1
|
Lasota M, Jankowski D, Wiśniewska A, Sarna M, Kaczor-Kamińska M, Misterka A, Szczepaniak M, Dulińska-Litewka J, Górecki A. The Potential of Congo Red Supplied Aggregates of Multitargeted Tyrosine Kinase Inhibitor (Sorafenib, BAY-43-9006) in Enhancing Therapeutic Impact on Bladder Cancer. Int J Mol Sci 2023; 25:269. [PMID: 38203437 PMCID: PMC10779242 DOI: 10.3390/ijms25010269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 12/15/2023] [Accepted: 12/21/2023] [Indexed: 01/12/2024] Open
Abstract
Bladder cancer is a common malignancy associated with high recurrence rates and potential progression to invasive forms. Sorafenib, a multi-targeted tyrosine kinase inhibitor, has shown promise in anti-cancer therapy, but its cytotoxicity to normal cells and aggregation in solution limits its clinical application. To address these challenges, we investigated the formation of supramolecular aggregates of sorafenib with Congo red (CR), a bis-azo dye known for its supramolecular interaction. We analyzed different mole ratios of CR-sorafenib aggregates and evaluated their effects on bladder cancer cells of varying levels of malignancy. In addition, we also evaluated the effect of the test compounds on normal uroepithelial cells. Our results demonstrated that sorafenib inhibits the proliferation of bladder cancer cells and induces apoptosis in a dose-dependent manner. However, high concentrations of sorafenib also showed cytotoxicity to normal uroepithelial cells. In contrast, the CR-BAY aggregates exhibited reduced cytotoxicity to normal cells while maintaining anti-cancer activity. The aggregates inhibited cancer cell migration and invasion, suggesting their potential for metastasis prevention. Dynamic light scattering and UV-VIS measurements confirmed the formation of stable co-aggregates with distinctive spectral properties. These CR-sorafenib aggregates may provide a promising approach to targeted therapy with reduced cytotoxicity and improved stability for drug delivery in bladder cancer treatment. This work shows that the drug-excipient aggregates proposed and described so far, as Congo red-sorafenib, can be a real step forward in anti-cancer therapies.
Collapse
Affiliation(s)
- Małgorzata Lasota
- Chair of Medical Biochemistry, Jagiellonian University Medical College, Kopernika 7, 31-034 Krakow, Poland; (M.K.-K.); (A.M.); (J.D.-L.)
- SSG of Targeted Therapy and Supramolecular Systems, Jagiellonian University Medical College, Kopernika 7, 31-034 Krakow, Poland; (D.J.); (M.S.)
| | - Daniel Jankowski
- SSG of Targeted Therapy and Supramolecular Systems, Jagiellonian University Medical College, Kopernika 7, 31-034 Krakow, Poland; (D.J.); (M.S.)
- Department of Physical Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland;
| | - Anna Wiśniewska
- Chair of Pharmacology, Faculty of Medicine, Jagiellonian University Medical College, Grzegórzecka 16, 31-531 Krakow, Poland;
| | - Michał Sarna
- Department of Biophysics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland;
| | - Marta Kaczor-Kamińska
- Chair of Medical Biochemistry, Jagiellonian University Medical College, Kopernika 7, 31-034 Krakow, Poland; (M.K.-K.); (A.M.); (J.D.-L.)
| | - Anna Misterka
- Chair of Medical Biochemistry, Jagiellonian University Medical College, Kopernika 7, 31-034 Krakow, Poland; (M.K.-K.); (A.M.); (J.D.-L.)
- SSG of Targeted Therapy and Supramolecular Systems, Jagiellonian University Medical College, Kopernika 7, 31-034 Krakow, Poland; (D.J.); (M.S.)
| | - Mateusz Szczepaniak
- SSG of Targeted Therapy and Supramolecular Systems, Jagiellonian University Medical College, Kopernika 7, 31-034 Krakow, Poland; (D.J.); (M.S.)
