Department of Surgery, Indiana University School of Medicine, Indianapolis, United States of America
| | - Sheng Liu
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, United States of America
| | - Jun Wan
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, United States of America
| | - Pearlly Yan
- Comprehensive Cancer Center, Ohio State University, Columbus, United States of America
| | - Ralf Bundschuh
- Department of Physics, Ohio State University, Columbus, United States of America
| | - Savita Khanna
- Department of Surgery, Indiana University School of Medicine, Indianapolis, United States of America
| | - Gayle M Gordillo
- Department of Surgery, Indiana University School of Medicine, Indianapolis, United States of America
| | - Michael P Murphy
- Department of Surgery, Indiana University School of Medicine, Indianapolis, United States of America
| | - Sashwati Roy
- Department of Surgery, Indiana University School of Medicine, Indianapolis, United States of America
| | - Chandan K Sen
- Department of Surgery, Indiana University School of Medicine, Indianapolis, United States of America
| |
Collapse
|
21
|
Rac1 as a Target to Treat Dysfunctions and Cancer of the Bladder. Biomedicines 2022; 10:biomedicines10061357. [PMID: 35740379 PMCID: PMC9219850 DOI: 10.3390/biomedicines10061357] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 06/06/2022] [Accepted: 06/07/2022] [Indexed: 12/28/2022] Open
Abstract
Bladder pathologies, very common in the aged population, have a considerable negative impact on quality of life. Novel targets are needed to design drugs and combinations to treat diseases such as overactive bladder and bladder cancers. A promising new target is the ubiquitous Rho GTPase Rac1, frequently dysregulated and overexpressed in bladder pathologies. We have analyzed the roles of Rac1 in different bladder pathologies, including bacterial infections, diabetes-induced bladder dysfunctions and bladder cancers. The contribution of the Rac1 protein to tumorigenesis, tumor progression, epithelial-mesenchymal transition of bladder cancer cells and their metastasis has been analyzed. Small molecules selectively targeting Rac1 have been discovered or designed, and two of them—NSC23766 and EHT 1864—have revealed activities against bladder cancer. Their mode of interaction with Rac1, at the GTP binding site or the guanine nucleotide exchange factors (GEF) interaction site, is discussed. Our analysis underlines the possibility of targeting Rac1 with small molecules with the objective to combat bladder dysfunctions and to reduce lower urinary tract symptoms. Finally, the interest of a Rac1 inhibitor to treat advanced chemoresistance prostate cancer, while reducing the risk of associated bladder dysfunction, is discussed. There is hope for a better management of bladder pathologies via Rac1-targeted approaches.
Collapse
|
22
|
Chang LL, Lu PH, Yang W, Hu Y, Zheng L, Zhao Q, Lin NM, Zhang WZ. AKR1C1 promotes non-small cell lung cancer proliferation via crosstalk between HIF-1α and metabolic reprogramming. Transl Oncol 2022; 20:101421. [PMID: 35429904 PMCID: PMC9034391 DOI: 10.1016/j.tranon.2022.101421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 03/08/2022] [Accepted: 04/05/2022] [Indexed: 11/28/2022] Open
Abstract
AKR1C1 accelerates the proliferation of NSCLC cells. AKR1C1 remodels metabolism in NSCLC cells. HIF-1α may play a vital role in AKR1C1-mediated metabolic reprogramming.
Non-small cell lung cancer (NSCLC) ranks first among cancer death worldwide. Despite efficacy and safety priority, targeted therapy only benefits ∼30% patients, leading to the unchanged survival rates for whole NSCLC patients. Metabolic reprogramming occurs to offer energy and intermediates for fuelling cancer cells proliferation. Thus, mechanistic insights into metabolic reprogramming may shed light upon NSCLC proliferation and find new proper targets for NSCLC treatment. Herein, we used loss- and gain-of-function experiments to uncover that highly expressed aldo-keto reductase family1 member C1 (AKR1C1) accelerated NSCLC cells proliferation via metabolic reprogramming. Further molecular profiling analyses demonstrated that AKR1C1 augmented the expression of hypoxia-inducible factor 1-alpha (HIF-1α), which could drive tumour metabolic reprogramming. What's more, AKR1C1 significantly correlated with HIF-1α signaling, which predicted poor prognosis for NSCLC patients. Collectively, our data display that AKR1C1 reprograms tumour metabolism to promote NSCLC cells proliferation by activating HIF-1α. These newly acquired data not only establish the specific role for AKR1C1 in metabolic reprogramming, but also hint to the possibility that AKR1C1 may be a new therapeutic target for NSCLC treatment.
Collapse
Affiliation(s)
- Lin-Lin Chang
- Department of Pharmacy, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou 450008, China
| | - Pei-Hua Lu
- Department of Medical Oncology, Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
| | - Wei Yang
- Department of Pharmacy, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou 450008, China
| | - Yan Hu
- Department of Pharmacy, Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China
| | - Lin Zheng
- Zhejiang University, Hangzhou, China
| | - Qiong Zhao
- Shulan International Medical College, Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University, Hangzhou, China
| | - Neng-Ming Lin
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, No.261 Huansha Road, Hangzhou, Zhejiang 310006, China; Cancer Center, Zhejiang University, Hangzhou, China.
| | - Wen-Zhou Zhang
- Department of Pharmacy, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou 450008, China.
| |
Collapse
|
23
|
Passeri G, Northcote-Smith J, Perera R, Gubic N, Suntharalingam K. An Osteosarcoma Stem Cell Potent Nickel(II)-Polypyridyl Complex Containing Flufenamic Acid. Molecules 2022; 27:3277. [PMID: 35630754 PMCID: PMC9143476 DOI: 10.3390/molecules27103277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 05/14/2022] [Accepted: 05/16/2022] [Indexed: 11/25/2022] Open
Abstract
Apoptosis resistance is inherent to stem cell-like populations within tumours and is one of the major reasons for chemotherapy failures in the clinic. Necroptosis is a non-apoptotic mode of programmed cell death that could help bypass apoptosis resistance. Here we report the synthesis, characterisation, biophysical properties, and anti-osteosarcoma stem cell (OSC) properties of a new nickel(II) complex bearing 3,4,7,8-tetramethyl-1,10-phenanthroline and two flufenamic acid moieties, 1. The nickel(II) complex 1 is stable in both DMSO and cell media. The nickel(II) complex 1 kills bulk osteosarcoma cells and OSCs grown in monolayer cultures and osteospheres grown in three-dimensional cultures within the micromolar range. Remarkably, 1 exhibits higher potency towards osteospheres than the metal-based drugs used in current osteosarcoma treatment regimens, cisplatin and carboplatin, and an established anti-cancer stem cell agent, salinomycin (up to 7.7-fold). Cytotoxicity studies in the presence of prostaglandin E2 suggest that 1 kills OSCs in a cyclooxygenase-2 (COX-2) dependent manner. Furthermore, the potency of 1 towards OSCs decreased significantly upon co-treatment with necrostatin-1 or dabrafenib, well-known necroptosis inhibitors, implying that 1 induces necroptosis in OSCs. To the best of our knowledge, 1 is the first compound to implicate both COX-2 and necroptosis in its mechanism of action in OSCs.
Collapse
|
24
|
Kores K, Kolenc Z, Furlan V, Bren U. Inverse Molecular Docking Elucidating the Anticarcinogenic Potential of the Hop Natural Product Xanthohumol and Its Metabolites. Foods 2022; 11:foods11091253. [PMID: 35563976 PMCID: PMC9104229 DOI: 10.3390/foods11091253] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 04/19/2022] [Accepted: 04/25/2022] [Indexed: 01/27/2023] Open
Abstract
Natural products from plants exert a promising potential to act as antioxidants, antimicrobials, anti-inflammatory, and anticarcinogenic agents. Xanthohumol, a natural compound from hops, is indeed known for its anticarcinogenic properties. Xanthohumol is converted into three metabolites: isoxanthohumol (non-enzymatically) as well as 8- and 6-prenylnaringenin (enzymatically). An inverse molecular docking approach was applied to xanthohumol and its three metabolites to discern their potential protein targets. The aim of our study was to disclose the potential protein targets of xanthohumol and its metabolites in order to expound on the potential anticarcinogenic mechanisms of xanthohumol based on the found target proteins. The investigated compounds were docked into the predicted binding sites of all human protein structures from the Protein Data Bank, and the best docking poses were examined. Top scoring human protein targets with successfully docked compounds were identified, and their experimental connection with the anticarcinogenic function or cancer was investigated. The obtained results were carefully checked against the existing experimental findings from the scientific literature as well as further validated using retrospective metrics. More than half of the human protein targets of xanthohumol with the highest docking scores have already been connected with the anticarcinogenic function, and four of them (including two important representatives of the matrix metalloproteinase family, MMP-2 and MMP-9) also have a known experimental correlation with xanthohumol. Another important protein target is acyl-protein thioesterase 2, to which xanthohumol, isoxanthohumol, and 6-prenylnaringenin were successfully docked with the lowest docking scores. Moreover, the results for the metabolites show that their most promising protein targets are connected with the anticarcinogenic function as well. We firmly believe that our study can help to elucidate the anticarcinogenic mechanisms of xanthohumol and its metabolites as after consumption, all four compounds can be simultaneously present in the organism.
Collapse
Affiliation(s)
- Katarina Kores
- Laboratory of Physical Chemistry and Chemical Thermodynamics, Faculty for Chemistry and Chemical Technology, University of Maribor, Smetanova 17, SI-2000 Maribor, Slovenia; (K.K.); (Z.K.); (V.F.)
| | - Zala Kolenc
- Laboratory of Physical Chemistry and Chemical Thermodynamics, Faculty for Chemistry and Chemical Technology, University of Maribor, Smetanova 17, SI-2000 Maribor, Slovenia; (K.K.); (Z.K.); (V.F.)
| | - Veronika Furlan
- Laboratory of Physical Chemistry and Chemical Thermodynamics, Faculty for Chemistry and Chemical Technology, University of Maribor, Smetanova 17, SI-2000 Maribor, Slovenia; (K.K.); (Z.K.); (V.F.)
| | - Urban Bren
- Laboratory of Physical Chemistry and Chemical Thermodynamics, Faculty for Chemistry and Chemical Technology, University of Maribor, Smetanova 17, SI-2000 Maribor, Slovenia; (K.K.); (Z.K.); (V.F.)
