1
|
Cao H, Liu B, Gong K, Wu H, Wang Y, Zhang H, Shi C, Wang P, Du H, Zhou H, Wang S. Association between cathepsins and benign prostate diseases: a bidirectional two-sample Mendelian randomization study. Front Endocrinol (Lausanne) 2024; 15:1348310. [PMID: 38904040 PMCID: PMC11188316 DOI: 10.3389/fendo.2024.1348310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 05/20/2024] [Indexed: 06/22/2024] Open
Abstract
Objectives The relationship between cathepsins and prostate cancer (PCa) has been reported. However, there is a lack of research on cathepsins and benign prostate diseases (BPDs). This study investigated the potential genetic link between cathepsins and BPDs through the utilization of Mendelian randomization (MR) analysis to determine if a causal relationship exists. Methods Publicly accessible summary statistics on BPDs were obtained from FinnGen Biobank. The data comprised 149,363 individuals, with 30,066 cases and 119,297 controls for BPH, and 123,057 individuals, with 3,760 cases and 119,297 controls for prostatitis. The IEU OpenGWAS provided the Genome-wide association data on ten cathepsins. To evaluate the causal relationship between BPDs and cathepsins, five distinct MR analyses were employed, with the primary method being the inverse variance weighted (IVW) approach. Additionally, sensitivity analyses were conducted to examine the horizontal pleiotropy and heterogeneity of the findings. Results The examination of IVW MR findings showed that cathepsin O had a beneficial effect on BPH (IVW OR=0.94, 95% CI 0.89-0.98, P=0.0055), while cathepsin X posed a threat to prostatitis (IVW OR=1.08, 95% CI 1.00-1.16, P=0.047). Through reverse MR analysis, it was revealed that prostatitis had an adverse impact on cathepsin V (IVW OR=0.89, 95% CI 0.80-0.99, P=0.035), while no favorable association was observed between BPH and cathepsins. The results obtained from MR-Egger, weighted median, simple mode, and weighted mode methods were consistent with the findings of the IVW approach. Based on sensitivity analyses, heterogeneity, and horizontal pleiotropy are unlikely to distort the results. Conclusion This study offers the initial evidence of a genetic causal link between cathepsins and BPDs. Our findings revealed that cathepsin O was beneficial in preventing BPH, whereas cathepsin X posed a potential threat to prostatitis. Additionally, prostatitis negatively affected cathepsin V level. These three cathepsins could be targets of diagnosis and treatment for BPDs, which need further research.
Collapse
Affiliation(s)
- Hongliang Cao
- Department of Urology II, The First Hospital of Jilin University, Changchun, China
| | - Bin Liu
- Department of Urology II, The First Hospital of Jilin University, Changchun, China
| | - Kejian Gong
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, China
| | - Hao Wu
- Department of Urology II, The First Hospital of Jilin University, Changchun, China
| | - Yishu Wang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, China
| | - Haiyang Zhang
- Department of Prosthodontics, Hospital of Stomatology, Jilin University, Changchun, China
| | - Chengdong Shi
- Department of Urology II, The First Hospital of Jilin University, Changchun, China
| | - Pengyu Wang
- Department of Urology II, The First Hospital of Jilin University, Changchun, China
| | - Hao Du
- Department of Urology II, The First Hospital of Jilin University, Changchun, China
| | - Honglan Zhou
- Department of Urology II, The First Hospital of Jilin University, Changchun, China
| | - Song Wang
- Department of Urology II, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
2
|
Kumar DN, Chaudhuri A, Dehari D, Gamper AM, Kumar D, Agrawal AK. Enhanced Therapeutic Efficacy Against Melanoma through Exosomal Delivery of Hesperidin. Mol Pharm 2024; 21:3061-3076. [PMID: 38757678 DOI: 10.1021/acs.molpharmaceut.4c00490] [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] [Indexed: 05/18/2024]
Abstract
Melanoma, characterized as the most aggressive and metastatic form of skin cancer, currently has limited treatment options, predominantly chemotherapy and radiation therapy. However, the drawbacks associated with parenterally administered chemotherapy underscore the urgent need for alternative compounds to combat melanoma effectively. Hesperidin (HES), a flavonoid present in various citrus fruits, exhibits promising anticancer activity. Nevertheless, the clinical utility of HES is hindered by challenges such as poor water solubility, a short half-life, and low oral bioavailability. In response to these limitations, we introduced a novel approach by formulating HES-loaded exosomes (Exo-HES). Isolation of exosomes was achieved through the ultracentrifugation method, and HES was efficiently loaded using the sonication method. The resulting formulations displayed a desirable particle size (∼106 nm) and exhibited a spherical morphology, as confirmed by scanning electron and atomic force microscopy. In vitro studies conducted on B16F10 cell lines demonstrated higher cytotoxicity of Exo-HES compared to free HES, supported by enhanced cellular uptake validated through coumarin-6-loaded exosomes. This superior cytotoxicity was further evidenced by DNA fragmentation, increased generation of free radicals (ROS), loss of mitochondrial membrane potential, and effective inhibition of colony formation. The antimetastatic properties of Exo-HES were confirmed through wound healing and transwell migration assays. Oral pharmacokinetics studies revealed a remarkable increase of approximately 2.5 times in oral bioavailability and half-life of HES when loaded into exosomes. Subsequent in vivo experiments utilizing a B16F10-induced melanoma model in Swiss mice established that Exo-HES exhibited superior anticancer activity compared to HES after oral administration. Importantly, no biochemical, hematological, or histological toxicities were observed in tumor-bearing mice treated with Exo-HES. These findings suggest that exosomes loaded with HES represent a promising nanocarrier strategy to enhance the therapeutic effectiveness of hesperidin in melanoma treatment.
Collapse
Affiliation(s)
- Dulla Naveen Kumar
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU) Varanasi, Varanasi 221005, India
| | - Aiswarya Chaudhuri
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU) Varanasi, Varanasi 221005, India
| | - Deepa Dehari
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU) Varanasi, Varanasi 221005, India
| | - Armin M Gamper
- Department of Oncology, University of Alberta, Edmonton, Alberta T6G 1Z2, Canada
- Cross Cancer Institute, Edmonton, Alberta T6G 1Z2, Canada
| | - Dinesh Kumar
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU) Varanasi, Varanasi 221005, India
| | - Ashish Kumar Agrawal
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU) Varanasi, Varanasi 221005, India
| |
Collapse
|
3
|
Liu F, Zhou T, Zhang S, Li Y, Chen Y, Miao Z, Wang X, Yang G, Li Q, Zhang L, Liu Y. Cathepsin B: The dawn of tumor therapy. Eur J Med Chem 2024; 269:116329. [PMID: 38508117 DOI: 10.1016/j.ejmech.2024.116329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 03/10/2024] [Accepted: 03/11/2024] [Indexed: 03/22/2024]
Abstract
Cathepsin B (CTSB) is a key lysosomal protease that plays a crucial role in the development of cancer. This article elucidates the relationship between CTSB and cancer from the perspectives of its structure, function, and role in tumor growth, migration, invasion, metastasis, angiogenesis and autophagy. Further, we summarized the research progress of cancer treatment related drugs targeting CTSB, as well as the potential and advantages of Traditional Chinese medicine in treating tumors by regulating the expression of CTSB.
Collapse
Affiliation(s)
- Fuxian Liu
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities, Gansu University of Chinese Medicine, Lanzhou, China
| | - Ting Zhou
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities, Gansu University of Chinese Medicine, Lanzhou, China; Experimental & Training Teaching Centers, Gansu University of Chinese Medicine, Lanzhou, China
| | - Shangzu Zhang
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities, Gansu University of Chinese Medicine, Lanzhou, China
| | - Yangyang Li
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities, Gansu University of Chinese Medicine, Lanzhou, China
| | - Yan Chen
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities, Gansu University of Chinese Medicine, Lanzhou, China
| | - Zhiming Miao
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities, Gansu University of Chinese Medicine, Lanzhou, China
| | - Xin Wang
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities, Gansu University of Chinese Medicine, Lanzhou, China
| | - Gengqiang Yang
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities, Gansu University of Chinese Medicine, Lanzhou, China
| | - Qiyang Li
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities, Gansu University of Chinese Medicine, Lanzhou, China
| | - Liying Zhang
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities, Gansu University of Chinese Medicine, Lanzhou, China; College of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, China.
| | - Yongqi Liu
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities, Gansu University of Chinese Medicine, Lanzhou, China; College of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, China; Key Laboratory of Dunhuang Medicine and Transformation at Provincial and Ministerial Level, Gansu University of Chinese Medicine, Lanzhou, China.
| |
Collapse
|
4
|
Samaržija I. The Potential of Extracellular Matrix- and Integrin Adhesion Complex-Related Molecules for Prostate Cancer Biomarker Discovery. Biomedicines 2023; 12:79. [PMID: 38255186 PMCID: PMC10813710 DOI: 10.3390/biomedicines12010079] [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: 11/18/2023] [Revised: 12/16/2023] [Accepted: 12/26/2023] [Indexed: 01/24/2024] Open
Abstract
Prostate cancer is among the top five cancer types according to incidence and mortality. One of the main obstacles in prostate cancer management is the inability to foresee its course, which ranges from slow growth throughout years that requires minimum or no intervention to highly aggressive disease that spreads quickly and resists treatment. Therefore, it is not surprising that numerous studies have attempted to find biomarkers of prostate cancer occurrence, risk stratification, therapy response, and patient outcome. However, only a few prostate cancer biomarkers are used in clinics, which shows how difficult it is to find a novel biomarker. Cell adhesion to the extracellular matrix (ECM) through integrins is among the essential processes that govern its fate. Upon activation and ligation, integrins form multi-protein intracellular structures called integrin adhesion complexes (IACs). In this review article, the focus is put on the biomarker potential of the ECM- and IAC-related molecules stemming from both body fluids and prostate cancer tissue. The processes that they are involved in, such as tumor stiffening, bone turnover, and communication via exosomes, and their biomarker potential are also reviewed.
Collapse
Affiliation(s)
- Ivana Samaržija
- Laboratory for Epigenomics, Division of Molecular Medicine, Ruđer Bošković Institute, 10000 Zagreb, Croatia
| |
Collapse
|
5
|
Furuta H, Sheng Y, Takahashi A, Nagano R, Kataoka N, Perks CM, Barker R, Hakuno F, Takahashi SI. The IGF-Independent Role of IRS-2 in the Secretion of MMP-9 Enhances the Growth of Prostate Carcinoma Cell Line PC3. Int J Mol Sci 2023; 24:15065. [PMID: 37894751 PMCID: PMC10606031 DOI: 10.3390/ijms242015065] [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: 09/07/2023] [Revised: 10/06/2023] [Accepted: 10/06/2023] [Indexed: 10/29/2023] Open
Abstract
Insulin receptor substrate-2 (IRS-2), a substrate of the insulin-like growth factor (IGF)-I receptor, is highly expressed in the prostate cancer cell line, PC3. We recently demonstrated that extracellular signal-regulated kinase (Erk1/2), a kinase downstream of IGF signaling, is activated in PC3 cells under serum starvation, and this activation can be inhibited by IRS-2 knockdown. Here, we observed that adding an IGF-I-neutralizing antibody to the culture medium inhibited the activation of Erk1/2. Suppression of Erk1/2 in IRS-2 knockdown cells was restored by the addition of a PC3 serum-free conditioned medium. In contrast, the IRS-2-silenced PC3 conditioned medium could not restore Erk1/2 activation, suggesting that IRS-2 promotes the secretion of proteins that activate the IGF signaling pathway. Furthermore, gelatin zymography analysis of the conditioned medium showed that matrix metalloproteinase-9 (MMP-9) was secreted extracellularly in an IRS-2 dependent manner when PC3 was cultured under serum starvation conditions. Moreover, MMP-9 knockdown suppressed Erk1/2 activation, DNA synthesis, and migratory activity. The IRS-2 levels were positively correlated with Gleason grade in human prostate cancer tissues. These data suggest that highly expressed IRS-2 activates IGF signaling by enabling the secretion of MMP-9, which is associated with hyperproliferation and malignancy of prostate cancer cell line, PC3.
Collapse
Affiliation(s)
- Haruka Furuta
- Department of Animal Sciences and Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo 113-8654, Japan; (H.F.); (Y.S.); (A.T.); (R.N.); (N.K.)
| | - Yina Sheng
- Department of Animal Sciences and Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo 113-8654, Japan; (H.F.); (Y.S.); (A.T.); (R.N.); (N.K.)
| | - Ayaka Takahashi
- Department of Animal Sciences and Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo 113-8654, Japan; (H.F.); (Y.S.); (A.T.); (R.N.); (N.K.)
| | - Raku Nagano
- Department of Animal Sciences and Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo 113-8654, Japan; (H.F.); (Y.S.); (A.T.); (R.N.); (N.K.)
| | - Naoyuki Kataoka
- Department of Animal Sciences and Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo 113-8654, Japan; (H.F.); (Y.S.); (A.T.); (R.N.); (N.K.)
| | - Claire Marie Perks
- IGFs & Metabolic Endocrinology Group, Learning & Research Building, Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol BS8 1TH, UK; (C.M.P.); (R.B.)
| | - Rachel Barker
- IGFs & Metabolic Endocrinology Group, Learning & Research Building, Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol BS8 1TH, UK; (C.M.P.); (R.B.)
| | - Fumihiko Hakuno
- Department of Animal Sciences and Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo 113-8654, Japan; (H.F.); (Y.S.); (A.T.); (R.N.); (N.K.)
| | - Shin-Ichiro Takahashi
- Department of Animal Sciences and Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo 113-8654, Japan; (H.F.); (Y.S.); (A.T.); (R.N.); (N.K.)
| |
Collapse
|
6
|
Egorova VS, Kolesova EP, Lopus M, Yan N, Parodi A, Zamyatnin AA. Smart Delivery Systems Responsive to Cathepsin B Activity for Cancer Treatment. Pharmaceutics 2023; 15:1848. [PMID: 37514035 PMCID: PMC10386206 DOI: 10.3390/pharmaceutics15071848] [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: 05/29/2023] [Revised: 06/23/2023] [Accepted: 06/27/2023] [Indexed: 07/30/2023] Open
Abstract
Cathepsin B is a lysosomal cysteine protease, contributing to vital cellular homeostatic processes including protein turnover, macroautophagy of damaged organelles, antigen presentation, and in the extracellular space, it takes part in tissue remodeling, prohormone processing, and activation. However, aberrant overexpression of cathepsin B and its enzymatic activity is associated with different pathological conditions, including cancer. Cathepsin B overexpression in tumor tissues makes this enzyme an important target for smart delivery systems, responsive to the activity of this enzyme. The generation of technologies which therapeutic effect is activated as a result of cathepsin B cleavage provides an opportunity for tumor-targeted therapy and controlled drug release. In this review, we summarized different technologies designed to improve current cancer treatments responsive to the activity of this enzyme that were shown to play a key role in disease progression and response to the treatment.