- Department of Biophysics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland;
| | - Joanna Dulińska-Litewka
- Chair of Medical Biochemistry, Jagiellonian University Medical College, Kopernika 7, 31-034 Krakow, Poland; (M.K.-K.); (A.M.); (J.D.-L.)
| | - Andrzej Górecki
- Department of Physical Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland;
| |
Collapse
|
2
|
Trybus W, Król T, Trybus E. Rhein induces changes in the lysosomal compartment of HeLa cells. J Cell Biochem 2022; 123:1506-1524. [PMID: 35901236 DOI: 10.1002/jcb.30311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 06/17/2022] [Accepted: 07/15/2022] [Indexed: 11/10/2022]
Abstract
Rhein is an anthraquinone found in Rheum palmatum, used in Chinese medicine. Due to potential anticancer properties, the study assessed its effect on the lysosomal compartment, which indirectly influences cell death. The experiment was performed on HeLa cells by treating them with rhein at concentrations of 100-300 µM. LC3-II protein and caspase 3/7 activity, level of apoptosis, the concentration of reactive oxide species (ROS), and mitochondrial potential (Δψm) were evaluated by the cytometric method. To evaluate the permeability of the lysosomal membrane (LMP), staining with acridine orange and the assessment of activity of cathepsin D and L in the lysosomal and extralysosomal fractions were used. Cell viability was assessed by -(3-(4,5-Dimethylthiazol-2-yl)-2,5-Diphenyltetrazolium Bromide) (MTT) and neutral red (NR) assays. Changes in cells were also demonstrated at the level of electron, optical, confocal, and fluorescence microscopy. Inhibition of autophagy was done using chloroquine. Rhein-induced degradation processes were confirmed by an increase in the number of primary lysosomes, autophagosomes, and autolysosomes. At high concentrations, rhein caused the generation of ROS, which induced LMP expressed by quenching of acridine orange fluorescence. These results correlated with a reduction of lysosomes, as visualized in graphical modeling, with the decreased uptake of NR by lysosomes, and increased activity of cathepsin D and L in the extralysosomal fraction. The studies also showed an increase in the activity of caspase 3/7 and a decrease in the expression of Bcl-2 protein, indicative of rhein-stimulated apoptosis. At the same time, we demonstrated that preincubation of cells with chloroquine inhibited rhein-induced autophagy and contributed to increased cytotoxicity to HeLa cells. Rhein also induced DNA damage and led to cycle arrest in the S phase. Our results indicate that rhein, by inducing changes in the lysosomal compartment, indirectly affects apoptosis of HeLa cells and in combination with autophagy inhibitors may be an effective form of anticancer therapy.
Collapse
Affiliation(s)
- Wojciech Trybus
- Department of Medical Biology, Institute of Biology, The Jan Kochanowski University, Kielce, Poland
| | - Teodora Król
- Department of Medical Biology, Institute of Biology, The Jan Kochanowski University, Kielce, Poland
| | - Ewa Trybus
- Department of Medical Biology, Institute of Biology, The Jan Kochanowski University, Kielce, Poland
| |
Collapse
|
3
|
Tsai JP, Lee CC, Huang PY, Hsieh YH, Chen YS. Melatonin combined with sorafenib synergistically inhibit the invasive ability through targeting metastasis-associated protein 2 expression in human renal cancer cells. Tzu Chi Med J 2022; 34:192-199. [PMID: 35465276 PMCID: PMC9020234 DOI: 10.4103/tcmj.tcmj_204_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 08/09/2021] [Accepted: 08/19/2021] [Indexed: 11/04/2022] Open
Abstract
Objectives: Materials and Methods: Results: Conclusion:
Collapse
|
4
|
Abstract
The intimate involvement of pathogens with the heightened risk for developing certain cancers is an area of research that has captured a great deal of attention over the last 10 years. One firmly established paradigm that highlights this aspect of disease progression is in the instance of Helicobacter pylori infection and the contribution it makes in elevating the risk for developing gastric cancer. Whilst the molecular mechanisms that pinpoint the contribution that this microorganism inflicts towards host cells during gastric cancer initiation have come into greater focus, another picture that has also emerged is one that implicates the host's immune system, and the chronic inflammation that can arise therefrom, as being a central contributory factor in disease progression. Consequently, when taken with the underlying role that the extracellular matrix plays in the development of most cancers, and how this dynamic can be modulated by proteases expressed from the tumor or inflammatory cells, a complex and detailed relationship shared between the individual cellular components and their surroundings is coming into focus. In this review article, we draw attention to the emerging role played by the cathepsin proteases in modulating the stage-specific progression of Helicobacter pylori-initiated gastric cancer and the underlying immune response, while highlighting the therapeutic significance of this dynamic and how it may be amenable for novel intervention strategies within a basic research or clinical setting.