- Department of Applied Natural Sciences, Faculty of Mathematics, Natural Sciences and Information Technologies, University of Primorska, Glagoljaška 8, SI-6000 Koper, Slovenia
- Correspondence: ; Tel.: +386-2-229-4421
| |
Collapse
|
25
|
Mezzasoma L, Bellezza I, Romani R, Talesa VN. Extracellular Vesicles and the Inflammasome: An Intricate Network Sustaining Chemoresistance. Front Oncol 2022; 12:888135. [PMID: 35530309 PMCID: PMC9072732 DOI: 10.3389/fonc.2022.888135] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 03/23/2022] [Indexed: 12/24/2022] Open
Abstract
Extracellular vesicles (EVs) are membrane enclosed spherical particles devoted to intercellular communication. Cancer-derived EVs (Ca-EVs) are deeply involved in tumor microenvironment remodeling, modifying the inflammatory phenotype of cancerous and non-cancerous residing cells. Inflammation plays a pivotal role in initiation, development, and progression of many types of malignancies. The key feature of cancer-related inflammation is the production of cytokines that incessantly modify of the surrounding environment. Interleukin-1β (IL-1β) is one of the most powerful cytokines, influencing all the initiation-to-progression stages of many types of cancers and represents an emerging critical contributor to chemoresistance. IL-1β production strictly depends on the activation of inflammasome, a cytoplasmic molecular platform sensing exogenous and endogenous danger signals. It has been recently shown that Ca-EVs can activate the inflammasome cascade and IL-1β production in tumor microenvironment-residing cells. Since inflammasome dysregulation has been established as crucial regulator in inflammation-associated tumorigenesis and chemoresistance, it is conceivable that the use of inflammasome-inhibiting drugs may be employed as adjuvant chemotherapy to counteract chemoresistance. This review focuses on the role of cancer-derived EVs in tuning tumor microenvironment unveiling the intricate network between inflammasome and chemoresistance.
Collapse
|
26
|
Fu Z, Li S, Liu J, Zhang C, Jian C, Wang L, Zhang Y, Shi C. Natural Product Alantolactone Targeting AKR1C1 Suppresses Cell Proliferation and Metastasis in Non-Small-Cell Lung Cancer. Front Pharmacol 2022; 13:847906. [PMID: 35370661 PMCID: PMC8965451 DOI: 10.3389/fphar.2022.847906] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 02/14/2022] [Indexed: 12/29/2022] Open
Abstract
Non-small-cell lung cancer (NSCLC) is one of the leading causes of cancer-related deaths, characterized by high invasion and metastasis. Aldo-keto reductase family 1 member C1 (AKR1C1) plays an important role in cancer cell proliferation and metastasis, and has gained attention as an anticancer drug target. Here, we report that the natural sesquiterpene lactone alantolactone (ALA) was shown to bind directly to AKR1C1 through the Proteome Integral Solubility Alteration (PISA) analysis, a label-free target identification approach based on thermal proteome profiling. Acting as a specific inhibitor of AKR1C1, ALA selectively inhibits the activity of AKR1C1 and ALA treatment in human non-small-cell lung cancer (NSCLC) cell results in a reduction in cell proliferation and metastasis, inhibition of AKR1C1 expression, and deactivation of STAT3. Moreover, ALA inhibited tumor growth in vivo, and the inhibition of AKR1C1 and STAT3 activation were also found in the murine xenograft model. Collectively, our work not only gives mechanistic insights to explain the bioactivity of ALA in anticancer but also provides opportunities of developing novel sesquiterpene lactone-based AKR1C1 inhibitors for the treatment of NSCLC.
Collapse
Affiliation(s)
- Zhiwen Fu
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan, China
| | - Shijun Li
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan, China
| | - Jinmei Liu
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan, China
| | - Cong Zhang
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan, China
| | - Chen Jian
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan, China
| | - Lulu Wang
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan, China
| | - Yu Zhang
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan, China
| | - Chen Shi
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan, China
| |
Collapse
|
27
|
Conod A, Silvano M, Ruiz I Altaba A. On the origin of metastases: Induction of pro-metastatic states after impending cell death via ER stress, reprogramming, and a cytokine storm. Cell Rep 2022; 38:110490. [PMID: 35263600 DOI: 10.1016/j.celrep.2022.110490] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 12/07/2021] [Accepted: 02/14/2022] [Indexed: 12/12/2022] Open
Abstract
How metastatic cells arise is unclear. Here, we search for the induction of recently characterized pro-metastatic states as a surrogate for the origin of metastasis. Since cell-death-inducing therapies can paradoxically promote metastasis, we ask if such treatments induce pro-metastatic states in human colon cancer cells. We find that post-near-death cells acquire pro-metastatic states (PAMEs) and form distant metastases in vivo. These PAME ("let's go" in Greek) cells exhibit a multifactorial cytokine storm as well as signs of enhanced endoplasmic reticulum (ER) stress and nuclear reprogramming, requiring CXCL8, INSL4, IL32, PERK-CHOP, and NANOG. PAMEs induce neighboring tumor cells to become PAME-induced migratory cells (PIMs): highly migratory cells that re-enact the storm and enhance PAME migration. Metastases are thus proposed to originate from the induction of pro-metastatic states through intrinsic and extrinsic cues in a pro-metastatic tumoral ecosystem, driven by an impending cell-death experience involving ER stress modulation, metastatic reprogramming, and paracrine recruitment via a cytokine storm.
Collapse
Affiliation(s)
- Arwen Conod
- Department of Genetic Medicine and Development, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Marianna Silvano
- Department of Genetic Medicine and Development, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Ariel Ruiz I Altaba
- Department of Genetic Medicine and Development, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
| |
Collapse
|
28
|
Chang LL, Li YK, Zhao CX, Zeng CM, Ge FJ, Du JM, Zhang WZ, Lu PH, He QJ, Zhu H, Yang B. AKR1C1 connects autophagy and oxidative stress by interacting with SQSTM1 in a catalytic-independent manner. Acta Pharmacol Sin 2022; 43:703-711. [PMID: 34017066 PMCID: PMC8888619 DOI: 10.1038/s41401-021-00673-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 03/30/2021] [Indexed: 02/04/2023] Open
Abstract
Targeting autophagy might be a promising anticancer strategy; however, the dual roles of autophagy in cancer development and malignancy remain unclear. NSCLC (non-small cell lung cancer) cells harbour high levels of SQSTM1 (sequestosome 1), the autophagy receptor that is critical for the dual roles of autophagy. Therefore, mechanistic insights into SQSTM1 modulation may point towards better approaches to treat NSCLC. Herein, we used multiple autophagy flux models and autophagy readouts to show that aldo-keto reductase family 1 member C1 (AKR1C1), which is highly expressed in NSCLC, promotes autophagy by directly binding to SQSTM1 in a catalytic-independent manner. This interaction may be strengthened by reactive oxygen species (ROS), important autophagy inducers. Further mechanistic research demonstrated that AKR1C1 interacts with SQSTM1 to augment SQSTM1 oligomerization, contributing to the SQSTM1 affinity for binding cargo. Collectively, our data reveal a catalytic-independent role of AKR1C1 for interacting with SQSTM1 and promoting autophagy. All these findings not only reveal a novel functional role of AKR1C1 in the autophagy process but also indicate that modulation of the AKR1C1-SQSTM1 interaction may be a new strategy for targeting autophagy.
Collapse
Affiliation(s)
- Lin-lin Chang
- grid.13402.340000 0004 1759 700XZhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China ,grid.414008.90000 0004 1799 4638Department of Pharmacy, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou 450000, China
| | - Yue-kang Li
- grid.13402.340000 0004 1759 700XZhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Chen-xi Zhao
- grid.13402.340000 0004 1759 700XZhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Chen-ming Zeng
- grid.13402.340000 0004 1759 700XZhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Fu-jing Ge
- grid.13402.340000 0004 1759 700XZhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jia-min Du
- grid.13402.340000 0004 1759 700XZhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Wen-zhou Zhang
- grid.414008.90000 0004 1799 4638Department of Pharmacy, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou 450000, China
| | - Pei-hua Lu
- grid.460176.20000 0004 1775 8598Department of Medical Oncology, Wuxi People’s Hospital of Nanjing Medical University, Wuxi 214023, China
| | - Qiao-jun He
- grid.13402.340000 0004 1759 700XZhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Hong Zhu
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.
| | - Bo Yang
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
29
|
Interactions of Analgesics with Cisplatin: Modulation of Anticancer Efficacy and Potential Organ Toxicity. MEDICINA (KAUNAS, LITHUANIA) 2021; 58:medicina58010046. [PMID: 35056355 PMCID: PMC8781901 DOI: 10.3390/medicina58010046] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 12/19/2021] [Accepted: 12/20/2021] [Indexed: 12/17/2022]
Abstract
Cisplatin (CDDP), one of the most eminent cancer chemotherapeutic agents, has been successfully used to treat more than half of all known cancers worldwide. Despite its effectiveness, CDDP might cause severe toxic adverse effects on multiple body organs during cancer chemotherapy, including the kidneys, heart, liver, gastrointestinal tract, and auditory system, as well as peripheral nerves causing severely painful neuropathy. The latter, among other pains patients feel during chemotherapy, is an indication for the use of analgesics during treatment with CDDP. Different types of analgesics, such as acetaminophen, non-steroidal anti-inflammatory drugs (NSAIDS), and narcotic analgesics, could be used according to the severity of pain. Administered analgesics might modulate CDDP’s efficacy as an anticancer drug. NSAIDS, on one hand, might have cytotoxic effects on their own and few of them can potentiate CDDP’s anticancer effects via inhibiting the CDDP-induced cyclooxygenase (COX) enzyme, or through COX-independent mechanisms. On the other hand, some narcotic analgesics might ameliorate CDDP’s anti-neoplastic effects, causing chemotherapy to fail. Concerning safety, some analgesics share the same adverse effects on normal tissues as CDDP, augmenting its potentially hazardous effects on organ impairment. This article offers an overview of the reported literature on the interactions between analgesics and CDDP, paying special attention to possible mechanisms that modulate CDDP’s cytotoxic efficacy and potential adverse reactions.