Collapse
Affiliation(s)
- Vera S Egorova
- Scientific Center for Translation Medicine, Sirius University of Science and Technology, Sochi 354340, Russia
| | - Ekaterina P Kolesova
- Scientific Center for Translation Medicine, Sirius University of Science and Technology, Sochi 354340, Russia
| | - Manu Lopus
- School of Biological Sciences, UM-DAE Centre for Excellence in Basic Sciences, University of Mumbai Kalina Campus, Vidyanagari, Mumbai 400098, India
| | - Neng Yan
- School of Environmental Studies, China University of Geosciences, Wuhan 430074, China
| | - Alessandro Parodi
- Scientific Center for Translation Medicine, Sirius University of Science and Technology, Sochi 354340, Russia
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow 119991, Russia
| | - Andrey A Zamyatnin
- Scientific Center for Translation Medicine, Sirius University of Science and Technology, Sochi 354340, Russia
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow 119991, Russia
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119991, Russia
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow 119991, Russia
| |
Collapse
|
7
|
Wang Y, Xi W, Zhang X, Bi X, Liu B, Zheng X, Chi X. CTSB promotes sepsis-induced acute kidney injury through activating mitochondrial apoptosis pathway. Front Immunol 2023; 13:1053754. [PMID: 36713420 PMCID: PMC9880165 DOI: 10.3389/fimmu.2022.1053754] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 12/28/2022] [Indexed: 01/15/2023] Open
Abstract
Background Acute kidney injury is a common and severe complication of sepsis. Sepsis -induced acute kidney injury(S-AKI) is an independent risk factor for mortality among sepsis patients. However, the mechanisms of S-AKI are complex and poorly understand. Therefore, exploring the underlying mechanisms of S-AKI may lead to the development of therapeutic targets. Method A model of S-AKI was established in male C57BL/6 mice using cecal ligation and puncture (CLP). The data-independent acquisition (DIA)-mass spectrometry-based proteomics was used to explore the protein expression changes and analyze the key proteomics profile in control and CLP group. The methodology was also used to identify the key proteins and pathways. S-AKI in vitro was established by treating the HK-2 cells with lipopolysaccharide (LPS). Subsequently, the effect and mechanism of Cathepsin B (CTSB) in inducing apoptosis in HK-2 cells were observed and verified. Results The renal injury scores, serum creatinine, blood urea nitrogen, and kidney injury molecule 1 were higher in septic mice than in non-septic mice. The proteomic analysis identified a total of 449 differentially expressed proteins (DEPs). GO and KEGG analysis showed that DEPs were mostly enriched in lysosomal-related cell structures and pathways. CTSB and MAPK were identified as key proteins in S-AKI. Electron microscopy observed enlarged lysosomes, swelled and ruptured mitochondria, and cytoplasmic vacuolization in CLP group. TUNEL staining and CTSB activity test showed that the apoptosis and CTSB activity were higher in CLP group than in control group. In HK-2 cell injury model, the CTSB activity and mRNA expression were increased in LPS-treated cells. Acridine orange staining showed that LPS caused lysosomal membrane permeabilization (LMP). CA074 as an inhibitor of CTSB could effectively inhibit CTSB activity. CCK8 and Annexin V/PI staining results indicated that CA074 reversed LPS-induced apoptosis of HK-2 cells. The JC-1 and western blot results showed that LPS inhibited mitochondrial membrane potential and activated mitochondrial apoptosis pathway, which could be reversed by CA074. Conclusions LMP and CTSB contribute to pathogenesis of S-AKI. LPS treatment induced HK-2 cell injury by activating mitochondrial apoptosis pathway. Inhibition of CTSB might be a new therapeutic strategy to alleviate sepsis-induced acute kidney injury.
Collapse
Affiliation(s)
- Yuting Wang
- Department of Anesthesiology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Wenjie Xi
- Department of Anesthesiology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Xinyi Zhang
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Xinwen Bi
- Department of Anesthesiology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Boyang Liu
- Department of Anesthesiology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Xiaoming Zheng
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China,*Correspondence: Xiaoming Zheng, ; Xinjin Chi,
| | - Xinjin Chi
- Department of Anesthesiology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China,*Correspondence: Xiaoming Zheng, ; Xinjin Chi,
| |
Collapse
|
8
|
Johnson CS, Cook LM. Osteoid cell-derived chemokines drive bone-metastatic prostate cancer. Front Oncol 2023; 13:1100585. [PMID: 37025604 PMCID: PMC10070788 DOI: 10.3389/fonc.2023.1100585] [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: 11/16/2022] [Accepted: 03/07/2023] [Indexed: 04/08/2023] Open
Abstract
One of the greatest challenges in improving prostate cancer (PCa) survival is in designing new therapies to effectively target bone metastases. PCa regulation of the bone environment has been well characterized; however, bone-targeted therapies have little impact on patient survival, demonstrating a need for understanding the complexities of the tumor-bone environment. Many factors contribute to creating a favorable microenvironment for prostate tumors in bone, including cell signaling proteins produced by osteoid cells. Specifically, there has been extensive evidence from both past and recent studies that emphasize the importance of chemokine signaling in promoting PCa progression in the bone environment. Chemokine-focused strategies present promising therapeutic options for treating bone metastasis. These signaling pathways are complex, with many being produced by (and exerting effects on) a plethora of different cell types, including stromal and tumor cells of the prostate tumor-bone microenvironment. This review highlights an underappreciated molecular family that should be interrogated for treatment of bone metastatic prostate cancer (BM-PCa).
Collapse
Affiliation(s)
- Catherine S. Johnson
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, United States
- Eppley Institute for Research in Cancer and Allied Diseases, Omaha, NE, United States
| | - Leah M. Cook
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, United States
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, United States
- *Correspondence: Leah M. Cook,
| |
Collapse
|
9
|
El‐Hussieny M, Mansour ST, Hashem AI, Fouad MA, Abd‐El‐Maksoud MA. Design, synthesis, and biological evaluation of new heterocycles bearing both silicon and phosphorus as potent
MMP
‐2 inhibitors. J CHIN CHEM SOC-TAIP 2022. [DOI: 10.1002/jccs.202200327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Affiliation(s)
- Marwa El‐Hussieny
- Organometallic and Organometalloid Chemistry Department National Research Centre Giza Egypt
| | - Shaimaa T. Mansour
- Organometallic and Organometalloid Chemistry Department National Research Centre Giza Egypt
| | - Ahmed I. Hashem
- Chemistry Department, Faculty of Science Ain Shams University Cairo Egypt
| | - Marwa A. Fouad
- Pharmaceutical Chemistry Department, Faculty of Pharmacy Cairo University Giza Egypt
| | | |
Collapse
|
10
|
Contursi A, Tacconelli S, Hofling U, Bruno A, Dovizio M, Ballerini P, Patrignani P. Biology and pharmacology of platelet-type 12-lipoxygenase in platelets, cancer cells, and their crosstalk. Biochem Pharmacol 2022; 205:115252. [PMID: 36130648 DOI: 10.1016/j.bcp.2022.115252] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 09/08/2022] [Accepted: 09/12/2022] [Indexed: 11/26/2022]
Abstract
Platelet-type lipoxygenase (pl12-LOX), encoded by ALOX12, catalyzes the production of the lipid mediator 12S-hydroperoxyeicosa-5,8,10,14-tetraenoic acid (12S-HpETE), which is quickly reduced by cellular peroxidases to form 12(S)-hydroxy-5,8,10,14-eicosatetraenoic acid (12S-HETE). Platelets express high levels of pl12-LOX and generate considerable amounts of 12S-HETE from arachidonic acid (AA; C20:4, n-6). The development of sensitive chiral liquid chromatography-tandem mass spectrometry (LC-MS/MS) methods has allowed the accurate quantification of 12S-HETE in biological samples. Moreover, advances in the knowledge of the mechanism of action of 12S-HETE have been achieved. The orphan G-protein-coupled receptor 31 (GPR31) has been identified as the high-affinity 12S-HETE receptor. Moreover, upon platelet activation, 12S-HETE is produced, and significant amounts are found esterified to membrane phospholipids (PLs), such as phosphatidylethanolamine (PE) and phosphatidylcholine (PC), promoting thrombin generation. Platelets play many roles in cancer metastasis. Among them, the platelets' ability to interact with cancer cells and transfer platelet molecules by the release of extracellular vesicles (EVs) is noteworthy. Recently, it was found that platelets induce epithelial-mesenchymal transition(EMT) in cancer cells, a phenomenon known to confer high-grade malignancy, through the transfer of pl12-LOX contained in platelet-derived EVs. These cancer cells now generate 12-HETE, considered a key modulator of cancer metastasis. Interestingly, 12-HETE was mainly found esterified in plasmalogen phospholipids of cancer cells. This review summarizes the current knowledge on the regulation and functions of pl12-LOX in platelets and cancer cells and their crosstalk.Novel approaches to preventing cancer and metastasis by the pharmacological inhibition of pl12-LOX and the internalization of mEVs are discussed.
Collapse
Affiliation(s)
- Annalisa Contursi
- Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University, Chieti, Italy; Department of Neuroscience, Imaging and Clinical Science, "G. d'Annunzio" University, Chieti, Italy
| | - Stefania Tacconelli
- Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University, Chieti, Italy; Department of Neuroscience, Imaging and Clinical Science, "G. d'Annunzio" University, Chieti, Italy
| | - Ulrika Hofling
- Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University, Chieti, Italy; Department of Neuroscience, Imaging and Clinical Science, "G. d'Annunzio" University, Chieti, Italy
| | - Annalisa Bruno
- Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University, Chieti, Italy; Department of Neuroscience, Imaging and Clinical Science, "G. d'Annunzio" University, Chieti, Italy
| | - Melania Dovizio
- Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University, Chieti, Italy; Department of Neuroscience, Imaging and Clinical Science, "G. d'Annunzio" University, Chieti, Italy
| | - Patrizia Ballerini
- Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University, Chieti, Italy; Department of Innovative Technologies in Medicine and Dentistry, "G. d'Annunzio" University, Chieti, Italy
| | - Paola Patrignani
- Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University, Chieti, Italy; Department of Neuroscience, Imaging and Clinical Science, "G. d'Annunzio" University, Chieti, Italy.
| |
Collapse
|
11
|
Augoff K, Hryniewicz-Jankowska A, Tabola R, Stach K. MMP9: A Tough Target for Targeted Therapy for Cancer. Cancers (Basel) 2022; 14:cancers14071847. [PMID: 35406619 PMCID: PMC8998077 DOI: 10.3390/cancers14071847] [Citation(s) in RCA: 85] [Impact Index Per Article: 42.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 03/27/2022] [Accepted: 03/31/2022] [Indexed: 02/01/2023] Open
Abstract
Having the capability to proteolyze diverse structural and signaling proteins, matrix metalloproteinase 9 (MMP9), one of the best-studied secretory endopeptidases, has been identified as a crucial mediator of processes closely associated with tumorigenesis, such as the extracellular matrix reorganization, epithelial to mesenchymal transition, cell migration, new blood vessel formation, and immune response. In this review, we present the current state of knowledge on MMP9 and its role in cancer growth in the context of cell adhesion/migration, cancer-related inflammation, and tumor microenvironment formation. We also summarize recent achievements in the development of selective MMP9 inhibitors and the limitations of using them as anticancer drugs.
Collapse
Affiliation(s)
- Katarzyna Augoff
- Department of Surgical Education, Wroclaw Medical University, 50-367 Wroclaw, Poland
- Department of Chemistry and Immunochemistry, Wroclaw Medical University, 50-367 Wroclaw, Poland;
- Correspondence:
| | | | - Renata Tabola
- Department of Thoracic Surgery, Wroclaw Medical University, 50-367 Wroclaw, Poland;
| | - Kamilla Stach
- Department of Chemistry and Immunochemistry, Wroclaw Medical University, 50-367 Wroclaw, Poland;
| |
Collapse
|
12
|
Shahzad MH, Feng L, Su X, Brassard A, Dhoparee-Doomah I, Ferri LE, Spicer JD, Cools-Lartigue JJ. Neutrophil Extracellular Traps in Cancer Therapy Resistance. Cancers (Basel) 2022; 14:1359. [PMID: 35267667 PMCID: PMC8909607 DOI: 10.3390/cancers14051359] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 03/01/2022] [Accepted: 03/05/2022] [Indexed: 02/04/2023] Open
Abstract
Neutrophils and their products are increasingly recognized to have a key influence on cancer progression and response to therapy. Their involvement has been shown in nearly every aspect of cancer pathophysiology with growing evidence now supporting their role in resistance to a variety of cancer therapies. Recently, the role of neutrophils in cancer progression and therapy resistance has been further complicated with the discovery of neutrophil extracellular traps (NETs). NETs are web-like structures of chromatin decorated with a variety of microbicidal proteins. They are released by neutrophils in a process called NETosis. NET-dependent mechanisms of cancer pathology are beginning to be appreciated, particularly with respect to tumor response to chemo-, immuno-, and radiation therapy. Several studies support the functional role of NETs in cancer therapy resistance, involving T-cell exhaustion, drug detoxification, angiogenesis, the epithelial-to-mesenchymal transition, and extracellular matrix remodeling mechanisms, among others. Given this, new and promising data suggests NETs provide a microenvironment conducive to limited therapeutic response across a variety of neoplasms. As such, this paper aims to give a comprehensive overview of evidence on NETs in cancer therapy resistance with a focus on clinical applicability.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Jonathan J. Cools-Lartigue
- Department of Surgery, Division of Thoracic and Upper Gastrointestinal Surgery, Montreal General Hospital, Montreal, QC H3G 1A4, Canada; (M.H.S.); (L.F.); (X.S.); (A.B.); (I.D.-D.); (L.E.F.); (J.D.S.)
| |
Collapse
|
13
|
Shmakova AA, Klimovich PS, Rysenkova KD, Popov VS, Gorbunova AS, Karpukhina AA, Karagyaur MN, Rubina KA, Tkachuk VA, Semina EV. Urokinase Receptor uPAR Downregulation in Neuroblastoma Leads to Dormancy, Chemoresistance and Metastasis. Cancers (Basel) 2022; 14:cancers14040994. [PMID: 35205745 PMCID: PMC8870350 DOI: 10.3390/cancers14040994] [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: 01/02/2022] [Revised: 02/05/2022] [Accepted: 02/12/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary uPAR is a membrane receptor that contributes to extracellular matrix remodeling and controls cellular adhesion, proliferation, survival, and migration. We demonstrate that the initially high uPAR expression predicts poor survival in neuroblastoma. However, relapsed neuroblastomas have a significantly decreased uPAR expression. uPAR downregulation in neuroblastoma cells leads to dormancy and resistance to chemotherapeutic drugs. In mice, low uPAR-expressing neuroblastoma cells formed smaller primary tumors but more frequent metastasis. Abstract uPAR is a membrane receptor that binds extracellular protease urokinase, contributes to matrix remodeling and plays a crucial role in cellular adhesion, proliferation, survival, and migration. uPAR overexpression in tumor cells promotes mitogenesis, opening a prospective avenue for targeted therapy. However, uPAR targeting in cancer has potential risks. We have recently shown that uPAR downregulation in neuroblastoma promotes epithelial-mesenchymal transition (EMT), potentially associated with metastasis and chemoresistance. We used data mining to evaluate the role of uPAR expression in primary and relapsed human neuroblastomas. To model the decreased uPAR expression, we targeted uPAR using CRISPR/Cas9 and shRNA in neuroblastoma Neuro2a cells and evaluated their chemosensitivity in vitro as well as tumor growth and metastasis in vivo. We demonstrate that the initially high PLAUR expression predicts poor survival in human neuroblastoma. However, relapsed neuroblastomas have a significantly decreased PLAUR expression. uPAR targeting in neuroblastoma Neuro2a cells leads to p38 activation and an increased p21 expression (suggesting a dormant phenotype). The dormancy in neuroblastoma cells can be triggered by the disruption of uPAR-integrin interaction. uPAR-deficient cells are less sensitive to cisplatin and doxorubicin treatment and exhibit lower p53 activation. Finally, low uPAR-expressing Neuro2a cells formed smaller primary tumors, but more frequent metastasis in mice. To the best of our knowledge, this is the first study revealing the pathological role of dormant uPAR-deficient cancer cells having a chemoresistant and motile phenotype.