Collapse
|
5
|
Riaz S, Abdulrahman N, Uddin S, Jabeen A, Gadeau AP, Fliegel L, Mraiche F. Anti-hypertrophic effect of Na +/H + exchanger-1 inhibition is mediated by reduced cathepsin B. Eur J Pharmacol 2020; 888:173420. [PMID: 32781168 DOI: 10.1016/j.ejphar.2020.173420] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 07/24/2020] [Accepted: 07/24/2020] [Indexed: 12/20/2022]
Abstract
Previous studies have established the role of Na+/H+ exchanger isoform-1 (NHE1) and cathepsin B (Cat B) in the development of cardiomyocyte hypertrophy (CH). Both NHE1 and Cat B are activated under acidic conditions suggesting that their activities might be interrelated. The inhibition of NHE1 has been demonstrated to reduce cardiac hypertrophy but the mechanism that contributes to the anti-hypertrophic effect of NHE1 inhibition still remains unclear. H9c2 cardiomyoblasts were stimulated with Angiotensin (Ang) II in the presence and absence of N-[2-methyl-4,5-bis(methylsulphonyl)-benzoyl]-guanidine, hydrochloride (EMD, EMD 87580), an NHE1 inhibitor or CA-074Me, a Cat B inhibitor, and various cardiac hypertrophic parameters, namely cell surface area, protein content and atrial natriuretic peptide (ANP) mRNA were analyzed. EMD significantly suppressed markers of cardiomyocyte hypertrophy and inhibited Ang II stimulated Cat B protein and gene expression. Cat B is located within the acidic environment of lysosomes. Cat B proteases are released into the cytoplasm upon disintegration of the lysosomes. EMD or CA-074Me prevented the dispersal of the lysosomes induced by Ang II and reduced the ratio of LC3-II to LC3-I, a marker of autophagy. Moreover, Cat B protein expression and MMP-9 activity in the extracellular space were significantly attenuated in the presence of EMD or CA-074Me. Our study demonstrates a novel mechanism for attenuation of the hypertrophic phenotype by NHE1 inhibition that is mediated by a regression in Cat B. The inhibition of Cat B via EMD or CA-074Me attenuates the autosomal-lysosomal pathway and MMP-9 activation.
Collapse
Affiliation(s)
- Sadaf Riaz
- College of Pharmacy, QU Health, Qatar University, Doha, Qatar; Hamad Medical Corporation, Doha, Qatar
| | - Nabeel Abdulrahman
- College of Pharmacy, QU Health, Qatar University, Doha, Qatar; Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Shahab Uddin
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Ayesha Jabeen
- College of Pharmacy, QU Health, Qatar University, Doha, Qatar
| | | | | | - Fatima Mraiche
- College of Pharmacy, QU Health, Qatar University, Doha, Qatar.