Collapse
|
30
|
Phoo NLL, Dejkriengkraikul P, Khaw-On P, Yodkeeree S. Transcriptomic Profiling Reveals AKR1C1 and AKR1C3 Mediate Cisplatin Resistance in Signet Ring Cell Gastric Carcinoma via Autophagic Cell Death. Int J Mol Sci 2021; 22:ijms222212512. [PMID: 34830394 PMCID: PMC8623627 DOI: 10.3390/ijms222212512] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 11/12/2021] [Accepted: 11/16/2021] [Indexed: 12/19/2022] Open
Abstract
Signet ring cell gastric carcinoma (SRCGC) is a lethal malignancy that has developed drug resistance to cisplatin therapies. The aim of this study was to characterize the acquisition of the cisplatin-resistance SRCGC cell line (KATO/DDP cells) and to understand the molecular mechanisms underlying cisplatin resistance. Transcriptomic and bioinformatic analyses were used to identify the candidate gene. This was confirmed by qPCR and Western blot. Aldoketoreductase1C1 and 1C3 (AKR1C1 and AKR1C3) were the most promising molecules in KATO/DDP cells. A specific inhibitor of AKR1C1 (5PBSA) and AKR1C3 (ASP9521) was used to enhance cisplatin-induced KATO/DPP cell death. Although cisplatin alone induced KATO/DDP apoptosis, a combination treatment of cisplatin and the AKR1C inhibitors had no influence on percent cell apoptosis. In conjunction with the autophagy inhibitor, 3MA, attenuated the effects of 5PBSA or ASP9521 to enhance cisplatin-induced cell death. These results indicated that AKR1C1 and 1C3 regulated cisplatin-induced KATO/DDP cell death via autophagy. Moreover, cisplatin in combination with AKR1C inhibitors and N-acetyl cysteine increased KATO/DDP cells' viability when compared with a combination treatment of cisplatin and the inhibitors. Taken together, our results suggested that AKR1C1 and 1C3 play a crucial role in cisplatin resistance of SRCGC by regulating redox-dependent autophagy.
Collapse
Affiliation(s)
- Nang Lae Lae Phoo
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (N.L.L.P.); (P.D.); (P.K.-O.)
| | - Pornngarm Dejkriengkraikul
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (N.L.L.P.); (P.D.); (P.K.-O.)
- Center for Research and Development of Natural Products for Health, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Patompong Khaw-On
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (N.L.L.P.); (P.D.); (P.K.-O.)
| | - Supachai Yodkeeree
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (N.L.L.P.); (P.D.); (P.K.-O.)
- Center for Research and Development of Natural Products for Health, Chiang Mai University, Chiang Mai 50200, Thailand
- Correspondence:
| |
Collapse
|
31
|
Overview of human 20 alpha-hydroxysteroid dehydrogenase (AKR1C1): Functions, regulation, and structural insights of inhibitors. Chem Biol Interact 2021; 351:109746. [PMID: 34780792 DOI: 10.1016/j.cbi.2021.109746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/28/2021] [Accepted: 11/10/2021] [Indexed: 11/22/2022]
Abstract
Human aldo-keto reductase family 1C1 (AKR1C1) is an important enzyme involved in human hormone metabolism, which is mainly responsible for the metabolism of progesterone in the human body. AKR1C1 is highly expressed and has an important relationship with the occurrence and development of various diseases, especially some cancers related to hormone metabolism. Nowadays, many inhibitors against AKR1C1 have been discovered, including some synthetic compounds and natural products, which have certain inhibitory activity against AKR1C1 at the target level. Here we briefly reviewed the physiological and pathological functions of AKR1C1 and the relationship with the disease, and then summarized the development of AKR1C1 inhibitors, elucidated the interaction between inhibitors and AKR1C1 through molecular docking results and existing co-crystal structures. Finally, we discussed the design ideals of selective AKR1C1 inhibitors from the perspective of AKR1C1 structure, discussed the prospects of AKR1C1 in the treatment of human diseases in terms of biomarkers, pre-receptor regulation and single nucleotide polymorphisms, aiming to provide new ideas for drug research targeting AKR1C1.
Collapse
|
32
|
Huang X, Pan T, Yan L, Jin T, Zhang R, Chen B, Feng J, Duan T, Xiang Y, Zhang M, Chen X, Yang Z, Zhang W, Ding X, Xie T, Sui X. The inflammatory microenvironment and the urinary microbiome in the initiation and progression of bladder cancer. Genes Dis 2021; 8:781-797. [PMID: 34522708 PMCID: PMC8427242 DOI: 10.1016/j.gendis.2020.10.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 10/05/2020] [Accepted: 10/06/2020] [Indexed: 12/24/2022] Open
Abstract
Accumulating evidence suggests that chronic inflammation may play a critical role in various malignancies, including bladder cancer. This hypothesis stems in part from inflammatory cells observed in the urethral microenvironment. Chronic inflammation may drive neoplastic transformation and the progression of bladder cancer by activating a series of inflammatory molecules and signals. Recently, it has been shown that the microbiome also plays an important role in the development and progression of bladder cancer, which can be mediated through the stimulation of chronic inflammation. In effect, the urinary microbiome can play a role in establishing the inflammatory urethral microenvironment that may facilitate the development and progression of bladder cancer. In other words, chronic inflammation caused by the urinary microbiome may promote the initiation and progression of bladder cancer. Here, we provide a detailed and comprehensive account of the link between chronic inflammation, the microbiome and bladder cancer. Finally, we highlight that targeting the urinary microbiome might enable the development of strategies for bladder cancer prevention and personalized treatment.
Collapse
Affiliation(s)
- Xingxing Huang
- Department of Medical Oncology, the Affiliated Hospital of Hangzhou Normal University, College of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang Province, 310015, PR China
- College of Pharmacy, School of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang Province, 311121, PR China
| | - Ting Pan
- Department of Medical Oncology, the Affiliated Hospital of Hangzhou Normal University, College of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang Province, 310015, PR China
- College of Pharmacy, School of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang Province, 311121, PR China
| | - Lili Yan
- Department of Medical Oncology, the Affiliated Hospital of Hangzhou Normal University, College of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang Province, 310015, PR China
- College of Pharmacy, School of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang Province, 311121, PR China
| | - Ting Jin
- Department of Medical Oncology, the Affiliated Hospital of Hangzhou Normal University, College of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang Province, 310015, PR China
- College of Pharmacy, School of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang Province, 311121, PR China
| | - Ruonan Zhang
- Department of Medical Oncology, the Affiliated Hospital of Hangzhou Normal University, College of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang Province, 310015, PR China
- College of Pharmacy, School of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang Province, 311121, PR China
| | - Bi Chen
- Department of Medical Oncology, the Affiliated Hospital of Hangzhou Normal University, College of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang Province, 310015, PR China
- College of Pharmacy, School of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang Province, 311121, PR China
| | - Jiao Feng
- Department of Medical Oncology, the Affiliated Hospital of Hangzhou Normal University, College of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang Province, 310015, PR China
- College of Pharmacy, School of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang Province, 311121, PR China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang Province, 311121, PR China
| | - Ting Duan
- Department of Medical Oncology, the Affiliated Hospital of Hangzhou Normal University, College of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang Province, 310015, PR China
- College of Pharmacy, School of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang Province, 311121, PR China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang Province, 311121, PR China
| | - Yu Xiang
- Department of Medical Oncology, the Affiliated Hospital of Hangzhou Normal University, College of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang Province, 310015, PR China
- College of Pharmacy, School of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang Province, 311121, PR China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang Province, 311121, PR China
| | - Mingming Zhang
- Department of Medical Oncology, the Affiliated Hospital of Hangzhou Normal University, College of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang Province, 310015, PR China
- College of Pharmacy, School of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang Province, 311121, PR China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang Province, 311121, PR China
| | - Xiaying Chen
- Department of Medical Oncology, the Affiliated Hospital of Hangzhou Normal University, College of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang Province, 310015, PR China
- College of Pharmacy, School of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang Province, 311121, PR China
| | - Zuyi Yang
- Department of Medical Oncology, the Affiliated Hospital of Hangzhou Normal University, College of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang Province, 310015, PR China
- College of Pharmacy, School of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang Province, 311121, PR China
| | - Wenzheng Zhang
- Department of Medical Oncology, the Affiliated Hospital of Hangzhou Normal University, College of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang Province, 310015, PR China
- College of Pharmacy, School of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang Province, 311121, PR China
| | - Xia Ding
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, PR China
| | - Tian Xie
- Department of Medical Oncology, the Affiliated Hospital of Hangzhou Normal University, College of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang Province, 310015, PR China
- College of Pharmacy, School of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang Province, 311121, PR China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang Province, 311121, PR China
| | - Xinbing Sui
- Department of Medical Oncology, the Affiliated Hospital of Hangzhou Normal University, College of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang Province, 310015, PR China
- College of Pharmacy, School of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang Province, 311121, PR China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang Province, 311121, PR China
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, PR China
| |
Collapse
|
33
|
Penning TM, Jonnalagadda S, Trippier PC, Rižner TL. Aldo-Keto Reductases and Cancer Drug Resistance. Pharmacol Rev 2021; 73:1150-1171. [PMID: 34312303 DOI: 10.1124/pharmrev.120.000122] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Human aldo-keto reductases (AKRs) catalyze the NADPH-dependent reduction of carbonyl groups to alcohols for conjugation reactions to proceed. They are implicated in resistance to cancer chemotherapeutic agents either because they are directly involved in their metabolism or help eradicate the cellular stress created by these agents (e.g., reactive oxygen species and lipid peroxides). Furthermore, this cellular stress activates the Nuclear factor-erythroid 2 p45-related factor 2 (NRF2)-Kelch-like ECH-associated protein 1 pathway. As many human AKR genes are upregulated by the NRF2 transcription factor, this leads to a feed-forward mechanism to enhance drug resistance. Resistance to major classes of chemotherapeutic agents (anthracyclines, mitomycin, cis-platin, antitubulin agents, vinca alkaloids, and cyclophosphamide) occurs by this mechanism. Human AKRs also catalyze the synthesis of androgens and estrogens and the elimination of progestogens and are involved in hormonal-dependent malignancies. They are upregulated by antihormonal therapy providing a second mechanism for cancer drug resistance. Inhibitors of the NRF2 system or pan-AKR1C inhibitors offer promise to surmount cancer drug resistance and/or synergize the effects of existing drugs. SIGNIFICANCE STATEMENT: Aldo-keto reductases (AKRs) are overexpressed in a large number of human tumors and mediate resistance to cancer chemotherapeutics and antihormonal therapies. Existing drugs and new agents in development may surmount this resistance by acting as specific AKR isoforms or AKR pan-inhibitors to improve clinical outcome.