Collapse
Affiliation(s)
- Anna A. Shmakova
- National Cardiology Research Center of the Ministry of Health of the Russian Federation, Institute of Experimental Cardiology, 121552 Moscow, Russia; (A.A.S.); (P.S.K.); (K.D.R.); (V.A.T.)
- Faculty of Medicine, Lomonosov Moscow State University, 119192 Moscow, Russia; (V.S.P.); (A.S.G.); (M.N.K.); (K.A.R.)
| | - Polina S. Klimovich
- National Cardiology Research Center of the Ministry of Health of the Russian Federation, Institute of Experimental Cardiology, 121552 Moscow, Russia; (A.A.S.); (P.S.K.); (K.D.R.); (V.A.T.)
- Faculty of Medicine, Lomonosov Moscow State University, 119192 Moscow, Russia; (V.S.P.); (A.S.G.); (M.N.K.); (K.A.R.)
| | - Karina D. Rysenkova
- National Cardiology Research Center of the Ministry of Health of the Russian Federation, Institute of Experimental Cardiology, 121552 Moscow, Russia; (A.A.S.); (P.S.K.); (K.D.R.); (V.A.T.)
- Faculty of Medicine, Lomonosov Moscow State University, 119192 Moscow, Russia; (V.S.P.); (A.S.G.); (M.N.K.); (K.A.R.)
| | - Vladimir S. Popov
- Faculty of Medicine, Lomonosov Moscow State University, 119192 Moscow, Russia; (V.S.P.); (A.S.G.); (M.N.K.); (K.A.R.)
| | - Anna S. Gorbunova
- Faculty of Medicine, Lomonosov Moscow State University, 119192 Moscow, Russia; (V.S.P.); (A.S.G.); (M.N.K.); (K.A.R.)
| | - Anna A. Karpukhina
- Koltzov Institute of Developmental Biology, Russian Academy of Science, 117334 Moscow, Russia;
| | - Maxim N. Karagyaur
- Faculty of Medicine, Lomonosov Moscow State University, 119192 Moscow, Russia; (V.S.P.); (A.S.G.); (M.N.K.); (K.A.R.)
| | - Kseniya A. Rubina
- Faculty of Medicine, Lomonosov Moscow State University, 119192 Moscow, Russia; (V.S.P.); (A.S.G.); (M.N.K.); (K.A.R.)
| | - Vsevolod A. Tkachuk
- National Cardiology Research Center of the Ministry of Health of the Russian Federation, Institute of Experimental Cardiology, 121552 Moscow, Russia; (A.A.S.); (P.S.K.); (K.D.R.); (V.A.T.)
- Faculty of Medicine, Lomonosov Moscow State University, 119192 Moscow, Russia; (V.S.P.); (A.S.G.); (M.N.K.); (K.A.R.)
| | - Ekaterina V. Semina
- National Cardiology Research Center of the Ministry of Health of the Russian Federation, Institute of Experimental Cardiology, 121552 Moscow, Russia; (A.A.S.); (P.S.K.); (K.D.R.); (V.A.T.)
- Faculty of Medicine, Lomonosov Moscow State University, 119192 Moscow, Russia; (V.S.P.); (A.S.G.); (M.N.K.); (K.A.R.)
- Correspondence:
| |
Collapse
|
14
|
Mason LD, Chava S, Reddi KK, Gupta R. The BRD9/7 Inhibitor TP-472 Blocks Melanoma Tumor Growth by Suppressing ECM-Mediated Oncogenic Signaling and Inducing Apoptosis. Cancers (Basel) 2021; 13:cancers13215516. [PMID: 34771678 PMCID: PMC8582741 DOI: 10.3390/cancers13215516] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 10/26/2021] [Accepted: 11/01/2021] [Indexed: 12/12/2022] Open
Abstract
Melanoma accounts for the majority of all skin cancer-related deaths and only 1/3rd of melanoma patients with distal metastasis survive beyond five years. However, current therapies including BRAF/MEK targeted therapies or immunotherapies only benefit a subset of melanoma patients due to the emergence of intrinsic or extrinsic resistance mechanisms. Effective treatment of melanoma will thus require new and more effective therapeutic agents. Towards the goal of identifying new therapeutic agents, we conducted an unbiased, druggable epigenetic drug screen using a library of 32 epigenetic inhibitors obtained from the Structural Genome Consortium that targets proteins encoding for epigenetic regulators. This chemical genetic screening identified TP-472, which targets bromodomain-7/9, as the strongest inhibitor of melanoma growth in both short- and long-term survival assays and in mouse models of melanoma tumor growth. Mechanistically, using a transcriptome-wide mRNA sequencing profile we identified TP-472 treatment downregulates genes encoding various extracellular matrix (ECM) proteins, including integrins, collagens, and fibronectins. Reactome-based functional pathway analyses revealed that many of the ECM proteins are involved in extracellular matrix interactions required for cancer cell growth and proliferation. TP-472 treatment also upregulated several pro-apoptotic genes that can inhibit melanoma growth. Collectively, our results identify BRD7/9 inhibitor TP-472 as a potentially useful therapeutic agent for melanoma therapy.
Collapse
Affiliation(s)
- Lawrence David Mason
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL 35322, USA; (L.D.M.); (S.C.); (K.K.R.)
| | - Suresh Chava
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL 35322, USA; (L.D.M.); (S.C.); (K.K.R.)
| | - Kiran Kumar Reddi
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL 35322, USA; (L.D.M.); (S.C.); (K.K.R.)
| | - Romi Gupta
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL 35322, USA; (L.D.M.); (S.C.); (K.K.R.)
- O’Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL 35322, USA
- Correspondence: ; Tel.: +1-205-934-6207
| |
Collapse
|
15
|
Kim DY, Kim SH, Yang EK. RNA interference mediated suppression of TRPV6 inhibits the progression of prostate cancer in vitro by modulating cathepsin B and MMP9 expression. Investig Clin Urol 2021; 62:447-454. [PMID: 34085788 PMCID: PMC8246020 DOI: 10.4111/icu.20200511] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 01/08/2021] [Accepted: 01/21/2021] [Indexed: 12/17/2022] Open
Abstract
Purpose The transient receptor potential vanilloid 6 (TRPV6) channel is overexpressed in prostate cancer and its silencing is known to inhibit the growth of LNCaP cells. However, the role of TRPV6 in the metastasis of prostate cancer cells and its relationship to the invasive markers, matrix metalloproteinase (MMP) and cathepsin B, is unclear. Thus, the present study was focused on understanding these tumor-related processes. Materials and Methods We performed a wound-healing assay and a Transwell migration and invasion assay to assess the migration and invasion of prostate cancer cells. Western blot analysis was used to measure the expression of cathepsin B, MMP2, and MMP9. Results TRPV6 siRNA significantly inhibited the proliferation of LNCaP prostate cancer cells. It also significantly attenuated the wound healing and migration capacities of LNCaP cells. Moreover, the invasiveness of LNCaP cells and the expression of MMP9 and cathepsin B in LNCaP cells were also significantly inhibited by TRPV6 siRNA. Conclusions The results indicate that TRPV6 may promote prostate cancer progression in association with MMP9 and cathepsin B, thereby validating further research into TRPV6 as a useful therapeutic target for local invasion or metastasis of advanced prostate cancer.
Collapse
Affiliation(s)
- Duk Yoon Kim
- Department of Urology, School of Medicine, Daegu Catholic University, Daegu, Korea
| | - Soon Hee Kim
- Department of Physiology, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Eun Kyoung Yang
- Department of Physiology, School of Medicine, Kyungpook National University, Daegu, Korea.
| |
Collapse
|
16
|
Zhang Z, Yue P, Lu T, Wang Y, Wei Y, Wei X. Role of lysosomes in physiological activities, diseases, and therapy. J Hematol Oncol 2021; 14:79. [PMID: 33990205 PMCID: PMC8120021 DOI: 10.1186/s13045-021-01087-1] [Citation(s) in RCA: 126] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 05/03/2021] [Indexed: 02/07/2023] Open
Abstract
Long known as digestive organelles, lysosomes have now emerged as multifaceted centers responsible for degradation, nutrient sensing, and immunity. Growing evidence also implicates role of lysosome-related mechanisms in pathologic process. In this review, we discuss physiological function of lysosomes and, more importantly, how the homeostasis of lysosomes is disrupted in several diseases, including atherosclerosis, neurodegenerative diseases, autoimmune disorders, pancreatitis, lysosomal storage disorders, and malignant tumors. In atherosclerosis and Gaucher disease, dysfunction of lysosomes changes cytokine secretion from macrophages, partially through inflammasome activation. In neurodegenerative diseases, defect autophagy facilitates accumulation of toxic protein and dysfunctional organelles leading to neuron death. Lysosomal dysfunction has been demonstrated in pathology of pancreatitis. Abnormal autophagy activation or inhibition has been revealed in autoimmune disorders. In tumor microenvironment, malignant phenotypes, including tumorigenesis, growth regulation, invasion, drug resistance, and radiotherapy resistance, of tumor cells and behaviors of tumor-associated macrophages, fibroblasts, dendritic cells, and T cells are also mediated by lysosomes. Based on these findings, a series of therapeutic methods targeting lysosomal proteins and processes have been developed from bench to bedside. In a word, present researches corroborate lysosomes to be pivotal organelles for understanding pathology of atherosclerosis, neurodegenerative diseases, autoimmune disorders, pancreatitis, and lysosomal storage disorders, and malignant tumors and developing novel therapeutic strategies.
Collapse
Affiliation(s)
- Ziqi Zhang
- Laboratory 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
| | - Pengfei Yue
- Laboratory 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
| | - Tianqi Lu
- Laboratory 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
- Laboratory 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
- Laboratory 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
- Laboratory 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
|
17
|
Arora M, Pandey G, Chauhan SS. Cysteine Cathepsins and Their Prognostic and Therapeutic Relevance in Leukemia. ANNALS OF THE NATIONAL ACADEMY OF MEDICAL SCIENCES (INDIA) 2021. [DOI: 10.1055/s-0041-1726151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
AbstractCysteine cathepsins are lysosomal proteases that require Cys-His ion pair in their catalytic site for enzymatic activity. While their aberrant expression and oncogenic functions have been widely reported in solid tumors, recent findings suggest that these proteases also play an important role in the pathogenesis of hematological malignancies. In this review, we summarize the potential clinical implications of cysteine cathepsins as diagnostic and prognostic markers in leukemia, and present evidences which supports the utility of these proteases as potential therapeutic targets in hematological malignancies. We also highlight the available information on the expression patterns, regulation, and potential functions of cysteine cathepsins in normal hematopoiesis and hematological malignancies. In hematopoiesis, cysteine cathepsins play a variety of physiological roles including regulation of hematopoietic stem cell adhesion in the bone marrow, trafficking, and maturation. They are also involved in several functions of immune cells which include the selection of lymphocytes in the thymus, antigen processing, and presentation. However, the expression of cysteine cathepsins is dysregulated in hematological malignancies where they have been shown to play diverse functions. Interestingly, several pieces of evidence over the past few years have demonstrated overexpression of cathepsins in leukemia and their association with worst survival outcomes in patients. Strategies aimed at altering the expression, activity, and subcellular localization of these cathepsins are emerging as potential therapeutic modalaties in the management of hematological malignancies. Recent findings also suggest the involvement of these proteases in modulating the immune response in leukemia and lymphomas.
Collapse
Affiliation(s)
- Mohit Arora
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Garima Pandey
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Shyam S. Chauhan
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
- Department of Biotechnology, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
18
|
Pt(IV) Prodrugs with NSAIDs as Axial Ligands. Int J Mol Sci 2021; 22:ijms22083817. [PMID: 33917027 PMCID: PMC8067705 DOI: 10.3390/ijms22083817] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 03/30/2021] [Accepted: 03/31/2021] [Indexed: 12/13/2022] Open
Abstract
A chemo-anti-inflammatory strategy is of interest for the treatment of aggressive cancers. The platinum (IV) prodrug with non-steroidal anti-inflammatory drugs (NSAIDs) as axial ligands is designed to efficiently enter tumor cells due to high lipophilicity and release the cytotoxic metabolite and NSAID intracellularly, thereby reducing side effects and increasing the therapeutic efficacy of platinum chemotherapy. Over the last 7 years, a number of publications have been devoted to the design of such Pt(IV) prodrugs in combination with anti-inflammatory chemotherapy, with high therapeutic efficacy in vitro and In vivo. In this review, we summarize the studies devoted to the development of Pt(IV) prodrugs with NSAIDs as axial ligands, the study of the mechanism of their cytotoxic action and anti-inflammatory activity, the structure-activity ratio, and therapeutic efficacy.
Collapse
|
19
|
The Role of the Metzincin Superfamily in Prostate Cancer Progression: A Systematic-Like Review. Int J Mol Sci 2021; 22:ijms22073608. [PMID: 33808504 PMCID: PMC8036576 DOI: 10.3390/ijms22073608] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 03/21/2021] [Accepted: 03/26/2021] [Indexed: 02/07/2023] Open
Abstract
Prostate cancer remains a leading cause of cancer-related morbidity in men. Potentially important regulators of prostate cancer progression are members of the metzincin superfamily of proteases, principally through their regulation of the extracellular matrix. It is therefore timely to review the role of the metzincin superfamily in prostate cancer and its progression to better understand their involvement in this disease. A systematic-like search strategy was conducted. Articles that investigated the roles of members of the metzincin superfamily and their key regulators in prostate cancer were included. The extracted articles were synthesized and data presented in tabular and narrative forms. Two hundred and five studies met the inclusion criteria. Of these, 138 investigated the role of the Matrix Metalloproteinase (MMP) subgroup, 34 the Membrane-Tethered Matrix Metalloproteinase (MT-MMP) subgroup, 22 the A Disintegrin and Metalloproteinase (ADAM) subgroup, 8 the A Disintegrin and Metalloproteinase with Thrombospondin Motifs (ADAMTS) subgroup and 53 the Tissue Inhibitor of Metalloproteinases (TIMP) family of regulators, noting that several studies investigated multiple family members. There was clear evidence that specific members of the metzincin superfamily are involved in prostate cancer progression, which can be either in a positive or negative manner. However, further understanding of their mechanisms of action and how they may be used as prognostic indicators or molecular targets is required.