| |
Collapse
|
6
|
Ashrafizadeh M, Zarrabi A, Samarghandian S, Najafi M. PTEN: What we know of the function and regulation of this onco-suppressor factor in bladder cancer? Eur J Pharmacol 2020; 881:173226. [PMID: 32485246 DOI: 10.1016/j.ejphar.2020.173226] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 05/14/2020] [Accepted: 05/26/2020] [Indexed: 12/13/2022]
Abstract
Bladder cancer accounts for high morbidity and mortality around the world and its incidence rate is suggested to be higher in following years. A number of factors involve in bladder cancer development such as lifestyle and drugs. However, it appears that genetic factors play a significant role in bladder cancer development and progression. Phosphatase and tensin homolog (PTEN) is a cancer-related transcription factor that is corelated with reduced proliferation and invasion of cancer cells by negatively targeting PI3K/Akt/mTOR signaling pathway. In the present review, we aimed to explore the role of PTEN in bladder cancer cells and how upstream modulators affect PTEN in this life-threatening disorder. Down-regulation of PTEN is associated with poor prognosis, chemoresistance and progression of cancer cells. Besides, microRNAs, long non-coding RNAs, circular RNAs and other molecular pathways such as NF-kB are able to target PTEN in bladder cancer cells. Notably, anti-tumor drugs such as kaempferol, β-elemene and sorafenib upregulate the expression of PTEN to exert their inhibitory effects on bladder cancer cells.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Department of Basic Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Ali Zarrabi
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, 34956, Istanbul, Turkey
| | - Saeed Samarghandian
- Healthy Ageing Research Center, Neyshabur University of Medical Sciences, Neyshabur, Iran
| | - Masoud Najafi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
7
|
Haghi A, Salami M, Mohammadi Kian M, Nikbakht M, Mohammadi S, Chahardouli B, Rostami SH, Malekzadeh K. Effects of Sorafenib and Arsenic Trioxide on U937 and KG-1 Cell Lines: Apoptosis or Autophagy? CELL JOURNAL 2019; 22:253-262. [PMID: 31863650 PMCID: PMC6947003 DOI: 10.22074/cellj.2020.6728] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Accepted: 04/23/2019] [Indexed: 12/21/2022]
Abstract
Objective Acute myeloid leukemia (AML) is a clonal disorder of hemopoietic progenitor cells. The Raf serine/threonine (Ser/Thr) protein kinase isoforms including B-Raf and RAF1, are the upstream in the MAPK cascade that play essential functions in regulating cellular proliferation and survival. Activated autophagy-related genes have a dual role in both cell death and cell survival in cancer cells. The cytotoxic activities of arsenic trioxide (ATO) were widely assessed in many cancers. Sorafenib is known as a multikinase inhibitor which acts through suppression of Ser/Thr kinase Raf that was reported to have a key role in tumor cell signaling, proliferation, and angiogenesis. In this study, we examined the combination effect of ATO and sorafenib in AML cell lines. Materials and Methods In this experimental study, we studied in vitro effects of ATO and sorafenib on human leukemia cell lines. The effective concentrations of compounds were determined by MTT assay in both single and combination treatments. Apoptosis was evaluated by annexin-V FITC staining. Finally, mRNA levels of apoptotic and autophagy genes were evaluated using real-time polymerase chain reaction (PCR). Results Data demonstrated that sorafenib, ATO, and their combination significantly increase the number of apoptotic cells. We found that the combination of ATO and sorafenib significantly reduces the viability of U937 and KG-1 cells. The expression level of selective autophagy genes, ULK1 and Beclin1 decreased but LC3-II increased in U937. Conclusion The expression levels of apoptotic and autophagy activator genes were increased in response to treatment. The crosstalk between apoptosis and autophagy is a complicated mechanism and further investigations seem to be necessary.