Collapse
Affiliation(s)
- Trevor M Penning
- Center of Excellence in Environmental Toxicology, Department of Systems Pharmacology & Translational Therapeutics, Philadelphia, Pennsylvania (T.M.P.); Department of Pharmaceutical Science (S.J., P.C.T.) and Fred and Pamela Buffett Cancer Center (P.C.T.), University of Nebraska Medical Center and UNMC Center for Drug Discovery, Omaha, Nebraska; and Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia (T.L.R.)
| | - Sravan Jonnalagadda
- Center of Excellence in Environmental Toxicology, Department of Systems Pharmacology & Translational Therapeutics, Philadelphia, Pennsylvania (T.M.P.); Department of Pharmaceutical Science (S.J., P.C.T.) and Fred and Pamela Buffett Cancer Center (P.C.T.), University of Nebraska Medical Center and UNMC Center for Drug Discovery, Omaha, Nebraska; and Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia (T.L.R.)
| | - Paul C Trippier
- Center of Excellence in Environmental Toxicology, Department of Systems Pharmacology & Translational Therapeutics, Philadelphia, Pennsylvania (T.M.P.); Department of Pharmaceutical Science (S.J., P.C.T.) and Fred and Pamela Buffett Cancer Center (P.C.T.), University of Nebraska Medical Center and UNMC Center for Drug Discovery, Omaha, Nebraska; and Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia (T.L.R.)
| | - Tea Lanišnik Rižner
- Center of Excellence in Environmental Toxicology, Department of Systems Pharmacology & Translational Therapeutics, Philadelphia, Pennsylvania (T.M.P.); Department of Pharmaceutical Science (S.J., P.C.T.) and Fred and Pamela Buffett Cancer Center (P.C.T.), University of Nebraska Medical Center and UNMC Center for Drug Discovery, Omaha, Nebraska; and Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia (T.L.R.)
| |
Collapse
|
34
|
Sinreih M, Jójárt R, Kele Z, Büdefeld T, Paragi G, Mernyák E, Rižner TL. Synthesis and evaluation of AKR1C inhibitory properties of A-ring halogenated oestrone derivatives. J Enzyme Inhib Med Chem 2021; 36:1500-1508. [PMID: 34227437 PMCID: PMC8266253 DOI: 10.1080/14756366.2021.1937142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Enzymes AKR1C regulate the action of oestrogens, androgens, and progesterone at the pre-receptor level and are also associated with chemo-resistance. The activities of these oestrone halides were investigated on recombinant AKR1C enzymes. The oestrone halides with halogen atoms at both C-2 and C-4 positions (13β-, 13α-methyl-17-keto halogen derivatives) were the most potent inhibitors of AKR1C1. The lowest IC50 values were for the 13α-epimers 2_2I,4Br and 2_2I,4Cl (IC50, 0.7 μM, 0.8 μM, respectively), both of which selectively inhibited the AKR1C1 isoform. The 13α-methyl-17-keto halogen derivatives 2_2Br and 2_4Cl were the most potent inhibitors of AKR1C2 (IC50, 1.5 μM, 1.8 μM, respectively), with high selectivity for the AKR1C2 isoform. Compound 1_2Cl,4Cl showed the best AKR1C3 inhibition, and it also inhibited AKR1C1 (Ki: AKR1C1, 0.69 μM; AKR1C3, 1.43 μM). These data show that halogenated derivatives of oestrone represent a new class of potent and selective AKR1C inhibitors as lead compounds for further optimisations.
Collapse
Affiliation(s)
- Maša Sinreih
- Faculty of Medicine, Institute of Biochemistry and Molecular Genetics, University of Ljubljana, Ljubljana, Slovenia
| | - Rebeka Jójárt
- Department of Organic Chemistry, University of Szeged, Szeged, Hungary
| | - Zoltán Kele
- Department of Medicinal Chemistry, University of Szeged, Szeged, Hungary
| | - Tomaž Büdefeld
- Faculty of Medicine, Institute of Biochemistry and Molecular Genetics, University of Ljubljana, Ljubljana, Slovenia
| | - Gábor Paragi
- MTA-SZTE Biomimetic Systems Research Group, University of Szeged, Szeged, Hungary.,Institute of Physics, University of Pécs, Pécs, Hungary
| | - Erzsébet Mernyák
- Department of Organic Chemistry, University of Szeged, Szeged, Hungary
| | - Tea Lanišnik Rižner
- Faculty of Medicine, Institute of Biochemistry and Molecular Genetics, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
35
|
Kleinstein SE, McCorrison J, Ahmed A, Hasturk H, Van Dyke TE, Freire M. Transcriptomics of type 2 diabetic and healthy human neutrophils. BMC Immunol 2021; 22:37. [PMID: 34134627 PMCID: PMC8207744 DOI: 10.1186/s12865-021-00428-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 06/06/2021] [Indexed: 01/21/2023] Open
Abstract
OBJECTIVES Chronic inflammatory diseases, including diabetes and cardiovascular disease, are heterogeneous and often co-morbid, with increasing global prevalence. Uncontrolled type 2 diabetes (T2D) can result in severe inflammatory complications. As neutrophils are essential to normal and aberrant inflammation, we conducted RNA-seq transcriptomic analyses to investigate the association between neutrophil gene expression and T2D phenotype. As specialized pro-resolving lipid mediators (SPM) act to resolve inflammation, we further surveyed the impact of neutrophil receptor binding SPM resolvin E1 (RvE1) on isolated diabetic and healthy neutrophils. METHODS Cell isolation and RNA-seq analysis of neutrophils from N = 11 T2D and N = 7 healthy individuals with available clinical data was conducted. Additionally, cultured neutrophils (N = 3 T2D, N = 3 healthy) were perturbed with increasing RvE1 doses (0 nM, 1 nM, 10 nM, or 100 nM) prior to RNA-seq. Data was evaluated through a bioinformatics pipeline including pathway analysis and post hoc false discovery rate (FDR)-correction. RESULTS We observed significant differential expression of 50 genes between T2D and healthy neutrophils (p < 0.05), including decreased T2D gene expression in inflammatory- and lipid-related genes SLC9A4, NECTIN2, and PLPP3 (p < 0.003). RvE1 treatment induced dose-dependent differential gene expression (uncorrected p < 0.05) across groups, including 59 healthy and 216 T2D neutrophil genes. Comparing T2D to healthy neutrophils, 1097 genes were differentially expressed across RvE1 doses, including two significant genes, LILRB5 and AKR1C1, involved in inflammation (p < 0.05). CONCLUSIONS The neutrophil transcriptomic database revealed novel chronic inflammatory- and lipid-related genes that were differentially expressed between T2D cells when compared to controls, and cells responded to RvE1 dose-dependently by gene expression changes. Unraveling the mechanisms regulating abnormalities in diabetic neutrophil responses could lead to better diagnostics and therapeutics targeting inflammation and inflammation resolution.
Collapse
Affiliation(s)
- Sarah E Kleinstein
- Genomic Medicine and Infectious Diseases, J. Craig Venter Institute, 4120 Capricorn Lane, La Jolla, CA, 92037, USA
| | - Jamison McCorrison
- Genomic Medicine and Infectious Diseases, J. Craig Venter Institute, 4120 Capricorn Lane, La Jolla, CA, 92037, USA
| | - Alaa Ahmed
- The Forsyth Institute, Cambridge, MA, USA
- Harvard School of Dental Medicine, Cambridge, MA, USA
| | - Hatice Hasturk
- The Forsyth Institute, Cambridge, MA, USA
- Harvard School of Dental Medicine, Cambridge, MA, USA
| | - Thomas E Van Dyke
- The Forsyth Institute, Cambridge, MA, USA
- Harvard School of Dental Medicine, Cambridge, MA, USA
| | - Marcelo Freire
- Genomic Medicine and Infectious Diseases, J. Craig Venter Institute, 4120 Capricorn Lane, La Jolla, CA, 92037, USA.