Collapse
|
20
|
Gallazzi M, Baci D, Mortara L, Bosi A, Buono G, Naselli A, Guarneri A, Dehò F, Capogrosso P, Albini A, Noonan DM, Bruno A. Prostate Cancer Peripheral Blood NK Cells Show Enhanced CD9, CD49a, CXCR4, CXCL8, MMP-9 Production and Secrete Monocyte-Recruiting and Polarizing Factors. Front Immunol 2021; 11:586126. [PMID: 33569050 PMCID: PMC7868409 DOI: 10.3389/fimmu.2020.586126] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 12/08/2020] [Indexed: 12/24/2022] Open
Abstract
Natural killer (NK) cells, effector lymphocytes of the innate immunity, have been shown to be altered in several cancers, both at tissue and peripheral levels. We have shown that in Non-Small Cell Lung Cancer (NSCLC) and colon cancer, tumour associated circulating NK (TA-NK) and tumour infiltrating NK (TI-NK) exhibit pro-angiogenic phenotype/functions. However, there is still a lack of knowledge concerning the phenotype of peripheral blood (PB) NK (pNK) cells in prostate cancer (PCa). Here, we phenotypically and functionally characterized pNK from PCa patients (PCa TA-NKs) and investigated their interactions with endothelial cells and monocytes/macrophages. NK cell subset distribution in PB of PCa patients was investigated, by multicolor flow cytometry, for surface antigens expression. Protein arrays were performed to characterize the secretome on FACS-sorted pNK cells. Conditioned media (CM) from FACS-sorted PCa pTA-NKs were used to determine their ability to induce pro-inflammatory/pro-angiogenic phenotype/functions in endothelial cells, monocytes, and macrophages. CM from three different PCa (PC-3, DU-145, LNCaP) cell lines, were used to assess their effects on human NK cell polarization in vitro, by multicolor flow cytometry. We found that PCa pTA-NKs acquire the CD56brightCD9+CD49a+CXCR4+ phenotype, increased the expression of markers of exhaustion (PD-1, TIM-3) and are impaired in their degranulation capabilities. Similar effects were observed on healthy donor-derived pNK cells, exposed to conditioned media of three different PCa cell lines, together with increased production of pro-inflammatory chemokines/chemokine receptors CXCR4, CXCL8, CXCL12, reduced production of TNFα, IFNγ and Granzyme-B. PCa TA-NKs released factors able to support inflammatory angiogenesis in an in vitro model and increased the expression of CXCL8, ICAM-1, and VCAM-1 mRNA in endothelial cells. Secretome analysis revealed the ability of PCa TA-NKs to release pro-inflammatory cytokines/chemokines involved in monocyte recruitment and M2-like polarization. Finally, CMs from PCa pTA-NKs recruit THP-1 and peripheral blood CD14+ monocyte and polarize THP-1 and peripheral blood CD14+ monocyte-derived macrophage towards M2-like/TAM macrophages. Our results show that PCa pTA-NKs acquire properties related to the pro-inflammatory angiogenesis in endothelial cells, recruit monocytes and polarize macrophage to an M2-like type phenotype. Our data provides a rationale for a potential use of pNK profiling in PCa patients.
Collapse
Affiliation(s)
- Matteo Gallazzi
- Laboratory of Immunology and General Pathology, Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| | - Denisa Baci
- Laboratory of Immunology and General Pathology, Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| | - Lorenzo Mortara
- Laboratory of Immunology and General Pathology, Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| | - Annalisa Bosi
- Laboratory of Pharmacology, Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | | | - Angelo Naselli
- Unit of Urology, San Giuseppe Hospital, IRCCS MultiMedica, Milan, Italy
| | - Andrea Guarneri
- Unit of Urology, San Giuseppe Hospital, IRCCS MultiMedica, Milan, Italy
| | - Federico Dehò
- S.C. of Urology, ASST Settelaghi, Ospedale di Circolo e Fondazione Macchi, Varese, Italy
| | - Paolo Capogrosso
- S.C. of Urology, ASST Settelaghi, Ospedale di Circolo e Fondazione Macchi, Varese, Italy
| | - Adriana Albini
- Laboratory of Vascular Biology and Angiogenesis, IRCCS MultiMedica, Milano, Italy
| | - Douglas M. Noonan
- Laboratory of Immunology and General Pathology, Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
- Laboratory of Vascular Biology and Angiogenesis, IRCCS MultiMedica, Milano, Italy
| | | |
Collapse
|
21
|
Ranjbar S, Shabanpoor MR, Dehghani Z, Firuzi O, Edraki N, Khoshneviszadeh M. Dihydronaphthalenone chalconoid derivatives as potential cathepsin B inhibitors; design, synthesis, cytotoxicity evaluation and docking analysis. BRAZ J PHARM SCI 2021. [DOI: 10.1590/s2175-979020200004181074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Affiliation(s)
| | | | | | | | | | - Mehdi Khoshneviszadeh
- Shiraz University of Medical Sciences, Iran; Shiraz University of Medical Sciences, Iran
| |
Collapse
|
22
|
Momeny M, Sankanian G, Hamzehlou S, Yousefi H, Esmaeili F, Alishahi Z, Karimi B, Zandi Z, Shamsaiegahkani S, Sabourinejad Z, Kashani B, Nasrollahzadeh A, Mousavipak SH, Mousavi SA, Ghaffari SH. Cediranib, an inhibitor of vascular endothelial growth factor receptor kinases, inhibits proliferation and invasion of prostate adenocarcinoma cells. Eur J Pharmacol 2020; 882:173298. [PMID: 32593665 DOI: 10.1016/j.ejphar.2020.173298] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 06/17/2020] [Accepted: 06/18/2020] [Indexed: 11/29/2022]
Abstract
Prostate Cancer is the second cause of cancer-related death in men and development of metastatic castration-resistant prostate cancer (mCRPC) is the major reason for its high mortality rate. Despite various treatments, all patients succumb to resistant disease, suggesting that there is a pressing need for novel and more efficacious treatments. Members of the vascular endothelial growth factor (VEGF) family play key roles in the tumorigenesis of mCRPC, indicating that VEGF-targeted therapies may have potential anti-tumor efficacy in this malignancy. However, due to compensatory activation of other family members, clinical trials with single-targeted VEGF inhibitors were discouraging. Here, we determined the anti-neoplastic activity of Cediranib, a pan-VEGF receptor inhibitor, in the mCRPC cell lines. Anti-growth effects of Cediranib were studied by MTT and BrdU cell proliferation assays and crystal violet staining. Annexin V/PI, radiation therapy and cell motility assays were carried out to examine the effects of Cediranib on apoptosis, radio-sensitivity and cell motility. Quantitative reverse transcription-PCR (qRT-PCR) and Western blot analyses were conducted to determine the molecular mechanisms underlying the anti-tumor activity of Cediranib. Cediranib decreased cell viability and induced apoptosis via inhibition of the anti-apoptotic proteins. Combination with Cediranib synergistically increased Docetaxel sensitivity and potentiated the effects of radiation therapy. Furthermore, Cediranib impaired cell motility via decrease in the expression of the epithelial-to-mesenchymal transition markers. These findings suggest that Cediranib may have anti-tumor activity in mCRPC cells and warrant further investigation on the therapeutic activity of this pan-VEGF receptor inhibitor in mCRPC.
Collapse
Affiliation(s)
- Majid Momeny
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland.
| | - Ghazaleh Sankanian
- Hematology/Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Sepideh Hamzehlou
- Hematology/Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hassan Yousefi
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, New Orleans, USA
| | - Fatemeh Esmaeili
- Hematology/Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Zivar Alishahi
- Hematology/Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Behnaz Karimi
- Hematology/Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Zandi
- Hematology/Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Sahar Shamsaiegahkani
- Hematology/Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Sabourinejad
- Hematology/Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Bahareh Kashani
- Hematology/Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Nasrollahzadeh
- Hematology/Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyyedeh H Mousavipak
- Hematology/Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed A Mousavi
- Hematology/Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed H Ghaffari
- Hematology/Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
23
|
Zhao Z, Liang S, Sun F. LncRNA DLX6-AS1 Promotes Malignant Phenotype and Lymph Node Metastasis in Prostate Cancer by Inducing LARGE Methylation. Front Oncol 2020; 10:1172. [PMID: 32850336 PMCID: PMC7424052 DOI: 10.3389/fonc.2020.01172] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 06/09/2020] [Indexed: 01/12/2023] Open
Abstract
Long non-coding RNAs (lncRNAs) have recently become recognized as crucial players in cancer cellular events including proliferation, migration, and invasion. Herein, we investigated the potential role of lncRNA DLX6-AS1 in prostate cancer cell malignant behaviors and lymph node metastasis. A differentially expressed lncRNA DLX6-AS1 and its downstream regulatory gene (LARGE) were predicted by analysis in silico. RT-qPCR and western blot analysis results demonstrated that DLX6-AS1 was highly expressed, but LARGE was poorly expressed in prostate cancer tissues and cells. The online website indicated that DLX6-AS1 negatively targeted LARGE expression, which was validated by Pearson correlation analysis and MSP. ChIP, RIP, and RNA pull-down assays further suggested that DLX6-AS1 downregulated LARGE expression through recruitment of DNMT1 to its promoter. We induced DLX6-AS1/LARGE overexpression or knockdown to examine their effects through Edu and Transwell assays, which revealed that DLX6-AS1 overexpression accelerated proliferation, invasion, and migration of prostate cancer cells, and that overexpression of LARGE rescued these effects. Tumors xenografts studies confirmed that DLX6-AS1 promoted lymph node metastasis by regulating LARGE, as evidenced by enhanced expression of MMP-9, uPAR, and cathepsin B. In summary, DLX6-AS1 stimulated prostate cancer malignant progression and lymph node metastasis by inducing DNMT1-mediated LARGE methylation, highlighting a potential therapeutic target against prostate cancer.
Collapse
Affiliation(s)
- Zhifeng Zhao
- Department of Urology, Linyi People's Hospital of Shandong Province, Linyi, China
| | - Shuxia Liang
- Special Needs Ward, Linyi People's Hospital of Shandong Province, Linyi, China
| | - Fuguang Sun
- Department of Urology, Linyi People's Hospital of Shandong Province, Linyi, China
| |
Collapse
|
24
|
Kurmi BD, Patel P, Paliwal R, Paliwal SR. Molecular approaches for targeted drug delivery towards cancer: A concise review with respect to nanotechnology. J Drug Deliv Sci Technol 2020. [DOI: 10.1016/j.jddst.2020.101682] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
25
|
Hölzen L, Parigiani MA, Reinheckel T. Tumor cell- and microenvironment-specific roles of cysteine cathepsins in mouse models of human cancers. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2020; 1868:140423. [PMID: 32247787 DOI: 10.1016/j.bbapap.2020.140423] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 03/20/2020] [Accepted: 03/29/2020] [Indexed: 12/22/2022]
Abstract
The human genome encodes for 11 papain-like endolysosomal cysteine peptidases, collectively known as the cysteine cathepsins. Based on their biochemical properties and with the help of experiments in cell culture, the cysteine cathepsins have acquired a reputation as promotors of progression and metastasis of various cancer entities. However, tumors are known to be complex tissues in which non-cancerous cells are also critical for tumorigenesis. Here we discuss the results of the intense investigation of cathepsins in mouse models of human cancers. We focus on models in immunocompetent mice, because only such models allow for analysis of cathepsins in a fully functional tumor microenvironment. An important outcome of those studies was the identification of cancer-promoting cathepsins in tumor-associated macrophages. Another interesting outcome of these animal studies was the identification of a homeostatic tumor-suppressive role for cathepsin L in skin and intestinal cancers. Taken together, these in vivo findings provide a basis for the use of cysteine cathepsins as therapeutic targets, prodrug activators, or as proteases for imaging tumors.
Collapse
Affiliation(s)
- Lena Hölzen
- Institute of Molecular Medicine and Cell Research, Medical Faculty, University of Freiburg, Freiburg, Germany; Faculty of Biology, University of Freiburg, Freiburg, Germany; German Cancer Research Center (DKFZ), Heidelberg, German Cancer Consortium (DKTK), Partner Site, Freiburg, Germany
| | - Maria Alejandra Parigiani
- Institute of Molecular Medicine and Cell Research, Medical Faculty, University of Freiburg, Freiburg, Germany; Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Thomas Reinheckel
- Institute of Molecular Medicine and Cell Research, Medical Faculty, University of Freiburg, Freiburg, Germany; German Cancer Research Center (DKFZ), Heidelberg, German Cancer Consortium (DKTK), Partner Site, Freiburg, Germany; BIOSS Centre for Biological Signalling Studies, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
26
|
Naruishi K. Carotenoids and Periodontal Infection. Nutrients 2020; 12:nu12010269. [PMID: 31968635 PMCID: PMC7019381 DOI: 10.3390/nu12010269] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 01/03/2020] [Accepted: 01/16/2020] [Indexed: 02/07/2023] Open
Abstract
Periodontitis is a polymicrobial infectious disease that leads to inflammation of the gingiva, resulting in teeth loss by various causes such as inflammation-mediated bone resorption. Recently, many investigators have reported that the periodontitis resulting from persistent low-grade infection of Gram-negative bacteria such as Porphyromonas gingivalis (Pg) is associated with increased atherosclerosis, diabetes mellitus, and other systemic diseases through blood stream. On the other hand, carotenoids belong among phytochemicals that are responsible for different colors of the foods. It is important to examine whether carotenoids are effective to the inhibition of periodontal infection/inflammation cascades. This review summarizes the advanced state of knowledge about suppression of periodontal infection by several carotenoids. A series of findings suggest that carotenoids intake may provide novel strategy for periodontitis treatment, although further study will be needed.
Collapse
Affiliation(s)
- Koji Naruishi
- Department of Periodontology and Endodontology, Tokushima University Graduate School of Biomedical Sciences, 3-18-15 Kuramoto, Tokushima 770-8504, Japan
| |
Collapse
|
27
|
Lee MG, Lee KS, Nam KS. Anti‑metastatic effects of arctigenin are regulated by MAPK/AP‑1 signaling in 4T‑1 mouse breast cancer cells. Mol Med Rep 2020; 21:1374-1382. [PMID: 32016480 DOI: 10.3892/mmr.2020.10937] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 10/29/2019] [Indexed: 11/06/2022] Open
Abstract
Arctigenin is a natural lignan that is found in burdock with anti‑viral, ‑oxidative, ‑inflammatory and anti‑tumor activities. In the current study, the effect of arctigenin on metastatic potential was examined in 4T‑1 mouse triple‑negative breast cancer cells. The results indicated that arctigenin inhibited cell motility and invasiveness, which was determined using wound healing and transwell invasion assays. Arctigenin suppressed matrix metalloprotease‑9 (MMP‑9) activity via gelatin zymography, and protein expression of cyclooxygenase‑2 (COX‑2) and MMP‑3. Furthermore, arctigenin attenuated the mRNA expression of metastatic factors, including MMP‑9, MMP‑3 and COX‑2. Based on these results, the effect of arctigenin on the mitogen‑activated protein kinase (MAPK)/activating protein‑1 (AP‑1) signaling pathway was assessed in an attempt to identify the regulatory mechanism responsible for its anti‑metastatic effects. Arctigenin was demonstrated to inhibit the phosphorylation of extracellular signal‑regulated protein kinase (ERK) and c‑Jun N‑terminal kinase (JNK), and the nuclear translocations of the AP‑1 subunits, c‑Jun and c‑Fos. In summary, the present study demonstrated that in 4T‑1 mouse triple‑negative breast cancer cells the anti‑metastatic effect of arctigenin is mediated by the inhibition of MMP‑9 activity and by the inhibition of the metastasis‑enhancing factors MMP‑9, MMP‑3 and COX‑2, due to the suppression of the MAPK/AP‑1 signaling pathway. The results of the current study demonstrated that arctigenin exhibits a potential for preventing cell migration and invasion in triple negative breast cancer.