Collapse
Affiliation(s)
- Atousa Haghi
- Hematology, Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran.,Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mahdieh Salami
- Hematology, Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran.,Hematologic Malignancies Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahnaz Mohammadi Kian
- Hematology, Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran.,Hematologic Malignancies Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohsen Nikbakht
- Hematology, Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran.,Hematologic Malignancies Research Center, Tehran University of Medical Sciences, Tehran, Iran. Electronic Address:
| | - Saeed Mohammadi
- Hematology, Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran.,Hematologic Malignancies Research Center, Tehran University of Medical Sciences, Tehran, Iran. Electronic Address:
| | - Bahram Chahardouli
- Hematology, Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran.,Hematologic Malignancies Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - S Haharbano Rostami
- Hematology, Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran.,Hematologic Malignancies Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Kianoosh Malekzadeh
- Molecular Medicine Research Center (MMRC), Hormozgan University of Medical Science (HUMS), Bandar Abbass, Iran
| |
Collapse
|
8
|
Typical and Atypical Inducers of Lysosomal Cell Death: A Promising Anticancer Strategy. Int J Mol Sci 2018; 19:ijms19082256. [PMID: 30071644 PMCID: PMC6121368 DOI: 10.3390/ijms19082256] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 07/30/2018] [Accepted: 07/30/2018] [Indexed: 12/22/2022] Open
Abstract
Lysosomes are conservative organelles with an indispensable role in cellular degradation and the recycling of macromolecules. However, in light of recent findings, it has emerged that the role of lysosomes in cancer cells extends far beyond cellular catabolism and includes a variety of cellular pathways, such as proliferation, metastatic potential, and drug resistance. It has been well described that malignant transformation leads to alterations in lysosomal structure and function, which, paradoxically, renders cancer cells more sensitive to lysosomal destabilization. Furthermore, lysosomes are implicated in the regulation and execution of cell death in response to diverse stimuli and it has been shown that lysosome-dependent cell death can be utilized to overcome apoptosis and drug resistance. Thus, the purpose of this review is to characterize the role of lysosome in cancer therapy and to describe how these organelles impact treatment resistance. We summarized the characteristics of typical inducers of lysosomal cell death, which exert its function primarily via alterations in the lysosomal compartment. The review also presents other anticancer agents with the predominant mechanism of action different from lysosomal destabilization, the activity of which is influenced by lysosomal signaling, including classical chemotherapeutics, kinase inhibitors, monoclonal antibodies, as well as photodynamic therapy.
Collapse
|
9
|
Yuan D, Liu C, Hu B. Dysfunction of Membrane Trafficking Leads to Ischemia-Reperfusion Injury After Transient Cerebral Ischemia. Transl Stroke Res 2018; 9:215-222. [PMID: 29022237 PMCID: PMC5895539 DOI: 10.1007/s12975-017-0572-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 09/19/2017] [Accepted: 09/21/2017] [Indexed: 01/12/2023]
Abstract
Neurons require an extraordinarily high level of membrane trafficking activities because of enriched axonal terminals and dendritic branches. For that reason, defects in the membrane trafficking pathway are a hallmark of most, and may be all, neurodegenerative disorders. A major cellular membrane trafficking pathway is the Golgi apparatus (Golgi hereafter)-late endosome-lysosome axis for supplying lysosomal enzymes. This pathway is regulated by N-ethylmaleimide-sensitive factor (NSF) ATPase. This review article is to discuss a novel hypothesis that brain ischemia inactivates NSF ATPase, resulting in a cascade of events of disruption of the Golgi-endosome-lysosome pathway, release of cathepsin B (CTSB), and induction of mitochondrial outer membrane permeabilization (MOMP) during the postischemic phase. This hypothesis is supported by recent studies demonstrating that NSF is trapped into inactive protein aggregates in neurons destined to die after brain ischemia. Consequently, Golgi, transport vesicles (TVs), and late endosomes (LEs) are accumulated and damaged, which is followed by CTSB release from these damaged structures. Moderate release of CTSB cleaves Bax-like BH3 protein (Bid) to become active truncated Bid (tBid). Active tBid is then translocated to the mitochondrial outer membrane, resulting in oligomerization of BCL2-associated X protein (Bax) forming the mitochondrial outer membrane pores, and releasing mitochondrial intramembranous proteins. Extensive CTSB release, however, can digest cellular proteins indiscriminately to induce cell death. Based on these new observations, we propose a novel hypothesis, i.e., brain ischemia leads to NSF inactivation, resulting in a massive buildup of damaged Golgi, TVs and LEs, fatal release of CTSB, induction of MOMP, and eventually brain ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Dong Yuan
- Department of Anesthesiology and Neurology, Shock Trauma and Anesthesiology Research Center, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Chunli Liu
- Department of Anesthesiology and Neurology, Shock Trauma and Anesthesiology Research Center, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Bingren Hu
- Department of Anesthesiology and Neurology, Shock Trauma and Anesthesiology Research Center, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
- Veterans Affairs Maryland Health Center System, 10 North Greene Street, Baltimore, MD, 21201, USA.