- Division of Infectious Diseases and Global Public Health Department of Medicine, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
36
|
Schneider L, Liu J, Zhang C, Azoitei A, Meessen S, Zheng X, Cremer C, Gorzelanny C, Kempe-Gonzales S, Brunner C, Wezel F, Bolenz C, Gunes C, John A. The Role of Interleukin-1-Receptor-Antagonist in Bladder Cancer Cell Migration and Invasion. Int J Mol Sci 2021; 22:ijms22115875. [PMID: 34070905 PMCID: PMC8198563 DOI: 10.3390/ijms22115875] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 05/21/2021] [Accepted: 05/27/2021] [Indexed: 11/30/2022] Open
Abstract
Background: The interleukin-1-receptor antagonist IL1RA (encoded by the IL1RN gene) is a potent competitive antagonist to interleukin-1 (IL1) and thereby is mainly involved in the regulation of inflammation. Previous data indicated a role of IL1RA in muscle-invasive urothelial carcinoma of the bladder (UCB) as well as an IL1-dependent decrease in tissue barrier function, potentially contributing to cancer cell invasion. Objective: Based on these observations, here we investigated the potential roles of IL1RA, IL1A, and IL1B in bladder cancer cell invasion in vitro. Methods: Cell culture, real-time impedance sensing, invasion assays (Boyden chamber, pig bladder model), qPCR, Western blot, ELISA, gene overexpression. Results: We observed a loss of IL1RA expression in invasive, high-grade bladder cancer cell lines T24, UMUC-3, and HT1197 while IL1RA expression was readily detectable in the immortalized UROtsa cells, the non-invasive bladder cancer cell line RT4, and in benign patient urothelium. Thus, we modified the invasive human bladder cancer cell line T24 to ectopically express IL1RA, and measured changes in cell migration/invasion using the xCELLigence Real-Time-Cell-Analysis (RTCA) system and the Boyden chamber assay. The real-time observation data showed a significant decrease of cell migration and invasion in T24 cells overexpressing IL1RA (T24-IL1RA), compared to cells harboring an empty vector (T24-EV). Concurrently, tumor cytokines, e.g., IL1B, attenuated the vascular endothelial barrier, which resulted in a reduction of the Cell Index (CI), an impedance-based dimensionless unit. This reduction could be reverted by the simultaneous incubation with IL1RA. Moreover, we used an ex vivo porcine organ culture system to evaluate cell invasion capacity and showed that T24-IL1RA cells showed significantly less invasive capacity compared to parental T24 cells or T24-EV. Conclusions: Taken together, our results indicate an inverse correlation between IL1RA expression and tumor cell invasive capacity and migration, suggesting that IL1RA plays a role in bladder carcinogenesis, while the exact mechanisms by which IL1RA influences tumor cells migration/invasion remain to be clarified in future studies. Furthermore, we confirmed that real-time impedance sensing and the porcine ex vivo organ culture methods are powerful tools to discover differences in cancer cell migration and invasion.
Collapse
Affiliation(s)
- Lisa Schneider
- Department of Urology, Ulm University Hospital, 89081 Ulm, Germany; (L.S.); (J.L.); (C.Z.); (A.A.); (S.M.); (X.Z.); (C.C.); (F.W.); (C.B.); (A.J.)
| | - Junnan Liu
- Department of Urology, Ulm University Hospital, 89081 Ulm, Germany; (L.S.); (J.L.); (C.Z.); (A.A.); (S.M.); (X.Z.); (C.C.); (F.W.); (C.B.); (A.J.)
| | - Cheng Zhang
- Department of Urology, Ulm University Hospital, 89081 Ulm, Germany; (L.S.); (J.L.); (C.Z.); (A.A.); (S.M.); (X.Z.); (C.C.); (F.W.); (C.B.); (A.J.)
| | - Anca Azoitei
- Department of Urology, Ulm University Hospital, 89081 Ulm, Germany; (L.S.); (J.L.); (C.Z.); (A.A.); (S.M.); (X.Z.); (C.C.); (F.W.); (C.B.); (A.J.)
| | - Sabine Meessen
- Department of Urology, Ulm University Hospital, 89081 Ulm, Germany; (L.S.); (J.L.); (C.Z.); (A.A.); (S.M.); (X.Z.); (C.C.); (F.W.); (C.B.); (A.J.)
| | - Xi Zheng
- Department of Urology, Ulm University Hospital, 89081 Ulm, Germany; (L.S.); (J.L.); (C.Z.); (A.A.); (S.M.); (X.Z.); (C.C.); (F.W.); (C.B.); (A.J.)
| | - Catharina Cremer
- Department of Urology, Ulm University Hospital, 89081 Ulm, Germany; (L.S.); (J.L.); (C.Z.); (A.A.); (S.M.); (X.Z.); (C.C.); (F.W.); (C.B.); (A.J.)
| | | | - Sybille Kempe-Gonzales
- Department of Otorhinolaryngology, Head and Neck Surgery, Ulm University Hospital, 89075 Ulm, Germany; (S.K.-G.); (C.B.)
| | - Cornelia Brunner
- Department of Otorhinolaryngology, Head and Neck Surgery, Ulm University Hospital, 89075 Ulm, Germany; (S.K.-G.); (C.B.)
| | - Felix Wezel
- Department of Urology, Ulm University Hospital, 89081 Ulm, Germany; (L.S.); (J.L.); (C.Z.); (A.A.); (S.M.); (X.Z.); (C.C.); (F.W.); (C.B.); (A.J.)
| | - Christian Bolenz
- Department of Urology, Ulm University Hospital, 89081 Ulm, Germany; (L.S.); (J.L.); (C.Z.); (A.A.); (S.M.); (X.Z.); (C.C.); (F.W.); (C.B.); (A.J.)
| | - Cagatay Gunes
- Department of Urology, Ulm University Hospital, 89081 Ulm, Germany; (L.S.); (J.L.); (C.Z.); (A.A.); (S.M.); (X.Z.); (C.C.); (F.W.); (C.B.); (A.J.)
- Correspondence: ; Tel.: +49-731-500-58019
| | - Axel John
- Department of Urology, Ulm University Hospital, 89081 Ulm, Germany; (L.S.); (J.L.); (C.Z.); (A.A.); (S.M.); (X.Z.); (C.C.); (F.W.); (C.B.); (A.J.)
| |
Collapse
|
37
|
Gao Y, Xu D, Li H, Xu J, Pan Y, Liao X, Qian J, Hu Y, Yu G. Avasimibe Dampens Cholangiocarcinoma Progression by Inhibiting FoxM1-AKR1C1 Signaling. Front Oncol 2021; 11:677678. [PMID: 34127944 PMCID: PMC8195695 DOI: 10.3389/fonc.2021.677678] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 04/28/2021] [Indexed: 12/16/2022] Open
Abstract
Avasimibe is a bioavailable acetyl-CoA acetyltransferase (ACAT) inhibitor and shows a good antitumor effect in various human solid tumors, but its therapeutic value in cholangiocarcinoma (CCA) and underlying mechanisms are largely unknown. In the study, we proved that avasimibe retard cell proliferation and tumor growth of CCAs and identified FoxM1/AKR1C1 axis as the potential novel targets of avasimibe. Aldo-keto reductase 1 family member C1 (AKR1C1) is gradually increased along with the disease progression and highly expressed in human CCAs. From survival analysis, AKR1C1 could be a vital predictor of tumor recurrence and prognostic factor. Enforced Forkhead box protein M1 (FoxM1) expression results in the upregulation of AKR1C1, whereas silencing FoxM1 do the opposite. FoxM1 directly binds to promoter of AKR1C1 and triggers its transcription, while FoxM1-binding site mutation decreases AKR1C1 promoter activity. Moreover, over-expressing exogenous FoxM1 reverses the growth retardation of CCA cells induced by avasimibe administration, while silencing AKR1C1 in FoxM1-overexpressing again retard cell growth. Furthermore, FoxM1 expression significantly correlates with the AKR1C1 expression in human CCA specimens. Our study demonstrates a novel positive regulatory between FoxM1 and AKR1C1 contributing cell growth and tumor progression of CCA and avasimibe may be an alternative therapeutic option for CCA by targeting this FoxM1/AKR1C1 signaling pathway.
Collapse
Affiliation(s)
- Yunshu Gao
- Department of Oncology, People's Liberation Army General Hospital, Beijing, China
| | - Dongyun Xu
- Department of Oncology, The 71st Group Army Hospital of People's Liberation Army, The Affiliated Huaihai Hospital of Xuzhou Medical University, Xuzhou, China
| | - Hongwei Li
- Department of Oncology, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jiahua Xu
- Department of Oncology, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yating Pan
- Department of Oncology, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xinyi Liao
- Department of Oncology, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jianxin Qian
- Department of Oncology, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yi Hu
- Department of Oncology, People's Liberation Army General Hospital, Beijing, China
| | - Guanzhen Yu
- Precision Medical Center laboratory, The First Affiliated Hospital of Wenzhou Medical University, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
38
|
De Las Rivas J, Brozovic A, Izraely S, Casas-Pais A, Witz IP, Figueroa A. Cancer drug resistance induced by EMT: novel therapeutic strategies. Arch Toxicol 2021; 95:2279-2297. [PMID: 34003341 PMCID: PMC8241801 DOI: 10.1007/s00204-021-03063-7] [Citation(s) in RCA: 100] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 04/28/2021] [Indexed: 02/06/2023]
Abstract
Over the last decade, important clinical benefits have been achieved in cancer patients by using drug-targeting strategies. Nevertheless, drug resistance is still a major problem in most cancer therapies. Epithelial-mesenchymal plasticity (EMP) and tumour microenvironment have been described as limiting factors for effective treatment in many cancer types. Moreover, epithelial-to-mesenchymal transition (EMT) has also been associated with therapy resistance in many different preclinical models, although limited evidence has been obtained from clinical studies and clinical samples. In this review, we particularly deepen into the mechanisms of which intermediate epithelial/mesenchymal (E/M) states and its interconnection to microenvironment influence therapy resistance. We also describe how the use of bioinformatics and pharmacogenomics will help to figure out the biological impact of the EMT on drug resistance and to develop novel pharmacological approaches in the future.