Collapse
Affiliation(s)
- Min-Gu Lee
- Department of Pharmacology and Intractable Disease Research Center, School of Medicine, Dongguk University, Gyeongju 38066, Republic of Korea
| | - Kyu-Shik Lee
- Department of Pharmacology and Intractable Disease Research Center, School of Medicine, Dongguk University, Gyeongju 38066, Republic of Korea
| | - Kyung-Soo Nam
- Department of Pharmacology and Intractable Disease Research Center, School of Medicine, Dongguk University, Gyeongju 38066, Republic of Korea
| |
Collapse
|
28
|
Pandey G, Bakhshi S, Kumar M, Thakur B, Jain P, Kaur P, Chauhan SS. Prognostic and therapeutic relevance of cathepsin B in pediatric acute myeloid leukemia. Am J Cancer Res 2019; 9:2634-2649. [PMID: 31911851 PMCID: PMC6943344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 10/18/2019] [Indexed: 06/10/2023] Open
Abstract
AML, the second most common childhood leukemia is also one of the deadliest cancers. High mortality rate in AML is due to high incidence of relapse after complete remission with chemotherapy and inadequate prognostic assessment of patients. Moreover, there is dearth of therapeutic targets for treatment of this malignancy. Previous pilot study (n = 24) by our group revealed strong association between cathepsin B (CTSB) overexpression in peripheral blood mononuclear cells (PBMCs) and poor survival outcome in pediatric AML patients. To further explore the clinical utility and role of this protease in pediatric AML, we measured its enzymatic activity and mRNA expression in PBMCs as well as bone marrow mononuclear cells (BMMCs) of patients (n = 101) and PBMCs of healthy controls. Our results revealed elevated CTSB activity (P < 0.01) and overexpression of its mRNA (P < 0.01) in AML patients. Interestingly CTSB in BMMCs of patients emerged as an independent prognostic marker when compared with other known risk factors. Moreover, chemical inhibition of CTSB activity compromised survival, and induced apoptosis in an AML cell line THP-1. We further demonstrate the inhibition of CTSB activity by chemotherapeutic agent doxorubicin in these cells. Docking and simulation studies suggested the binding of doxorubicin to CTSB with higher affinity than its known specific inhibitor CA-074 Me, thereby indicating that cell death induced by this drug may at least partly be mediated by CTSB inhibition. CTSB, therefore, may serve as a prognostic marker and an attractive chemotherapeutic target in pediatric AML.
Collapse
Affiliation(s)
- Garima Pandey
- Department of Biochemistry, All India Institute of Medical SciencesNew Delhi, India
| | - Sameer Bakhshi
- Department of Medical Oncology, All India Institute of Medical SciencesNew Delhi, India
| | - Manoj Kumar
- Department of Biophysics, All India Institute of Medical SciencesNew Delhi, India
| | - Bhaskar Thakur
- Department of Biostatistics, All India Institute of Medical SciencesNew Delhi, India
| | - Prerna Jain
- Department of Biochemistry, All India Institute of Medical SciencesNew Delhi, India
| | - Punit Kaur
- Department of Biophysics, All India Institute of Medical SciencesNew Delhi, India
| | - Shyam S Chauhan
- Department of Biochemistry, All India Institute of Medical SciencesNew Delhi, India
| |
Collapse
|
29
|
Momeny M, Alishahi Z, Eyvani H, Esmaeili F, Zaghal A, Ghaffari P, Tavakkoly-Bazzaz J, Alimoghaddam K, Ghavamzadeh A, Ghaffari SH. Anti-tumor activity of cediranib, a pan-vascular endothelial growth factor receptor inhibitor, in pancreatic ductal adenocarcinoma cells. Cell Oncol (Dordr) 2019; 43:81-93. [DOI: 10.1007/s13402-019-00473-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/30/2019] [Indexed: 12/18/2022] Open
|
30
|
Wang Y, Li B, Zhang X. Scutellaria barbata D. Don (SBD) protects oxygen glucose deprivation/reperfusion-induced injuries of PC12 cells by up-regulating Nrf2. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2019; 47:1797-1807. [PMID: 31062620 DOI: 10.1080/21691401.2019.1610413] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
This study aimed to investigate the potential effect of Scutellaria barbata D. Don (SBD) on oxygen glucose deprivation/reperfusion (OGD/R)-injured PC12 cells. PC12 cells were pretreated with various concentrations of 0.1-0.8 mg/ml SBD for indicated times (12-48 h) and then subjected to OGD/R injury. Cell viability, apoptosis and proliferation were detected using MTT assay, flow cytometry, Ki67 staining and western blot. Oxidative damage was assessed by detecting MDA content, SOD activity and GSH levels. The mitochondrial membrane potential (Δψm) was measured by Rh123 staining. Western blot was performed to assess the expression levels of Nrf2 and PI3K/AKT pathway-related proteins. We found that SBD pretreatment promoted cell viability and proliferation but inhibited apoptosis of OGD/R-injured PC12 cells in dosage- and time-dependent manner. Meanwhile, SBD attenuated oxidative damage and restored mitochondria dysfunction, as evidenced by the reduced MDA content, the increased SOD and GSH levels, and the increased Δψm. Furthermore, SBD induced the expression of Nrf2 in a PI3K/AKT-dependent signalling. Knockdown of Nrf2 blocked the protective effects of SBD on PC12 cells. In conclusion, this study demonstrates that SBD pretreatment protects PC12 cells against OGD/R-induced injury. The potential mechanism may be through up-regulating the expression of Nrf2 in a PI3K/AKT-dependent pathway.
Collapse
Affiliation(s)
- Yanhua Wang
- a Department of Critical Care Medicine , Jining No.1 People's Hospital , Jining , China.,b Affiliated Jining No.1 People's Hospital of Jining Medical University, Jining Medical University , Jining , China
| | - Bo Li
- a Department of Critical Care Medicine , Jining No.1 People's Hospital , Jining , China
| | - Xiaofen Zhang
- a Department of Critical Care Medicine , Jining No.1 People's Hospital , Jining , China
| |
Collapse
|
31
|
McGuire CM, Collins MP, Sun-Wada G, Wada Y, Forgac M. Isoform-specific gene disruptions reveal a role for the V-ATPase subunit a4 isoform in the invasiveness of 4T1-12B breast cancer cells. J Biol Chem 2019; 294:11248-11258. [PMID: 31167791 PMCID: PMC6643023 DOI: 10.1074/jbc.ra119.007713] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 05/30/2019] [Indexed: 12/17/2022] Open
Abstract
The vacuolar H+-ATPase (V-ATPase) is an ATP-driven proton pump present in various intracellular membranes and at the plasma membrane of specialized cell types. Previous work has reported that plasma membrane V-ATPases are key players in breast cancer cell invasiveness. The two subunit a-isoforms known to target the V-ATPase to the plasma membrane are a3 and a4, and expression of a3 has been shown to correlate with plasma membrane localization of the V-ATPase in various invasive human breast cancer cell lines. Here we analyzed the role of subunit a-isoforms in the invasive mouse breast cancer cell line, 4T1-12B. Quantitation of mRNA levels for each isoform by quantitative RT-PCR revealed that a4 is the dominant isoform expressed in these cells. Using a CRISPR/Cas9-based approach to disrupt the genes encoding each of the four V-ATPase subunit a-isoforms, we found that ablation of only the a4-encoding gene significantly inhibits invasion and migration of 4T1-12B cells. Additionally, cells with disrupted a4 exhibited reduced V-ATPase expression at the leading edge, suggesting that the a4 isoform is primarily responsible for targeting the V-ATPase to the plasma membrane in 4T1-12B cells. These findings suggest that different subunit a-isoforms may direct V-ATPases to the plasma membrane of different invasive breast cancer cell lines. They further suggest that expression of V-ATPases at the cell surface is the primary factor that promotes an invasive cancer cell phenotype.
Collapse
Affiliation(s)
- Christina M McGuire
- Department of Developmental, Molecular, and Chemical Biology, Tufts University School of Medicine, Tufts University, Boston, Massachusetts 02111
- Program in Biochemistry, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, Massachusetts 02111
| | - Michael P Collins
- Department of Developmental, Molecular, and Chemical Biology, Tufts University School of Medicine, Tufts University, Boston, Massachusetts 02111
- Program in Cell, Molecular, and Developmental Biology, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, Massachusetts 02111
| | - GeHong Sun-Wada
- Department of Biochemistry, Faculty of Pharmaceutical Sciences, Doshisha Women's College, Kyotanabe, Kyoto 610-0395, Japan
| | - Yoh Wada
- Division of Biological Science, Institute of Scientific and Industrial Research, Osaka University, Ibaraki, Osaka 567-0047, Japan
| | - Michael Forgac
- Department of Developmental, Molecular, and Chemical Biology, Tufts University School of Medicine, Tufts University, Boston, Massachusetts 02111
- Program in Biochemistry, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, Massachusetts 02111
- Program in Cell, Molecular, and Developmental Biology, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, Massachusetts 02111
| |
Collapse
|
32
|
Kalmouni M, Al-Hosani S, Magzoub M. Cancer targeting peptides. Cell Mol Life Sci 2019; 76:2171-2183. [PMID: 30877335 PMCID: PMC11105397 DOI: 10.1007/s00018-019-03061-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 02/08/2019] [Accepted: 03/07/2019] [Indexed: 12/19/2022]
Abstract
Despite continuing advances in the development of biomacromolecules for therapeutic purposes, successful application of these often large and hydrophilic molecules has been hindered by their inability to efficiently traverse the cellular plasma membrane. In recent years, cell-penetrating peptides (CPPs) have received considerable attention as a promising class of delivery vectors due to their ability to mediate the efficient import of a large number of cargoes in vitro and in vivo. However, the lack of target specificity of CPPs remains a major obstacle to their clinical development. To address this issue, researchers have developed strategies in which chemotherapeutic drugs are conjugated to cancer targeting peptides (CTPs) that exploit the unique characteristics of the tumor microenvironment or cancer cells, thereby improving cancer cell specificity. This review highlights several of these strategies that are currently in use, and discusses how multi-component nanoparticles conjugated to CTPs can be designed to provide a more efficient cancer therapeutic delivery strategy.
Collapse
Affiliation(s)
- Mona Kalmouni
- Biology Program, New York University Abu Dhabi, PO Box 129188, Saadiyat Island Campus, Abu Dhabi, United Arab Emirates
| | - Sumaya Al-Hosani
- Biology Program, New York University Abu Dhabi, PO Box 129188, Saadiyat Island Campus, Abu Dhabi, United Arab Emirates
| | - Mazin Magzoub
- Biology Program, New York University Abu Dhabi, PO Box 129188, Saadiyat Island Campus, Abu Dhabi, United Arab Emirates.
| |
Collapse
|
33
|
Mijanović O, Branković A, Panin AN, Savchuk S, Timashev P, Ulasov I, Lesniak MS. Cathepsin B: A sellsword of cancer progression. Cancer Lett 2019; 449:207-214. [PMID: 30796968 PMCID: PMC6488514 DOI: 10.1016/j.canlet.2019.02.035] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 02/12/2019] [Accepted: 02/13/2019] [Indexed: 12/13/2022]
Abstract
Clinical, biochemical and molecular biology studies have identified lysosome-encapsulated cellular proteases as critical risk factors for cancer progression. Cathepsins represent a group of such proteases aimed at maintenance of cellular homeostasis. Nevertheless, recent reports suggest that Cathepsin B executes other cellular programs such as controlling tumor growth, migration, invasion, angiogenesis, and metastases development. In fact, elevated levels of Cathepsins are found under different pathological conditions including inflammation, infection, neurodegenerative disease, and cancer. Furthermore, the discovery of Cathepsin B secretion and function as an extracellular matrix protein has broadened our appreciation for the impact of Cathepsin B on cancer progression. Underneath a façade of an intracellular protease with limited therapeutic potential hides a central role of cathepsins in extracellular functions. Moreover, this role is incredibly diverse from one condition to the next - from driving caspase-dependent apoptosis to facilitating tumor neovascularization and metastasis. Here we discuss the role of Cathepsin B in the oncogenic process and perspective the use of Cathepsin B for diagnostic and therapeutic applications.
Collapse
Affiliation(s)
- Olja Mijanović
- Institute of Regenerative Medicine, Sechenov First Moscow State Medical University, Moscow, 119991, Russia
| | - Ana Branković
- Department of Forensics, Academy of Criminalistic and Police Studies, Belgrade, Serbia
| | - Alexander N Panin
- Moscow State University of Food Production, 11, Volokolamskoe Shosse, Moscow, 125080, Russia
| | - Solomiia Savchuk
- The University of Illinois at Chicago (UIC), Chicago, IL, USA; Northwestren University, Chicago, IL, 60611, USA
| | - Peter Timashev
- Institute of Regenerative Medicine, Sechenov First Moscow State Medical University, Moscow, 119991, Russia
| | - Ilya Ulasov
- Institute of Regenerative Medicine, Sechenov First Moscow State Medical University, Moscow, 119991, Russia
| | - Maciej S Lesniak
- The University of Illinois at Chicago (UIC), Chicago, IL, USA; Northwestren University, Chicago, IL, 60611, USA.
| |
Collapse
|
34
|
Zhang Y, Zhang C, Min D. Ailanthone up-regulates miR-449a to restrain acute myeloid leukemia cells growth, migration and invasion. Exp Mol Pathol 2019; 108:114-120. [PMID: 31002772 DOI: 10.1016/j.yexmp.2019.04.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 02/25/2019] [Accepted: 04/16/2019] [Indexed: 01/27/2023]
Abstract
BACKGROUND Ailanthone (AIL) is a quassinoid isolated from traditional Chinese herbal medicine Ailanthus altissima. The anti-tumor activities of AIL have been reported in various solid tumors. This study aimed to reveal the in vitro effect of AIL on acute myeloid leukemia (AML) cells. METHODS The effects of AIL on five AML cell lines (KG1, HL60, U-937, THP-1 and OCI-AML2) as well as myeloid progenitor cells were evaluated by performing CCK-8 assay, flow cytometry, Transwell assay and Western blotting. KG1 and HL60 cells were transfected with miR-449a inhibitor or its negative control, and then were treated by AIL. The above mentioned assays were performed again to study the involvement of miR-449a in AIL's function. RESULTS AIL dose-dependently inhibited the viability of AML cells and myeloid progenitor cells. The IC50 value of AIL towards KG1 and HL60 cells was 0.58 and 0.57 μM, respectively. AIL with concentration of 0.5 μM significantly induced the apoptosis of AML cells rather than myeloid progenitor cells. Meanwhile, 0.5 μM AIL significantly reduced migration and invasion of AML cells. miR-449a was highly expressed in response to the treatment of 0.5 μM AIL. Besides this, the anti-tumor activities of AIL in AML cells were attenuated by miR-449a silence. Further, the blockage of Notch and PI3K/AKT signaling pathways induced by AIL was reversed by miR-449a silence. CONCLUSION AIL restrained AML cells growth, migration and invasion through up-regulation of miR-449a, and deactivation of Notch and PI3K/AKT signaling pathways.