| |
Collapse
|
10
|
Santoni M, Massari F, Piva F, Carrozza F, Di Nunno V, Cimadamore A, Martignetti A, Montironi R, Battelli N. Tivozanib for the treatment of renal cell carcinoma. Expert Opin Pharmacother 2018; 19:1021-1025. [PMID: 29851529 DOI: 10.1080/14656566.2018.1480722] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Abstract
INTRODUCTION Renal cell carcinoma (RCC) represents a heterogeneous group of cancers with distinct histological features, molecular alterations, prognosis, and response to therapy. Target agents directed against vascular endothelial growth factor and its receptor and mammalian target of rapamycin (mTOR) inhibitors have completely changed the landscape of RCC. However, the rate of complete response is still low, thus supporting the research of novel therapeutic agents. Area covered: The authors describe the chemical features of tivozanib, its pharmacodynamic and pharmacokinetic properties, and the results obtained in human phase I-III clinical trials. Tivozanib received its first global approval in EU, Iceland, and Norway on 28 August 2017 for the first-line treatment of adult patients with advanced RCC and for adult patients who are VEGFR and mTOR inhibitor-naive following disease progression after one prior treatment with cytokines. Expert opinion: The US Food and Drug Administration did not approve tivozanib due to the lack of a significant advantage in terms of survival compared to sorafenib. To date, the role of tivozanib in the pharmaceutical landscape of mRCC appears to be very limited. However, ongoing trials on the association between tivozanib and immunotherapy may represent a promising strategy to be assessed in future clinical trials.
Collapse
Affiliation(s)
| | - Francesco Massari
- b Division of Oncology , S. Orsola-Malpighi Hospital , Bologna , Italy
| | - Francesco Piva
- c Department of Specialistic Clinical and Odontostomatological Sciences , Polytechnic University of Marche , Ancona , Italy
| | | | - Vincenzo Di Nunno
- b Division of Oncology , S. Orsola-Malpighi Hospital , Bologna , Italy
| | - Alessia Cimadamore
- e Section of Pathological Anatomy , Polytechnic University of the Marche Region, School of Medicine, United Hospitals , Ancona , Italy
| | - Angelo Martignetti
- f Dipartimento oncologico uslsudest toscana- area senese , Poggibonsi , Italy
| | - Rodolfo Montironi
- e Section of Pathological Anatomy , Polytechnic University of the Marche Region, School of Medicine, United Hospitals , Ancona , Italy
| | | |
Collapse
|
11
|
Nobiletin (NOB) suppresses autophagic degradation via over-expressing AKT pathway and enhances apoptosis in multidrug-resistant SKOV3/TAX ovarian cancer cells. Biomed Pharmacother 2018; 103:29-37. [PMID: 29635125 DOI: 10.1016/j.biopha.2018.03.126] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 03/19/2018] [Accepted: 03/20/2018] [Indexed: 12/21/2022] Open
Abstract
Chemotherapy could be used as an effective therapeutic treatment for ovarian cancer and subsequent peritoneal metastasis. However, the occurrence of drug resistance reduced the treatment effect originated from cancer chemotherapy. Accumulating evidences indicated the significant role of autophagy in tumor cell resistance to chemotherapy. Thus, inhibition of autophagy using natural compounds could be a promising candidate to overcome multidrug resistance in human ovarian cancer cells. Nobiletin (NOB), a polymethoxyflavonoid found in citrus fruits such as Citrus depressa and Citrus reticulate, exhibits a number of bioactivities. In the present study, NOB selectively suppressed the growth and proliferation of human SKOV3/TAX cells, inducing G0/G1 phase arrest and reducing