Collapse
Affiliation(s)
- Javier De Las Rivas
- Bioinformatics and Functional Genomics Group, Cancer Research Center (CiC-IBMCC, CSIC/USAL/IBSAL), Consejo Superior de Investigaciones Científicas (CSIC), University of Salamanca (USAL), Salamanca, Spain
| | - Anamaria Brozovic
- Division of Molecular Biology, Ruđer Bošković Institute, Bijenička 54, 10000, Zagreb, Croatia
| | - Sivan Izraely
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel-Aviv University, Tel Aviv, Israel
| | - Alba Casas-Pais
- Epithelial Plasticity and Metastasis Group, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, Spain.,Universidade da Coruña (UDC), Coruña, Spain
| | - Isaac P Witz
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel-Aviv University, Tel Aviv, Israel
| | - Angélica Figueroa
- Epithelial Plasticity and Metastasis Group, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, Spain. .,Universidade da Coruña (UDC), Coruña, Spain.
| |
Collapse
|
39
|
Gelardi ELM, Colombo G, Picarazzi F, Ferraris DM, Mangione A, Petrarolo G, Aronica E, Rizzi M, Mori M, La Motta C, Garavaglia S. A Selective Competitive Inhibitor of Aldehyde Dehydrogenase 1A3 Hinders Cancer Cell Growth, Invasiveness and Stemness In Vitro. Cancers (Basel) 2021; 13:cancers13020356. [PMID: 33478031 PMCID: PMC7835878 DOI: 10.3390/cancers13020356] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/13/2021] [Accepted: 01/14/2021] [Indexed: 01/05/2023] Open
Abstract
Simple Summary The aldehyde dehydrogenases enzymes (ALDHs) are promising drug targets in cancer therapy. ALDHs are members of an enzymatic superfamily composed by 19 isoforms involved in the oxidation of aldehydes, with a scavenger role. Among them, the isoform ALDH1A3 is a cancer biomarker since it is highly expressed in cancer stem cells characterized by a marked drug resistance and the capacity to promote self-renewal, clonogenic growth and tumour-initiating capacity. In this paper, we present the first highly potent and selective ALDH1A3 inhibitor able to induce cytotoxic effects and to reduce cell migration and stemness of ALDH1A3-positive cancer cells. We propose the targeting of the ALDH1A3 enzyme as a promising approach for improving the treatments outcomes of patients affected by ALDH1A3-positive cancers. Abstract Aldehyde dehydrogenase 1A3 (ALDH1A3) belongs to an enzymatic superfamily composed by 19 different isoforms, with a scavenger role, involved in the oxidation of a plethora of aldehydes to the respective carboxylic acids, through a NAD+-dependent reaction. Previous clinical studies highlighted the high expression of ALDH1A3 in cancer stem cells (CSCs) correlated to a higher risk of cancer relapses, chemoresistance and a poor clinical outcome. We report on the structural, biochemical, and cellular characterization of NR6, a new selective ALDH1A3 inhibitor derived from an already published ALDH non-selective inhibitor with cytotoxic activity on glioblastoma and colorectal cancer cells. Crystal structure, through X-Ray analysis, showed that NR6 binds a non-conserved tyrosine residue of ALDH1A3 which drives the selectivity towards this isoform, as supported by computational binding simulations. Moreover, NR6 shows anti-metastatic activity in wound healing and invasion assays and induces the downregulation of cancer stem cell markers. Overall, our work confirms the role of ALDH1A3 as an important target in glioblastoma and colorectal cells and propose NR6 as a promising molecule for future preclinical studies.
Collapse
Affiliation(s)
- Edoardo L. M. Gelardi
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, A. Avogadro, 28100 Novara, Italy; (E.L.M.G.); (G.C.); (D.M.F.); (A.M.); (M.R.)
| | - Giorgia Colombo
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, A. Avogadro, 28100 Novara, Italy; (E.L.M.G.); (G.C.); (D.M.F.); (A.M.); (M.R.)
| | - Francesca Picarazzi
- Dipartimento di Biotecnologie, Chimica e Farmacia, University of Siena, 53100 Siena, Italy; (F.P.); (M.M.)
| | - Davide M. Ferraris
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, A. Avogadro, 28100 Novara, Italy; (E.L.M.G.); (G.C.); (D.M.F.); (A.M.); (M.R.)
| | - Andrea Mangione
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, A. Avogadro, 28100 Novara, Italy; (E.L.M.G.); (G.C.); (D.M.F.); (A.M.); (M.R.)
| | - Giovanni Petrarolo
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (G.P.); (C.L.M.)
| | - Eleonora Aronica
- Department of (Neuro)Pathology, Amsterdam UMC, University of Amsterdam, 1105 Amsterdam, The Netherlands;
- Stichting Epilepsie Instellingen Nederland (SEIN), 2103 Heemstede, The Netherlands
| | - Menico Rizzi
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, A. Avogadro, 28100 Novara, Italy; (E.L.M.G.); (G.C.); (D.M.F.); (A.M.); (M.R.)
| | - Mattia Mori
- Dipartimento di Biotecnologie, Chimica e Farmacia, University of Siena, 53100 Siena, Italy; (F.P.); (M.M.)
| | - Concettina La Motta
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (G.P.); (C.L.M.)
- CISUP—Centre for Instrumentation Sharing, University of Pisa, 56126 Pisa, Italy
| | - Silvia Garavaglia
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, A. Avogadro, 28100 Novara, Italy; (E.L.M.G.); (G.C.); (D.M.F.); (A.M.); (M.R.)
- Correspondence: ; Tel.: +39-0321375714
| |
Collapse
|
40
|
John A, Günes C, Bolenz C, Vidal-Y-Sy S, Bauer AT, Schneider SW, Gorzelanny C. Bladder cancer-derived interleukin-1 converts the vascular endothelium into a pro-inflammatory and pro-coagulatory surface. BMC Cancer 2020; 20:1178. [PMID: 33267794 PMCID: PMC7709388 DOI: 10.1186/s12885-020-07548-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 10/21/2020] [Indexed: 12/31/2022] Open
Abstract
Background Bladder cancer cells orchestrate tumour progression by pro-inflammatory cytokines. Cytokines modulate the local tumour microenvironment and increase the susceptibility of tumour distant tissues for metastasis. Here, we investigated the impact of human bladder cancer cell derived factors on the ability to modulate and activate human vascular endothelial cells. Methods The pro-inflammatory and pro-coagulatory potential of four different bladder cancer cell lines was accessed by qRT-PCR arrays and ELISA. Modulation and activation of endothelial cells was studied in microfluidic devices. Clinical relevance of our findings was confirmed by immune histology in tissue samples of bladder cancer patients and public transcriptome data. Results The unbalanced ratio between interleukin (IL)-1 and IL-1 receptor antagonist (IL-1ra) in the secretome of bladder cancer cells converted the quiescent vascular endothelium into a pro-adhesive, pro-inflammatory, and pro-coagulatory surface. Microfluidic experiments showed that tumour cell induced endothelial cell activation promoted leukocyte recruitment and platelet adhesion. Human bladder cancer tissue analysis confirmed that loss of IL-1ra and elevated IL-1 expression was associated with enhanced cancer progression. Conclusions Our data indicate that IL-1 and IL-1ra were dysregulated in bladder cancer and could facilitate tumour dissemination through endothelial cell activation. Targeting the IL-1/IL-1ra axis might attenuate tumour-mediated inflammation and metastasis formation. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-020-07548-z.
Collapse
Affiliation(s)
- A John
- Department of Urology, University of Ulm, Ulm, Germany
| | - C Günes
- Department of Urology, University of Ulm, Ulm, Germany
| | - C Bolenz
- Department of Urology, University of Ulm, Ulm, Germany
| | - S Vidal-Y-Sy
- Department of Dermatology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - A T Bauer
- Department of Dermatology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - S W Schneider
- Department of Dermatology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - C Gorzelanny
- Department of Dermatology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany.
| |
Collapse
|
41
|
Li S, Ji J, Zhang Z, Peng Q, Hao L, Guo Y, Zhou W, Cui Q, Shi X. Cisplatin promotes the expression level of PD-L1 in the microenvironment of hepatocellular carcinoma through YAP1. Mol Cell Biochem 2020; 475:79-91. [PMID: 32761300 DOI: 10.1007/s11010-020-03861-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 07/26/2020] [Indexed: 12/26/2022]
Abstract
Hepatocellular carcinoma (HCC) is one of the most lethal malignancies worldwide. However, the immune tolerance limits the effect of chemotherapeutic drugs. Therefore, the mechanism of cisplatin in promoting PD-L1 expression by YAP1 was investigated in the present study, and we found that cisplatin increased the expression level of YAP1 in the mouse liver with H22 cells. Meanwhile, cisplatin improved the expression level of PD-L1, IL-1β and CCL2 in the tumor microenvironment. Further, cisplatin also enhanced the expression level of YAP1 in shYAP1 HepG2215 cells. The expression of PD-L1 was decreased by Verteporfin, YAP1 inhibitor, during the treatment of DEN/TCPOBOP-induced liver cancer in C57BL/6 mice. These results suggested that cisplatin could deteriorate the immunosuppressive microenvironment through increasing PD-L1, CCL2, IL-1β by upregulated YAP1 expression. Therefore, the study suggested that YAP1 blockade destroyed the immunosuppressive microenvironment of cancer to improve the effect of chemotherapy in HCC.