Collapse
MESH Headings
- Apoptosis/drug effects
- Cell Cycle/drug effects
- Cell Growth Processes/drug effects
- Cell Line, Tumor
- Cell Movement/drug effects
- Drugs, Chinese Herbal/pharmacology
- Humans
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Neoplasm Invasiveness
- Phosphatidylinositol 3-Kinases/metabolism
- Quassins/pharmacology
- Receptors, Notch/metabolism
- Signal Transduction/drug effects
- Up-Regulation/drug effects
Collapse
Affiliation(s)
- Yang Zhang
- Department of Hematology, Jining No.1 People's Hospital, Jining 272000, Shandong, China.; Affiliated Jining No.1 People's Hospital of Jining Medical University, Jining Medical University, Jining 272067, Shandong, China
| | - Chunzhi Zhang
- Department of Clinical Laboratory, Jining No.1 People's Hospital, Jining 272000, Shandong, China
| | - Dejin Min
- Department of Hepatobiliary Surgery, Jining No.1 People's Hospital, Jining 272000, Shandong, China.
| |
Collapse
|
35
|
Wach S, Brandl M, Borchardt H, Weigelt K, Lukat S, Nolte E, Al-Janabi O, Hart M, Grässer F, Giedl J, Jung R, Stöhr R, Hartmann A, Lieb V, Höbel S, Peters A, Stäubert C, Wullich B, Taubert H, Aigner A. Exploring the MIR143-UPAR Axis for the Inhibition of Human Prostate Cancer Cells In Vitro and In Vivo. MOLECULAR THERAPY. NUCLEIC ACIDS 2019; 16:272-283. [PMID: 30933831 PMCID: PMC6444223 DOI: 10.1016/j.omtn.2019.02.020] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 01/16/2019] [Accepted: 02/21/2019] [Indexed: 01/12/2023]
Abstract
MIR143 is pathologically downregulated and may function as a tumor suppressor in prostate cancer. Likewise, the urokinase plasminogen activator receptor (UPAR) is overexpressed in prostate carcinoma, representing a negative prognostic marker and putative therapeutic target gene. In this paper, we establish UPAR as a new direct target of MIR143. Luciferase reporter gene constructs identify one of the two in silico-predicted binding sites as functionally relevant for direct MIR143 binding to the 3′ UTR, and, concomitantly, transfection of MIR143 reduces UPAR protein levels in prostate carcinoma cells in vitro. Inhibitory effects on cell proliferation and colony formation, spheroid growth and integrity, and cell viability are extensively analyzed, and they are compared to direct small interfering RNA (siRNA)-mediated uPAR knockdown or combined microRNA (miRNA)-siRNA treatment. Switching to a therapeutically more relevant in vivo model, we demonstrate tumor-inhibitory effects of MIR143 replacement therapy by systemic treatment of mice bearing subcutaneous PC-3 tumor xenografts with MIR143 formulated in polymeric nanoparticles. This efficient, nanoparticle-mediated delivery of intact MIR143 mediates the marked downregulation of uPAR protein, but not mRNA levels, thus indicating translational inhibition rather than mRNA degradation. In summary, we identify UPAR as a direct target gene of MIR143, and we establish the therapeutic anti-tumor potential of nanoparticle-based MIR143 replacement in prostate cancer.
Collapse
Affiliation(s)
- Sven Wach
- Department of Urology, Friedrich Alexander University Hospital Erlangen-Nürnberg, Erlangen, Germany
| | - Madeleine Brandl
- Rudolf-Boehm-Institute for Pharmacology and Toxicology, Clinical Pharmacology, University of Leipzig, Leipzig, Germany
| | - Hannes Borchardt
- Rudolf-Boehm-Institute for Pharmacology and Toxicology, Clinical Pharmacology, University of Leipzig, Leipzig, Germany
| | - Katrin Weigelt
- Department of Urology, Friedrich Alexander University Hospital Erlangen-Nürnberg, Erlangen, Germany
| | - Sabine Lukat
- Department of Urology, Friedrich Alexander University Hospital Erlangen-Nürnberg, Erlangen, Germany
| | - Elke Nolte
- Department of Urology, Friedrich Alexander University Hospital Erlangen-Nürnberg, Erlangen, Germany
| | - Omar Al-Janabi
- Department of Urology, Friedrich Alexander University Hospital Erlangen-Nürnberg, Erlangen, Germany
| | - Martin Hart
- Institute of Virology, University of Saarland Medical School, Kirrbergerstrasse, Homburg/Saar, Germany
| | - Friedrich Grässer
- Institute of Virology, University of Saarland Medical School, Kirrbergerstrasse, Homburg/Saar, Germany
| | - Johannes Giedl
- Institute of Pathology, Friedrich Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Rudolf Jung
- Institute of Pathology, Friedrich Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Robert Stöhr
- Institute of Pathology, Friedrich Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Arndt Hartmann
- Institute of Pathology, Friedrich Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Verena Lieb
- Department of Urology, Friedrich Alexander University Hospital Erlangen-Nürnberg, Erlangen, Germany
| | - Sabrina Höbel
- Rudolf-Boehm-Institute for Pharmacology and Toxicology, Clinical Pharmacology, University of Leipzig, Leipzig, Germany
| | - Anna Peters
- Rudolf-Schönheimer-Institute for Biochemistry, University of Leipzig, Leipzig, Germany
| | - Claudia Stäubert
- Rudolf-Schönheimer-Institute for Biochemistry, University of Leipzig, Leipzig, Germany
| | - Bernd Wullich
- Department of Urology, Friedrich Alexander University Hospital Erlangen-Nürnberg, Erlangen, Germany
| | - Helge Taubert
- Department of Urology, Friedrich Alexander University Hospital Erlangen-Nürnberg, Erlangen, Germany.
| | - Achim Aigner
- Rudolf-Boehm-Institute for Pharmacology and Toxicology, Clinical Pharmacology, University of Leipzig, Leipzig, Germany
| |
Collapse
|
36
|
Down-regulation of cathepsin S and matrix metalloproteinase-9 via Src, a non-receptor tyrosine kinase, suppresses triple-negative breast cancer growth and metastasis. Exp Mol Med 2018; 50:1-14. [PMID: 30185799 PMCID: PMC6123788 DOI: 10.1038/s12276-018-0135-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 05/28/2018] [Indexed: 02/07/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is a highly metastatic breast cancer with poor prognosis. In the present study, we demonstrated that Src, a non-receptor tyrosine kinase, might provide an effective therapeutic strategy to overcome TNBC invasion and metastasis, which are mediated via the synergistic action of the lysosomal enzyme cathepsin S (CTSS) and gelatinase MMP-9. Knock-down of MMP-9 and CTSS using siRNAs resulted in a synergistic suppression of MDA-MB-231 cell invasion, which was similarly observed with pharmacological inhibitors. During the screening of new drug candidates that suppress both CTSS and MMP-9, BJ-2302, a novel 7-azaindolin-2-one derivative, was discovered. Src, an upstream activator of both pathways (PI3K/Akt and Ras/Raf/ERK) responsible for the expression of CTSS and MMP-9, was identified as a high-affinity target of BJ-2302 (IC90: 3.23 µM) through a Src kinase assay and a drug affinity responsive target stability (DARTS) assay. BJ-2302 effectively suppressed MDA-MB-231 cell invasion (Matrigel invasion assay) and metastasis (chorioallantoic membrane assay xenografted with MDA-MB-231-luc2-tdTomato cancer cells). Unlike Z-FL-COCHO (potent CTSS inhibitor), BJ-2302 did not induce any cytotoxicity in MCF-10A normal breast epithelial cells. Additionally, BJ-2302 (1 mg/kg) strongly suppressed TNBC cell proliferation in vitro and tumor growth in a xenograft mouse tumor model. The anti-metastatic and anti-tumor effects of BJ-2302 were superior to those of Z-FL-COCHO (1 mg/kg) or batimastat (30 mg/kg), a pan-MMP inhibitor. In summary, inhibition of Src kinase suppressed TNBC tumor growth and metastasis, and Src inhibitors such as BJ-2302 may constitute a novel therapeutic tool to treat breast cancer that expresses high levels of CTSS and MMP-9.
Collapse
|
37
|
Cheng L, Zang J, Dai HJ, Li F, Guo F. Ubiquitin ligase CHIP functions as an oncogene and activates the AKT signaling pathway in prostate cancer. Int J Oncol 2018; 53:203-214. [PMID: 29693147 DOI: 10.3892/ijo.2018.4377] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 04/12/2018] [Indexed: 11/06/2022] Open
Abstract
Carboxyl terminus of Hsc-70-interacting protein (CHIP) is an E3 ubiquitin ligase that induces the ubiquitination and degradation of numerous tumor-associated proteins and serves as a suppressor or promoter in tumor progression. To date, the molecular mechanism of CHIP in prostate cancer remains unknown. Therefore, the present study investigated the biological function of CHIP in prostate cancer cells and obtained evidence that CHIP expression is upregulated in prostate cancer tissues. The CHIP vector was introduced into DU145 cancer cells and the cell biological behaviour was examined through a series of experiments, including cell growth, cell apoptosis and migration and invasion assays. The results indicated that the overexpression of CHIP in DU145 prostatic cancer cells promoted cell proliferation through activation of the protein kinase B (AKT) signaling pathway, which subsequently increased cyclin D1 protein levels and decreased p21 and p27 protein levels. The overexpression of CHIP significantly increased the migration and invasion of the DU145 cells, which is possible due to activation of the AKT signaling pathway and upregulation of vimentin. The expression level of CHIP was observed to be increased in human prostate cancer tissues compared with the adjacent normal tissue. Furthermore, the CHIP expression level exhibited a positively association with the Gleason score of the patents. These findings indicate that CHIP functions as an oncogene in prostate cancer.
Collapse
Affiliation(s)
- Li Cheng
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Jin Zang
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Han-Jue Dai
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Feng Li
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Feng Guo
- Department of Oncology, Nanjing Medical University Affiliated Suzhou Hospital, Suzhou, Jiangsu 215001, P.R. China
| |
Collapse
|
38
|
Naruishi K, Nagata T. Biological effects of interleukin‐6 on Gingival Fibroblasts: Cytokine regulation in periodontitis. J Cell Physiol 2018; 233:6393-6400. [DOI: 10.1002/jcp.26521] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 01/30/2018] [Indexed: 12/21/2022]
Affiliation(s)
- Koji Naruishi
- Department of Periodontology and Endodontology, Institute of Biomedical SciencesTokushima University Graduate SchoolKuramotoTokushimaJapan
| | - Toshihiko Nagata
- Department of Periodontology and Endodontology, Institute of Biomedical SciencesTokushima University Graduate SchoolKuramotoTokushimaJapan
| |
Collapse
|
39
|
Zhao Y, Tang H, Zeng X, Ye D, Liu J. Resveratrol inhibits proliferation, migration and invasion via Akt and ERK1/2 signaling pathways in renal cell carcinoma cells. Biomed Pharmacother 2017; 98:36-44. [PMID: 29241073 DOI: 10.1016/j.biopha.2017.12.029] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 11/24/2017] [Accepted: 12/07/2017] [Indexed: 02/04/2023] Open
Abstract
Recent studies have shown that resveratrol (RES) inhibits cancer cell growth, migration and invasion. Here, we evaluated RES in two human renal cell carcinoma (RCC) cell lines, ACHN and A498. We investigated the effects of RES on proliferation, cell morphology, colony formation, migration, and invasion. We used a proliferation assay to demonstrate that RES inhibited cell growth with IC50 values 132.9±1.064μM in ACHN, and 112.8±1.191μM in A498, respectively. Using inverted contrast microscopy, we showed that RES reduced cell-to-cell contact and inhibited formation of filopodia. A wound healing assay showed that RES inhibited migration of RCC cells. A Transwell assay showed that RES inhibited RCC migration and invasion. Western blot analysis showed that RES suppresses expression of N-cadherin, Vimentin, Snail, MMP-2, MMP-9, p-Akt and p-ERK1/2, but increased expression of E-cadherin and TIMP-1. In the presence of PD98059, the inhibitor of ERK1/2 pathway, we repeated all of the above experiments, showed that RES acted via the ERK1/2 pathway. Taken together, our results suggested that RES suppressed RCC cell proliferation, migration, and invasion in a concentration- and time-dependent manner. These effects likely resulted from inactivation of the Akt and ERK1/2 signaling pathways.
Collapse
Affiliation(s)
- Yuwan Zhao
- Laboratory of Urology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Huancheng Tang
- Laboratory of Urology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Xin Zeng
- Laboratory of Urology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Dongcai Ye
- Laboratory of Urology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Jianjun Liu
- Laboratory of Urology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China.
| |
Collapse
|
40
|
Sharma A, Mendonca J, Ying J, Kim H, Verdone JE, Zarif JC, Carducci M, Hammers H, Pienta KJ, Kachhap S. The prostate metastasis suppressor gene NDRG1 differentially regulates cell motility and invasion. Mol Oncol 2017; 11:655-669. [PMID: 28371345 PMCID: PMC5467496 DOI: 10.1002/1878-0261.12059] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 03/20/2017] [Accepted: 03/21/2017] [Indexed: 12/12/2022] Open
Abstract
Experimental and clinical evidence suggests that N-myc downregulated gene 1 (NDRG1) functions as a suppressor of prostate cancer metastasis. Elucidating pathways that drive survival and invasiveness of NDRG1-deficient prostate cancer cells can help in designing therapeutics to target metastatic prostate cancer cells. However, the molecular mechanisms that lead NDRG1-deficient prostate cancer cells to increased invasiveness remain largely unknown. In this study, we demonstrate that NDRG1-deficient prostate tumors have decreased integrin expression and reduced cell adhesion and motility. Our data indicate that loss of NDRG1 differentially affects Rho GTPases. Specifically, there is a downregulation of active RhoA and Rac1 GTPases with a concomitant upregulation of active Cdc42 in NDRG1-deficient cells. Live cell imaging using a fluorescent sensor that binds to polymerized actin revealed that NDRG1-deficient cells have restricted actin dynamics, thereby affecting cell migration. These cellular and molecular characteristics are in sharp contrast to what is expected after loss of a metastasis suppressor. We further demonstrate that NDRG1-deficient cells have increased resistance to anoikis and increased invasiveness which is independent of its elevated Cdc42 activity. Furthermore, NDRG1 regulates expression and glycosylation of EMMPRIN, a master regulator of matrix metalloproteases. NDRG1 deficiency leads to an increase in EMMPRIN expression with a concomitant increase in matrix metalloproteases and thus invadopodial activity. Using a three-dimensional invasion assay and an in vivo metastasis assay for human prostate xenografts, we demonstrate that NDRG1-deficient prostate cancer cells exhibit a collective invasion phenotype and are highly invasive. Thus, our findings provide novel insights suggesting that loss of NDRG1 leads to a decrease in actin-mediated cellular motility but an increase in cellular invasion, resulting in increased tumor dissemination which positively impacts metastatic outcome.