Collapse
Affiliation(s)
- Shenghao Li
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, No.3 Xingyuan Road, Shijiazhuang, 050200, Hebei, China.,Hebei Key Laboratory of Integrative Medicine On Liver Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang, 050200, China
| | - Jingmin Ji
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, No.3 Xingyuan Road, Shijiazhuang, 050200, Hebei, China
| | - Zhiqin Zhang
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, No.3 Xingyuan Road, Shijiazhuang, 050200, Hebei, China
| | - Qing Peng
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, No.3 Xingyuan Road, Shijiazhuang, 050200, Hebei, China
| | - Liyuan Hao
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, No.3 Xingyuan Road, Shijiazhuang, 050200, Hebei, China
| | - Yinglin Guo
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, No.3 Xingyuan Road, Shijiazhuang, 050200, Hebei, China
| | - Wenhan Zhou
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, No.3 Xingyuan Road, Shijiazhuang, 050200, Hebei, China
| | - Qingzhuo Cui
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, No.3 Xingyuan Road, Shijiazhuang, 050200, Hebei, China
| | - Xinli Shi
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, No.3 Xingyuan Road, Shijiazhuang, 050200, Hebei, China. .,Hebei Key Laboratory of Integrative Medicine On Liver Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang, 050200, China.
| |
Collapse
|
42
|
Li XM, Cao LL. Identification of GOLM1 as a positively regulator of tumor metastasis by regulating MMP13 in gastric carcinoma. Cancer Biomark 2020; 26:421-430. [PMID: 31640085 DOI: 10.3233/cbm-190301] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Metastatic gastric carcinoma (GC) is a typically incurable disease. The progression of anti-metastatic treatment is hampered because the underlying mechanisms regulating the metastasis of GC cell are not well illuminated. OBJECTIVE Therefore, further elucidation of the molecular mechanism behind the metastatic traits of GC cells is needed for optimizing GC treatment. METHODS The levels of GOLM1 and MMP13 in GC cells and tissues were measured by using qPCR assay. The growth of GC cells in vitro was detected using MTS and colony formation assays. The migration and invasion of GC cells was analyzed using wound healing test and Transwell invasion assay. The level of MMP13 in GC cell was measured using immunoblotting and the level of GOLM1 was measured using immunofluorescence staining. The role of GOLM1 on the distant metastasis of GC SGC7910 cell was analyzed using experimental metastasis assay. Transplanted tumor model was constructed to analyze the influence of GOLM1 on GC cell growth in vivo. RESULTS Here, we report that GOLM1 is over-expressed in GC and knockdown GOLM1 impairs the aggressive phenotypes of GC cell in vitro. Furthermore, downregulation of GOLM1 restrains the tumor growth of GC cell in nude mice. Nevertheless, upregulation of GOLM1 distinctly elevated the growth, migration ability and invasiveness of GC SGC7910 cell. Finally, GOLM1 increases the metastatic phenotypes of GC cell in a MMP13-dependent manner. CONCLUSIONS Altogether, this investigation demonstrates the crucial function of GOLM1 in the progression of GC, which indicating GOLM1 as a potential target for GC treatment.
Collapse
|
43
|
Zhou C, Shen G, Yang F, Duan J, Wu Z, Yang M, Liu Y, Du X, Zhang X, Xiao S. Loss of AKR1C1 is a good prognostic factor in advanced NPC cases and increases chemosensitivity to cisplatin in NPC cells. J Cell Mol Med 2020; 24:6438-6447. [PMID: 32307891 PMCID: PMC7294127 DOI: 10.1111/jcmm.15291] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 03/24/2020] [Accepted: 03/26/2020] [Indexed: 12/14/2022] Open
Abstract
Cisplatin resistance is one of the main obstacles in the treatment of advanced nasopharyngeal carcinoma (NPC). AKR1C1 is a member of the Aldo-keto reductase superfamily (AKRs), which converts aldehydes and ketones to their corresponding alcohols and has been reported to be involved in chemotherapeutic resistance of multiple drugs. The expression and function of AKR1C1 in NPC have not been reported until now. The aim of this research was to investigate the expression of AKR1C1 and it is role in cisplatin resistance in NPC. AKR1C1 protein expression was detected by immunohistochemistry in human NPC tissues and by Western blot assays in NPC and immortalized nasopharyngeal epithelial cells. The effects of AKR1C1 knock-down by siRNA on proliferation, migration and invasion in NPC cells were evaluated by CCK8, wound healing and transwell assays. To evaluate the effects of AKR1C1 silencing on cisplatin sensitivity in NPC cells, CCK8 assays were used to detect cell proliferation, flow cytometry was used to detect cell cycle distribution, and flow cytometry and DAPI staining were used to detect cell apoptosis. AKR1C1 down-regulation was associated with advanced clinicopathological characters such as larger tumor size, more lymphatic nodes involvement, with metastasis and later clinical stages, while AKR1C1 down-regulation was a good prognostic factor for overall survival (OS) in NPC patients. In vitro study showed that AKR1C1 was not directly involved in the malignant biological behaviours such as proliferation, cell cycle progression and migration of NPC cells, whereas AKR1C1 knock-down could enhance cisplatin sensitivity of NPC cells. These results suggest that AKR1C1 is a potential marker for predicting cisplatin response and could serve as a molecular target to increase cisplatin sensitivity in NPC.
Collapse
Affiliation(s)
- Chen Zhou
- Department of Pathology, The Second Affiliated Hospital, Guilin Medical University, Guilin, China
| | - Guowen Shen
- Department of Pathology, The Second Affiliated Hospital, Guilin Medical University, Guilin, China
| | - Fan Yang
- Department of Pathology, The Second Affiliated Hospital, Guilin Medical University, Guilin, China
| | - Jingling Duan
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Zhen Wu
- Xiangya Medical College of South Central University, Changsha, China
| | - Mingqing Yang
- Department of Pathology, The Second Affiliated Hospital, Guilin Medical University, Guilin, China
| | - Yi Liu
- Department of Pathology, The Second Affiliated Hospital, Guilin Medical University, Guilin, China
| | - Xueli Du
- Department of Pathology, The Second Affiliated Hospital, Guilin Medical University, Guilin, China
| | - Xiaoling Zhang
- Department of Physiology, Faculty of Basic Medical Science, Guilin Medical University, Guilin, China
| | - Shengjun Xiao
- Department of Pathology, The Second Affiliated Hospital, Guilin Medical University, Guilin, China
| |
Collapse
|
44
|
Li ZY, Yin YF, Guo Y, Li H, Xu MQ, Liu M, Wang JR, Feng ZH, Duan XC, Zhang S, Zhang SQ, Wang GX, Liao A, Wang SM, Zhang X. Enhancing Anti-Tumor Activity of Sorafenib Mesoporous Silica Nanomatrix in Metastatic Breast Tumor and Hepatocellular Carcinoma via the Co-Administration with Flufenamic Acid. Int J Nanomedicine 2020; 15:1809-1821. [PMID: 32214813 PMCID: PMC7083629 DOI: 10.2147/ijn.s240436] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 02/21/2020] [Indexed: 12/13/2022] Open
Abstract
INTRODUCTION Because tumor-associated inflammation is a hallmark of cancer treatment, in the present study, sorafenib mesoporous silica nanomatrix (MSNM@SFN) co-administrated with flufenamic acid (FFA, a non-steroidal anti-inflammatory drug (NSAID)) was investigated to enhance the anti-tumor activity of MSNM@SFN. METHODS Metastatic breast tumor 4T1/luc cells and hepatocellular carcinoma HepG2 cells were selected as cell models. The effects of FFA in vitro on cell migration, PGE2 secretion, and AKR1C1 and AKR1C3 levels in 4T1/luc and HepG2 cells were investigated. The in vivo anti-tumor activity of MSNM@SFN co-administrating with FFA (MSNM@SFN+FFA) was evaluated in a 4T1/luc metastatic tumor model, HepG2 tumor-bearing nude mice model, and HepG2 orthotopic tumor-bearing nude mice model, respectively. RESULTS The results indicated that FFA could markedly decrease cell migration, PGE2 secretion, and AKR1C1 and AKR1C3 levels in both 4T1/luc and HepG2 cells. The enhanced anti-tumor activity of MSNM@SFN+FFA compared with that of MSNM@SFN was confirmed in the 4T1/luc metastatic tumor model, HepG2 tumor-bearing nude mice model, and HepG2 orthotopic tumor-bearing nude mice model in vivo, respectively. DISCUSSION MSNM@SFN co-administrating with FFA (MSNM@SFN+FFA) developed in this study is an alternative strategy for improving the therapeutic efficacy of MSNM@SFN via co-administration with NSAIDs.
Collapse
Affiliation(s)
- Zhuo-Yue Li
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing100191, People’s Republic of China
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing100191, People’s Republic of China
| | - Yi-Fan Yin
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing100191, People’s Republic of China
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing100191, People’s Republic of China
| | - Yang Guo
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing100191, People’s Republic of China
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing100191, People’s Republic of China
| | - Hui Li
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing100191, People’s Republic of China
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing100191, People’s Republic of China
| | - Mei-Qi Xu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing100191, People’s Republic of China
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing100191, People’s Republic of China
| | - Man Liu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing100191, People’s Republic of China
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing100191, People’s Republic of China
| | - Jing-Ru Wang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing100191, People’s Republic of China
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing100191, People’s Republic of China
| | - Zhen-Han Feng
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing100191, People’s Republic of China
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing100191, People’s Republic of China
| | - Xiao-Chuan Duan
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing100191, People’s Republic of China
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing100191, People’s Republic of China
| | - Shuang Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing100191, People’s Republic of China
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing100191, People’s Republic of China
| | - Shuai-Qiang Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing100191, People’s Republic of China
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing100191, People’s Republic of China
| | - Guang-Xue Wang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing100191, People’s Republic of China
| | - Ai Liao
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing100191, People’s Republic of China
| | - Shu-Min Wang
- Department of Ultrasound, Peking University Third Hospital, Peking University, Beijing100191, People’s Republic of China
| | - Xuan Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing100191, People’s Republic of China
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing100191, People’s Republic of China
| |
Collapse
|
45
|
Jiang Y, Li Y, Cheng J, Ma J, Li Q, Pang T. Upregulation of AKR1C1 in mesenchymal stromal cells promotes the survival of acute myeloid leukaemia cells. Br J Haematol 2020; 189:694-706. [PMID: 31943135 DOI: 10.1111/bjh.16253] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 10/01/2019] [Indexed: 01/03/2023]
Abstract
The leukaemic bone marrow microenvironment, comprising abnormal mesenchymal stromal cells (MSCs), is responsible for the poor prognosis of acute myeloid leukaemia (AML). Therefore, it is essential to determine the mechanisms underlying the supportive role of MSCs in the survival of leukaemia cells. Through in silico analyses, we identified a total of 271 aberrantly expressed genes in the MSCs derived from acute myeloid leukemia (AML) patients that were associated with adipogenic differentiation, of which aldo-keto reductase 1C1 (AKR1C1) was significantly upregulated in the AML-MSCs. Knockdown of AKR1C1 in the MSCs suppressed adipogenesis and promoted osteogenesis, and inhibited the growth of co-cultured AML cell lines compared to the situation in wild- type AML-derived MSCs. Introduction of recombinant human AKR1C1 in the MSCs partially alleviated the effects of AKR1C1 knockdown. In addition, the absence of AKR1C1 reduced secretion of cytokines such as MCP-1, IL-6 and G-CSF from the MSCs, along with inactivation of STAT3 and ERK1/2 in the co-cultured AML cells. AKR1C1 is an essential factor driving the adipogenic differentiation of leukaemic MSCs and mediates its pro-survival effects on AML cells by promoting cytokine secretion and activating the downstream pathways in the AML cells.
Collapse
Affiliation(s)
- Yajing Jiang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Disease, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Ying Li
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Disease, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Jingying Cheng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Disease, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Jiao Ma
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Disease, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Qinghua Li
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Disease, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Tianxiang Pang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Disease, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| |
Collapse
|
46
|
Zhu H, Hu Y, Zeng C, Chang L, Ge F, Wang W, Yan F, Zhao Q, Cao J, Ying M, Gu Y, Zheng L, He Q, Yang B. The SIRT2-mediated deacetylation of AKR1C1 is required for suppressing its pro-metastasis function in Non-Small Cell Lung Cancer. Am J Cancer Res 2020; 10:2188-2200. [PMID: 32104503 PMCID: PMC7019158 DOI: 10.7150/thno.39151] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 12/09/2019] [Indexed: 12/16/2022] Open
Abstract
Aldo-keto reductase family 1 member C1 (AKR1C1) promotes malignancy of Non-Small Cell Lung Cancer (NSCLC) by activating Signal Transducer and Activator of Transcription 3 (STAT3) pathway. However, how the pro-metastatic functions of AKR1C1 are switched on/off remains unknown. Methods: Immunoprecipitation and LC-MS/MS analyses were performed to identify the acetylation on AKR1C1 protein, and the functional analyses (in vitro and in vivo) were performed to depict the contribution of acetylation to the pro-metastatic effects of AKR1C1. Results: Here we report that acetylated AKR1C1 on two lysine residues K185 & K201 is critical to its pro-metastatic role. The acetylation modification has no impact on the canonical enzymatic activity of AKR1C1, while it is required for the interaction between AKR1C1 to STAT3, which triggers the downstream transduction events, ultimately mobilizing cells. Importantly, the deacetylase Sirtuin 2 (SIRT2) is capable of deacetylating AKR1C1, inhibiting the transactivation of STAT3 target genes, thus suppressing the migration of cells. Conclusion: Acetylation on Lysines 185 and 201 of AKR1C1 dictates its pro-metastatic potential both in vitro and in vivo, and the reverting of acetylation by Sirtuin 2 provides potential therapeutic targets for treatment against metastatic NSCLC patients with high AKR1C1 expression.
Collapse
|
47
|
Shadpour P, Zamani M, Aghaalikhani N, Rashtchizadeh N. Inflammatory cytokines in bladder cancer. J Cell Physiol 2019; 234:14489-14499. [PMID: 30779110 DOI: 10.1002/jcp.28252] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Revised: 12/29/2018] [Accepted: 01/22/2019] [Indexed: 01/24/2023]
Abstract
The presence of inflammatory cells and their products in the tumor microenvironment plays a crucial role in the pathogenesis of a tumor. Releasing the cytokines from a host in response to infection and inflammation can inhibit tumor growth and progression. However, tumor cells can also respond to the host cytokines with increasing the growth/invasion/metastasis. Bladder cancer (BC) is one of the most common cancers in the world. The microenvironment of a bladder tumor has been indicated to be rich in growth factors/inflammatory cytokines that can induce the tumor growth/progression and also suppress the immune system. On the contrary, modulate of the cancer progression has been shown following upregulation of the cytokines-related pathways that suggested the cytokines as potential therapeutic targets. In this study, we provide a summary of cytokines that are involved in BC formation/regression with both inflammatory and anti-inflammatory properties. A more accurate understanding of tumor microenvironment creates favorable conditions for cytokines targeting to treat BC.
Collapse
Affiliation(s)
- Pejman Shadpour
- Hasheminejad Kidney Center (HKC), Hospital Management Research Center (HMRC), Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Mojtaba Zamani
- Department of Agronomy and Plant Breeding, School of Agriculture, University of Tehran, Karaj, Iran
| | - Nazi Aghaalikhani
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nadereh Rashtchizadeh
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
48
|
Chang WM, Chang YC, Yang YC, Lin SK, Chang PMH, Hsiao M. AKR1C1 controls cisplatin-resistance in head and neck squamous cell carcinoma through cross-talk with the STAT1/3 signaling pathway. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:245. [PMID: 31182137 PMCID: PMC6558898 DOI: 10.1186/s13046-019-1256-2] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 05/30/2019] [Indexed: 01/22/2023]
Abstract
Background Cisplatin is the first-line chemotherapy used against most upper aerodigestive tract carcinomas. In head and neck cancer, sensitivity to cisplatin remains the key issue in treatment response and outcome. Genetic heterogeneity and aberrant gene expression may be the intrinsic factors that cause primary cisplatin-resistance. Methods Combination of the HNSCC gene expression data and the cisplatin sensitivity results from public database. We found that aldo-keto reductase family 1 member C1 (AKR1C1) may be associated with cisplatin sensitivity in HNSCC treatment of naïve cells. We examined the AKR1C1 expression and its correlation with cisplatin IC50 and prognosis in patients. The in vitro and in vivo AKR1C1 functions in cisplatin-resistance through overexpression or knockdown assays, respectively. cDNA microarrays were used to identify the upstream regulators that modulate AKR1C1-induced signaling in HNSCC. Finally, we used the cigarette metabolites to promote AKR1C1 expression and ruxolitinib to overcome AKR1C1-induced cisplatin-resistance. Results AKR1C1 positively correlates to cisplatin-resistance in HNSCC cells. AKR1C1 is a poor prognostic factor for recurrence and death of HNSCC patients. Silencing of AKR1C1 not only reduced in vitro IC50 but also increased in vivo cisplatin responses and vise versa in overexpression cells. Cigarette metabolites also promote AKR1C1 expression. Transcriptome analyses revealed that STAT1 and STAT3 activation enable AKR1C1-induced cisplatin-resistance and can be overcome by ruxolitinib treatment. Conclusions AKR1C1 is a crucial regulator for cisplatin-resistance in HNSCC and also poor prognostic marker for patients. Targeting the AKR1C1-STAT axis may provide a new therapeutic strategy to treat patients who are refractory to cisplatin treatment. Electronic supplementary material The online version of this article (10.1186/s13046-019-1256-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Wei-Min Chang
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Yu-Chan Chang
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Yi-Chieh Yang
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Sze-Kwan Lin
- Department of Dentistry, National Taiwan University Hospital, Taipei, Taiwan
| | - Peter Mu-Hsin Chang
- Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan. .,Faculty of Medicine, College of Medicine, National Yang-Ming University, Taipei, Taiwan.
| | - Michael Hsiao
- Genomics Research Center, Academia Sinica, Taipei, Taiwan. .,Department of Biochemistry, Kaohsiung Medical University, Kaohsiung, Taiwan.
| |
Collapse
|
49
|
Resveratrol as a Tumor-Suppressive Nutraceutical Modulating Tumor Microenvironment and Malignant Behaviors of Cancer. Int J Mol Sci 2019; 20:ijms20040925. [PMID: 30791624 PMCID: PMC6412705 DOI: 10.3390/ijms20040925] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 02/17/2019] [Accepted: 02/18/2019] [Indexed: 12/15/2022] Open
Abstract
Tumor-suppressive effects of resveratrol have been shown in various types of cancer. However, regulation of tumor microenvironment by resveratrol is still unclear. Recent findings suggest resveratrol can potentiate its tumor-suppressive effect through modulation of the signaling pathways of cellular components (fibroblasts, macrophages and T cells). Also, studies have shown that resveratrol can suppress malignant phenotypes of cancer cells acquired in response to stresses of the tumor microenvironment, such as hypoxia, oxidative stress and inflammation. We discuss the effects of resveratrol on cancer cells in stress environment of tumors as well as interactions between cancer cells and non-cancer cells in this review.
Collapse
|
50
|
Wang L, Liu Y, Yu G. Avasimibe inhibits tumor growth by targeting FoxM1-AKR1C1 in osteosarcoma. Onco Targets Ther 2019; 12:815-823. [PMID: 30774369 PMCID: PMC6353227 DOI: 10.2147/ott.s165647] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Background Osteosarcoma (OS) is a rare bone tumor with a high propensity for lung metastasis and poor patient outcomes. It is crucial to identify novel therapeutic strategies and biomarkers. Patients and methods ARK1C1 staining was detected in OS specimens, and its clinical significance was assessed. A potential AKR1C1 inhibitor, avasimibe, was used to target AKR1C1. Results High expression of AKR1C1 was observed in OS and was associated with poor outcomes for patients with OS. Avasimibe was found to inhibit cell proliferation and tumor growth by reducing the expression of AKR1C1 and FoxM1 in vivo and in vitro. Conclusion These findings indicate that AKR1C1 is a promising prognostic factor and may serve as a novel therapeutic target of avasimibe for human OS.
Collapse
Affiliation(s)
- Liang Wang
- Department of Orthopedic, Changzheng Hospital, Shanghai 200003, China
| | - Yang Liu
- Department of Orthopedic, Changzheng Hospital, Shanghai 200003, China
| | - Guanzhen Yu
- Department of Oncology, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China,
| |
Collapse
|