Collapse
Affiliation(s)
- Anup Sharma
- Prostate Cancer ProgramDepartment of OncologyThe Sidney Kimmel Comprehensive Cancer CenterJohns Hopkins Medical InstitutionsBaltimoreMDUSA
| | - Janet Mendonca
- Prostate Cancer ProgramDepartment of OncologyThe Sidney Kimmel Comprehensive Cancer CenterJohns Hopkins Medical InstitutionsBaltimoreMDUSA
| | - James Ying
- Prostate Cancer ProgramDepartment of OncologyThe Sidney Kimmel Comprehensive Cancer CenterJohns Hopkins Medical InstitutionsBaltimoreMDUSA
| | - Hea‐Soo Kim
- Prostate Cancer ProgramDepartment of OncologyThe Sidney Kimmel Comprehensive Cancer CenterJohns Hopkins Medical InstitutionsBaltimoreMDUSA
| | - James E. Verdone
- Department of UrologyThe James Buchanan Brady Urological InstituteThe Johns Hopkins UniversityBaltimoreMDUSA
| | - Jelani C. Zarif
- Department of UrologyThe James Buchanan Brady Urological InstituteThe Johns Hopkins UniversityBaltimoreMDUSA
| | - Michael Carducci
- Prostate Cancer ProgramDepartment of OncologyThe Sidney Kimmel Comprehensive Cancer CenterJohns Hopkins Medical InstitutionsBaltimoreMDUSA
| | - Hans Hammers
- Prostate Cancer ProgramDepartment of OncologyThe Sidney Kimmel Comprehensive Cancer CenterJohns Hopkins Medical InstitutionsBaltimoreMDUSA
| | - Kenneth J. Pienta
- Department of UrologyThe James Buchanan Brady Urological InstituteThe Johns Hopkins UniversityBaltimoreMDUSA
| | - Sushant Kachhap
- Prostate Cancer ProgramDepartment of OncologyThe Sidney Kimmel Comprehensive Cancer CenterJohns Hopkins Medical InstitutionsBaltimoreMDUSA
| |
Collapse
|
41
|
Oh PS, Kim HS, Kim EM, Hwang H, Ryu HH, Lim S, Sohn MH, Jeong HJ. Inhibitory effect of blue light emitting diode on migration and invasion of cancer cells. J Cell Physiol 2017; 232:3444-3453. [PMID: 28098340 DOI: 10.1002/jcp.25805] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 01/14/2017] [Accepted: 01/17/2017] [Indexed: 12/20/2022]
Abstract
The aim of this study was to determine the effects and molecular mechanism of blue light emitting diode (LED) in tumor cells. A migration and invasion assay for the metastatic behavior of mouse colon cancer CT-26 and human fibrosarcoma HT-1080 cells was performed. Cancer cell migration-related proteins were identified by obtaining a 2-dimensional gel electrophoresis (2-DE) in total cellular protein profile of blue LED-irradiated cancer cells, followed by matrix-assisted laser desorption/ionization-time of flight (MALDI-TOF) analysis of proteins. Protein levels were examined by immunoblotting. Irradiation with blue LED inhibited CT-26 and HT-1080 cell migration and invasion. The anti-metastatic effects of blue LED irradiation were associated with inhibition of matrix metalloproteinase (MMP)-2 and MMP-9 expression. P38 MAPK phosphorylation was increased in blue LED-irradiated CT-26 and HT-1080 cells, but was inhibited after pretreatment with SB203580, a specific inhibitor of p38 MAPK. Inhibition of p38 MAPK phosphorylation by SB203580 treatment increased number of migratory cancer cells in CT-26 and HT-1080 cells, indicating that blue LED irradiation inhibited cancer cell migration via phosphorylation of p38 MAPK. Additionally blue LED irradiation of mice injected with CT-26 cells expressing luciferase decreased early stage lung metastasis compared to untreated control mice. These results indicate that blue LED irradiation inhibits cancer cell migration and invasion in vitro and in vivo.
Collapse
Affiliation(s)
- Phil-Sun Oh
- Molecular Imaging and Therapeutic Medicine Research Center, Cyclotron Research Center, Institute for Medical Sciences, Biomedical Research Institute, Department of Nuclear Medicine, Chonbuk National University Medical School and Hospital, Jeonju, Republic of Korea
| | - Hyun-Soo Kim
- Molecular Imaging and Therapeutic Medicine Research Center, Cyclotron Research Center, Institute for Medical Sciences, Biomedical Research Institute, Department of Nuclear Medicine, Chonbuk National University Medical School and Hospital, Jeonju, Republic of Korea
| | - Eun-Mi Kim
- Molecular Imaging and Therapeutic Medicine Research Center, Cyclotron Research Center, Institute for Medical Sciences, Biomedical Research Institute, Department of Nuclear Medicine, Chonbuk National University Medical School and Hospital, Jeonju, Republic of Korea
| | - Hyosook Hwang
- Molecular Imaging and Therapeutic Medicine Research Center, Cyclotron Research Center, Institute for Medical Sciences, Biomedical Research Institute, Department of Nuclear Medicine, Chonbuk National University Medical School and Hospital, Jeonju, Republic of Korea
| | - Hyang Hwa Ryu
- Molecular Imaging and Therapeutic Medicine Research Center, Cyclotron Research Center, Institute for Medical Sciences, Biomedical Research Institute, Department of Nuclear Medicine, Chonbuk National University Medical School and Hospital, Jeonju, Republic of Korea
| | - SeokTae Lim
- Molecular Imaging and Therapeutic Medicine Research Center, Cyclotron Research Center, Institute for Medical Sciences, Biomedical Research Institute, Department of Nuclear Medicine, Chonbuk National University Medical School and Hospital, Jeonju, Republic of Korea
| | - Myung-Hee Sohn
- Molecular Imaging and Therapeutic Medicine Research Center, Cyclotron Research Center, Institute for Medical Sciences, Biomedical Research Institute, Department of Nuclear Medicine, Chonbuk National University Medical School and Hospital, Jeonju, Republic of Korea
| | - Hwan-Jeong Jeong
- Molecular Imaging and Therapeutic Medicine Research Center, Cyclotron Research Center, Institute for Medical Sciences, Biomedical Research Institute, Department of Nuclear Medicine, Chonbuk National University Medical School and Hospital, Jeonju, Republic of Korea
| |
Collapse
|
42
|
uPAR promotes tumor-like biologic behaviors of fibroblast-like synoviocytes through PI3K/Akt signaling pathway in patients with rheumatoid arthritis. Cell Mol Immunol 2017; 15:171-181. [PMID: 28090093 DOI: 10.1038/cmi.2016.60] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Revised: 09/29/2016] [Accepted: 09/29/2016] [Indexed: 12/31/2022] Open
Abstract
Urokinase-type plasminogen activator receptor (uPAR), is a multifunctional receptor on cell surface, widely present in endothelial cells, fibroblasts, and a variety of malignant cells. Current studies have suggested that uPAR overexpressed on synovial tissues or in synovial fluid or plasma in patients with rheumatoid arthritis (RA). However, there are limited researches regarding the role of uPAR on fibroblast-like synoviocytes of rheumatoid arthritis (RA-FLSs) and its underlying mechanisms. Here, our studies show that the expression of uPAR protein was significantly higher in fibroblast-like synoviocytes (FLSs) from RA than those from osteoarthritis or traumatic injury patients. uPAR gene silencing significantly inhibited RA-FLSs cell proliferation, restrained cell transformation from the G0/G1 phase to S phase, aggravated cell apoptosis, interfered with RA-FLSs cell migration and invasion, and reduced activation of the PI3K/Akt signaling pathway, which may be associated with β1-integrin. Cell supernatants from uPAR gene-silenced RA-FLSs markedly inhibited the migration and tubule formation ability of the HUVECs (a human endothelial cell line). Therefore, we demonstrate that uPAR changes the biological characteristics of RA-FLSs, and affects neoangiogenesis of synovial tissues in patients with RA. All of these may be associated with the β1-integrin/PI3K/Akt signaling pathway. These results imply that targeting uPAR and its downstream signal pathway may provide therapeutic effects in RA.
Collapse
|
43
|
Lee T, Son HY, Choi Y, Shin Y, Oh S, Kim J, Huh YM, Haam S. Minimum hyaluronic acid (HA) modified magnetic nanocrystals with less facilitated cancer migration and drug resistance for targeting CD44 abundant cancer cells by MR imaging. J Mater Chem B 2017; 5:1400-1407. [DOI: 10.1039/c6tb02306a] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
We report minimal amount of hyaluronic acid (HA) conjugated magnetic nanocrystals (mHMs) for targeted imaging of CD44 abundant breast cancer cells with less side effects via MRI.
Collapse
Affiliation(s)
- Taeksu Lee
- Department of Chemical and Biomolecular Engineering
- College of Engineering
- Yonsei University
- Seoul 120-749
- Republic of Korea
| | - Hye Young Son
- Department of Radiology
- College of Medicine
- Yonsei University
- Seoul 120-752
- Republic of Korea
| | - Yuna Choi
- Department of Radiology
- College of Medicine
- Yonsei University
- Seoul 120-752
- Republic of Korea
| | - Youngmin Shin
- Department of Radiology
- College of Medicine
- Yonsei University
- Seoul 120-752
- Republic of Korea
| | - Seungjae Oh
- YUHS-KRIBB Medical Convergence Research Institute
- Yonsei University
- Seoul 120-752
- Republic of Korea
| | - Jinyoung Kim
- Department of Chemical and Biomolecular Engineering
- College of Engineering
- Yonsei University
- Seoul 120-749
- Republic of Korea
| | - Yong-Min Huh
- Department of Radiology
- College of Medicine
- Yonsei University
- Seoul 120-752
- Republic of Korea
| | - Seungjoo Haam
- Department of Chemical and Biomolecular Engineering
- College of Engineering
- Yonsei University
- Seoul 120-749
- Republic of Korea
| |
Collapse
|
44
|
Zhang Y, Duan J, Cai L, Ma D, Xue W. Supramolecular Aggregate as a High-Efficiency Gene Carrier Mediated with Optimized Assembly Structure. ACS APPLIED MATERIALS & INTERFACES 2016; 8:29343-29355. [PMID: 27739303 DOI: 10.1021/acsami.6b11390] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
For cancer gene therapy, a safe and high-efficient gene carrier is a must. To resolve the contradiction between gene transfection efficiency and cytotoxicity, many polymers with complex topological structures have been synthesized, although their synthesis processes and structure control are difficult as well as the high molecular weight also bring high cytotoxicity. We proposed an alternative strategy that uses supramolecular inclusion to construct the aggregate from the small molecules for gene delivery, and to further explore the relationship between the topological assembly structure and their ability to deliver gene. Herein, PEI-1.8k-conjugating β-CD through 6-hydroxyl (PEI-6-CD) and 2-hydroxyl (PEI-2-CD) have been synthesized respectively and then assembled with diferrocene (Fc)-ended polyethylene glycol (PEG-Fc). The obtained aggregates were then used to deliver MMP-9 shRNA plasmid for MCF-7 cancer therapy. It was found that the higher gene transfection efficiency can be obtained by selecting PEI-2-CD as the host and tuning the host/guest molar ratios. With the rational modulation of supramolecular architectures, the aggregate played the functions similar to macromolecules which exhibit higher transfection efficiency than PEI-25k, but show much lower cytotoxicity because of the nature of small/low molecules. In vitro and in vivo assays confirmed that the aggregate could deliver MMP-9 shRNA plasmid effectively into MCF-7 cells and then downregulate MMP-9 expression, which induced the significant MCF-7 cell apoptosis, as well inhibit MCF-7 tumor growth with low toxicity. The supramolecular aggregates maybe become a promising carrier for cancer gene therapy and also provided an alternative strategy for designing new gene carriers.
Collapse
Affiliation(s)
- Yi Zhang
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Department of Biomedical Engineering, Jinan University , Guangzhou 510632, China
| | - Junkun Duan
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Department of Biomedical Engineering, Jinan University , Guangzhou 510632, China
| | - Lingguang Cai
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Department of Biomedical Engineering, Jinan University , Guangzhou 510632, China
| | - Dong Ma
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Department of Biomedical Engineering, Jinan University , Guangzhou 510632, China
| | - Wei Xue
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Department of Biomedical Engineering, Jinan University , Guangzhou 510632, China
| |
Collapse
|
45
|
Proteomic analysis of silenced cathepsin B expression suggests non-proteolytic cathepsin B functionality. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:2700-2709. [DOI: 10.1016/j.bbamcr.2016.08.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2016] [Revised: 07/01/2016] [Accepted: 08/01/2016] [Indexed: 12/30/2022]
|
46
|
Shah T, Wildes F, Kakkad S, Artemov D, Bhujwalla ZM. Lymphatic endothelial cells actively regulate prostate cancer cell invasion. NMR IN BIOMEDICINE 2016; 29:904-911. [PMID: 27149683 DOI: 10.1002/nbm.3543] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Revised: 03/23/2016] [Accepted: 03/29/2016] [Indexed: 06/05/2023]
Abstract
Lymphatic vessels serve as the primary route for metastatic spread to lymph nodes. However, it is not clear how interactions between cancer cells and lymphatic endothelial cells (LECs), especially within hypoxic microenvironments, affect the invasion of cancer cells. Here, using an MR compatible cell perfusion assay, we investigated the role of LEC-prostate cancer (PCa) cell interaction in the invasion and degradation of the extracellular matrix (ECM) by two human PCa cell lines, PC-3 and DU-145, under normoxia and hypoxia, and determined the metabolic changes that occurred under these conditions. We observed a significant increase in the invasion of ECM by invasive PC-3 cells, but not poorly invasive DU-145 cells when human dermal lymphatic microvascular endothelial cells (HMVEC-dlys) were present. Enhanced degradation of ECM by PC-3 cells in the presence of HMVEC-dlys identified interactions between HMVEC-dlys and PCa cells influencing cancer cell invasion. The enhanced ECM degradation was partly attributed to increased MMP-9 enzymatic activity in PC-3 cells when HMVEC-dlys were in close proximity. Significantly higher uPAR and MMP-9 expression levels observed in PC-3 cells compared to DU-145 cells may be one mechanism for increased invasion and degradation of matrigel by these cells irrespective of the presence of HMVEC-dlys. Hypoxia significantly decreased invasion by PC-3 cells, but this decrease was significantly attenuated when HMVEC-dlys were present. Significantly higher phosphocholine was observed in invasive PC-3 cells, while higher glycerophosphocholine was observed in DU-145 cells. These metabolites were not altered in the presence of HMVEC-dlys. Significantly increased lipid levels and lipid droplets were observed in PC-3 and DU-145 cells under hypoxia reflecting an adaptive survival response to oxidative stress. These results suggest that in vivo, invasive cells in or near lymphatic endothelial cells are likely to be more invasive and degrade the ECM to influence the metastatic cascade. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Tariq Shah
- JHU ICMIC Program, Division of Cancer Imaging Research, Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Flonne Wildes
- JHU ICMIC Program, Division of Cancer Imaging Research, Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Samata Kakkad
- JHU ICMIC Program, Division of Cancer Imaging Research, Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Dmitri Artemov
- JHU ICMIC Program, Division of Cancer Imaging Research, Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Zaver M Bhujwalla
- JHU ICMIC Program, Division of Cancer Imaging Research, Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
47
|
Dyshlovoy SA, Menchinskaya ES, Venz S, Rast S, Amann K, Hauschild J, Otte K, Kalinin VI, Silchenko AS, Avilov SA, Alsdorf W, Madanchi R, Bokemeyer C, Schumacher U, Walther R, Aminin DL, Fedorov SN, Shubina LK, Stonik VA, Balabanov S, Honecker F, von Amsberg G. The marine triterpene glycoside frondoside A exhibits activity in vitro and in vivo in prostate cancer. Int J Cancer 2016; 138:2450-65. [PMID: 26695519 DOI: 10.1002/ijc.29977] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 12/09/2015] [Indexed: 12/17/2022]
Abstract
Despite recent advances in the treatment of metastatic castration-resistant prostate cancer (CRPC), outcome of patients remains poor due to the development of drug resistance. Thus, new drugs are urgently needed. We investigated efficacy, toxicity and mechanism of action of marine triterpene glycoside frondoside A (FrA) using CRPC cell lines in vitro and in vivo. FrA revealed high efficacy in human prostate cancer cells, while non-malignant cells were less sensitive. Remarkably, proliferation and colony formation of cells resistant to enzalutamide and abiraterone (due to the androgen receptor splice variant AR-V7) were also significantly inhibited by FrA. The marine compound caused cell type specific cell cycle arrest and induction of caspase-dependent or -independent apoptosis. Up-regulation or induction of several pro-apoptotic proteins (Bax, Bad, PTEN), cleavage of PARP and caspase-3 and down-regulation of anti-apoptotic proteins (survivin and Bcl-2) were detected in treated cells. Global proteome analysis revealed regulation of proteins involved in formation of metastases, tumor cell invasion, and apoptosis, like keratin 81, CrkII, IL-1β and cathepsin B. Inhibition of pro-survival autophagy was observed following FrA exposure. In vivo, FrA inhibited tumor growth of PC-3 and DU145 cells with a notable reduction of lung metastasis, as well as circulating tumor cells in the peripheral blood. Increased lymphocyte counts of treated animals might indicate an immune modulating effect of FrA. In conclusion, our results suggest that FrA is a promising new drug for the treatment of mCRPC. Induction of apoptosis, inhibition of pro-survival autophagy, and immune modulatory effects are suspected modes of actions.
Collapse
Affiliation(s)
- Sergey A Dyshlovoy
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald-Tumorzentrum, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Laboratory of Marine Natural Products Chemistry, G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far-East Branch, Russian Academy of Sciences, Vladivostok, Russian Federation.,Laboratory of bioactive compounds, Department of bioorganic chemistry and biotechnology, School of Natural Sciences, Far Eastern Federal University, Vladivostok, Russian Federation
| | - Ekaterina S Menchinskaya
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald-Tumorzentrum, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Laboratory of Marine Natural Products Chemistry, G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far-East Branch, Russian Academy of Sciences, Vladivostok, Russian Federation
| | - Simone Venz
- Department of Medical Biochemistry and Molecular Biology, University of Greifswald, Greifswald, Germany.,Department of Functional Genomics, Interfacultary Institute of Genetics and Functional Genomics, University of Greifswald, Greifswald, Germany
| | - Stefanie Rast
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald-Tumorzentrum, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Kerstin Amann
- Nephropathology Department, University Medical Center Erlangen, Erlangen, Germany
| | - Jessica Hauschild
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald-Tumorzentrum, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Katharina Otte
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald-Tumorzentrum, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Vladimir I Kalinin
- Laboratory of Marine Natural Products Chemistry, G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far-East Branch, Russian Academy of Sciences, Vladivostok, Russian Federation
| | - Alexandra S Silchenko
- Laboratory of Marine Natural Products Chemistry, G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far-East Branch, Russian Academy of Sciences, Vladivostok, Russian Federation
| | - Sergey A Avilov
- Laboratory of Marine Natural Products Chemistry, G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far-East Branch, Russian Academy of Sciences, Vladivostok, Russian Federation
| | - Winfried Alsdorf
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald-Tumorzentrum, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ramin Madanchi
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald-Tumorzentrum, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Carsten Bokemeyer
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald-Tumorzentrum, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Udo Schumacher
- Institute of Anatomy and Experimental Morphology, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Reinhard Walther
- Department of Medical Biochemistry and Molecular Biology, University of Greifswald, Greifswald, Germany
| | - Dmitry L Aminin
- Laboratory of Marine Natural Products Chemistry, G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far-East Branch, Russian Academy of Sciences, Vladivostok, Russian Federation
| | - Sergey N Fedorov
- Laboratory of Marine Natural Products Chemistry, G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far-East Branch, Russian Academy of Sciences, Vladivostok, Russian Federation
| | - Larisa K Shubina
- Laboratory of Marine Natural Products Chemistry, G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far-East Branch, Russian Academy of Sciences, Vladivostok, Russian Federation
| | - Valentin A Stonik
- Laboratory of Marine Natural Products Chemistry, G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far-East Branch, Russian Academy of Sciences, Vladivostok, Russian Federation
| | - Stefan Balabanov
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald-Tumorzentrum, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Division of Hematology, University Hospital Zurich, Zurich, Switzerland
| | - Friedemann Honecker
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald-Tumorzentrum, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Tumor and Breast Center ZeTuP St. Gallen, St. Gallen, Switzerland
| | - Gunhild von Amsberg
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald-Tumorzentrum, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
48
|
Xian J, Shao H, Chen X, Zhang S, Quan J, Zou Q, Jin H, Zhang L. Nucleophosmin Mutants Promote Adhesion, Migration and Invasion of Human Leukemia THP-1 Cells through MMPs Up-regulation via Ras/ERK MAPK Signaling. Int J Biol Sci 2016; 12:144-55. [PMID: 26884713 PMCID: PMC4737672 DOI: 10.7150/ijbs.13382] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 11/26/2015] [Indexed: 01/07/2023] Open
Abstract
Acute myeloid leukemia (AML) with mutated nucleophosmin (NPM1) has been defined as a unique subgroup in the new classification of myeloid neoplasm, and the AML patients with mutated NPM1 frequently present extramedullary infiltration, but how NPM1 mutants regulate this process remains elusive. In this study, we found that overexpression of type A NPM1 gene mutation (NPM1-mA) enhanced the adhesive, migratory and invasive potential in THP-1 AML cells lacking mutated NPM1. NPM1-mA had up-regulated expression and gelatinolytic matrix metalloprotease-2 (MMP-2)/MMP-9 activity, as assessed by real-time PCR, western blotting and gelatin zymography. Following immunoprecipitation analysis to identify the interaction of NPM1-mA with K-Ras, we focused on the effect of NPM1-mA overexpression on the Ras/Mitogen-activated protein kinase (MAPK) signaling axis and showed that NPM1-mA increased the MEK and ERK phosphorylation levels, as evaluated by western blotting. Notably, a specific inhibitor of the ERK/MAPK pathway (PD98059), but not p38/MAPK, JNK/MAPK or PI3-K/AKT inhibitors, markedly decreased the cell invasion numbers in a transwell assay. Further experiments demonstrated that blocking the ERK/MAPK pathway by PD98059 resulted in reduced MMP-2/9 protein levels and MMP-9 activity. Additionally, NPM1-mA overexpression had down-regulated gene expression and protein production of tissue inhibitor of MMP-2 (TIMP-2) in THP-1 cells. Furthermore, evaluation of gene expression data from The Cancer Genome Atlas (TCGA) dataset revealed that MMP-2 was overexpressed in AML patient samples with NPM1 mutated and high MMP-2 expression associated with leukemic skin infiltration. Taken together, our results reveal that NPM1 mutations contribute to the invasive potential of AML cells through MMPs up-regulation via Ras/ERK MAPK signaling pathway activation and offer novel insights into the potential role of NPM1 mutations in leukemogenesis.
Collapse
Affiliation(s)
- Jingrong Xian
- 1. Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of Education, College of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Huiyuan Shao
- 2. Department of clinical laboratory, Yantai Yuhuangding Hospital, Shandong, China
| | - Xianchun Chen
- 3. Department of clinical laboratory, People's hospital of Ganzhou, Jiangxi, China
| | - Shuaishuai Zhang
- 1. Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of Education, College of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Jing Quan
- 1. Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of Education, College of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Qin Zou
- 1. Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of Education, College of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Hongjun Jin
- 1. Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of Education, College of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Ling Zhang
- 1. Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of Education, College of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| |
Collapse
|
49
|
Cantres-Rosario YM, Hernandez N, Negron K, Perez-Laspiur J, Leszyk J, Shaffer SA, Meléndez LM. Interacting partners of macrophage-secreted cathepsin B contribute to HIV-induced neuronal apoptosis. AIDS 2015. [PMID: 26208400 DOI: 10.1097/qad.0000000000000823] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
OBJECTIVE HIV-1 infection of macrophages increases cathepsin B secretion and induces neuronal apoptosis, but the molecular mechanism remains unclear. DESIGN We identified macrophage-secreted cathepsin B protein interactions extracellularly and their contribution to neuronal death in vitro. METHODS Cathepsin B was immunoprecipitated from monocyte-derived macrophage supernatants after 12 days postinfection. The cathepsin B interactome was identified by label-free tandem mass spectrometry and compared with uninfected supernatants. Proteins identified were validated by western blot. Neurons were exposed to macrophage-conditioned media in presence or absence of antibodies against cathepsin B and interacting proteins. Apoptosis was measured using TUNEL labeling. Immunohistochemistry of postmortem brain tissue samples from healthy, HIV-infected and Alzheimer's disease patients was performed to observe the ex-vivo expression of the proteins identified. RESULTS Nine proteins co-immunoprecipitated differentially with cathepsin B between uninfected and HIV-infected macrophages. Serum amyloid P component (SAPC)-cathepsin B interaction increased in HIV-infected macrophage supernatants, while matrix metalloprotease 9 (MMP-9)-cathepsin B interaction decreased. Pre-treatment of HIV-infected macrophage-conditioned media with antibodies against cathepsin B and SAPC decreased neuronal apoptosis. The addition of MMP-9 antibodies was not neuro-protective SAPC was overexpressed in postmortem brain tissue from HIV-positive neurocognitive impaired patients compared with HIV positive with normal cognition and healthy controls, although MMP-9 expression was similar in all tissues. CONCLUSION Inhibiting SAPC-cathepsin B interaction protects against HIV-induced neuronal death and may help to find alternative treatments for HIV-associated neurocognitive disorders.
Collapse
|
50
|
Patra P, Mitra S, Das Gupta A, Pradhan S, Bhattacharya S, Ahir M, Mukherjee S, Sarkar S, Roy S, Chattopadhyay S, Adhikary A, Goswami A, Chattopadhyay D. Simple synthesis of biocompatible biotinylated porous hexagonal ZnO nanodisc for targeted doxorubicin delivery against breast cancer cell: In vitro and in vivo cytotoxic potential. Colloids Surf B Biointerfaces 2015; 133:88-98. [PMID: 26093304 DOI: 10.1016/j.colsurfb.2015.05.052] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Revised: 05/26/2015] [Accepted: 05/31/2015] [Indexed: 01/15/2023]
Abstract
Targeted drug delivery with porous materials features great promise as improved therapeutic potential for treatment of various diseases. In the present study we have attempted a microwave synthesis of porous hexagonal nanodisc of zinc oxide (PZHD) for the first time and its subsequent targeted delivery to breast cancer cells, MCF7. PZHD has been fabricated suitably with 3-aminopropyltriethoxysilane to impart additional stability and surface amines to anchor site directing ligand NHS-biotin. Biotinylated scaffold showed targeted delivery of anticancer drug doxorubicin and pH triggered release to MCF 7 cells with preferential distribution on specified domain. A detailed in vitro cytotoxicity study was associated with it to evaluate the mode of action of Dox loaded PZHD on MCF-7 cells by means of cell cycle analysis, apoptosis assays, Western blot and immuno-fluorescence image analysis. The efficacy of the Dox loaded PZHD was further validated from our in vivo tumor regression studies. Finally, the whole study has been supported by in vitro and in vivo bio-safety studies which also signified its biocompatibility with real time applications. To the best of our knowledge this is the first effort to use biotinylated PZHD for targeted delivery of doxorubicin within MCF 7 cells with a detailed study of its mechanistic application. This study might thus hold future prospects for therapeutic intervention for treatment of cancer.
Collapse
Affiliation(s)
- Prasun Patra
- Centre for Research in NanoScience and NanoTechnology, Technology Campus, University of Calcutta, JD 2, Sec III, Salt lake, Kolkata 700098, India.
| | - Shouvik Mitra
- Biological Sciences Division, Indian Statistical Institute, 203 B.T. Road, Kolkata 700108, India
| | - Amarto Das Gupta
- Department of Biotechnology, Birla Institute of Technology and Science, Vidya Vihar, Pilani, Rajasthan 333031, India
| | - Saheli Pradhan
- Biological Sciences Division, Indian Statistical Institute, 203 B.T. Road, Kolkata 700108, India
| | - Saurav Bhattacharya
- Centre for Research in NanoScience and NanoTechnology, Technology Campus, University of Calcutta, JD 2, Sec III, Salt lake, Kolkata 700098, India
| | - Manisha Ahir
- Centre for Research in NanoScience and NanoTechnology, Technology Campus, University of Calcutta, JD 2, Sec III, Salt lake, Kolkata 700098, India
| | - Sudeshna Mukherjee
- Department of Physiology, University College of Science and Technology, University of Calcutta, 92 A.P.C. Road, Kolkata 700009, India
| | - Sampad Sarkar
- Biological Sciences Division, Indian Statistical Institute, 203 B.T. Road, Kolkata 700108, India
| | - Subhrodeb Roy
- Biological Sciences Division, Indian Statistical Institute, 203 B.T. Road, Kolkata 700108, India
| | - Sreya Chattopadhyay
- Department of Physiology, University College of Science and Technology, University of Calcutta, 92 A.P.C. Road, Kolkata 700009, India
| | - Arghya Adhikary
- Centre for Research in NanoScience and NanoTechnology, Technology Campus, University of Calcutta, JD 2, Sec III, Salt lake, Kolkata 700098, India
| | - Arunava Goswami
- Biological Sciences Division, Indian Statistical Institute, 203 B.T. Road, Kolkata 700108, India
| | - Dhrubajyoti Chattopadhyay
- Centre for Research in NanoScience and NanoTechnology, Technology Campus, University of Calcutta, JD 2, Sec III, Salt lake, Kolkata 700098, India
| |
Collapse
|