G2/M phase, along with the increase of p53 and p21. In addition, NOB induced significant apoptosis in SKOV3/TAX cells through the intrinsic apoptosis pathway, as evidenced by the up-regulation of cleaved Caspase-9/-3 and PARP. Further, NOB impaired the autophagic degradation in SKOV3/TAX cells, resulting in autophagic flux inhibition. Moreover, the impaired autophagic flux enhanced NOB-induced apoptosis in SKOV3/TAX cells. Importantly, AKT signaling was activated by NOB, which was involved in autophagic degradation and apoptotic cell death. In conclusion, the findings here supplied the illustration that NOB could overcome multidrug resistance in human ovarian cancer cells through AKT-regulated suppression of autophagic degradation.
Collapse
|
12
|
Nakano M, Tanaka M, Kuromatsu R, Nagamatsu H, Satani M, Niizeki T, Okamura S, Iwamoto H, Shimose S, Shirono T, Noda Y, Koga H, Torimura T. Alternative treatments in advanced hepatocellular carcinoma patients with progressive disease after sorafenib treatment: a prospective multicenter cohort study. Oncotarget 2018; 7:64400-64409. [PMID: 27462865 PMCID: PMC5325452 DOI: 10.18632/oncotarget.10794] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Accepted: 06/30/2016] [Indexed: 02/07/2023] Open
Abstract
Sorafenib is an oral multikinase inhibitor that has been approved to treat advanced hepatocellular carcinoma (HCC), though it is unclear how much benefit advanced HCC patients with progressive disease (PD) derive from sorafenib treatment. This study aimed to assess survival risk factors and evaluate therapeutic strategies for advanced HCC patients with PD after sorafenib treatment. We analyzed the clinical data and treatment outcomes for 315 consecutive advanced HCC patients treated with sorafenib. Univariate analyses of overall survival identified therapeutic effect as an independent risk factor in all patients. Among all patients, 141 developed PD. Of those, 58 (41%) were treated with sorafenib monotherapy, 70 (50%) with agents other than sorafenib, and 13 (9%) were not treated at all. The median survival time was 6.1 months for PD patients with sorafenib monotherapy and 12.2 months for those administered alternative treatments (p < 0.0001). Our results indicated that sorafenib treatment may have negative long-term therapeutic effects in advanced HCC patients with PD, and that alternative treatments should be considered for these patients after sorafenib administration.
Collapse
Affiliation(s)
- Masahito Nakano
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | | | - Ryoko Kuromatsu
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | | | - Manabu Satani
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Takashi Niizeki
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Shusuke Okamura
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Hideki Iwamoto
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Shigeo Shimose
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Tomotake Shirono
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Yu Noda
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Hironori Koga
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Takuji Torimura
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | | |
Collapse
|
13
|
Chen H, Lv M, Lv Z, Li C, Zhang W, Zhao X, Duan X, Jin C, Xiong J, Xu F, Li Y. Divergent roles of three cytochrome c in CTSB-modulating coelomocyte apoptosis in Apostichopus japonicus. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2017; 76:65-76. [PMID: 28549733 DOI: 10.1016/j.dci.2017.05.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 05/20/2017] [Accepted: 05/21/2017] [Indexed: 06/07/2023]
Abstract
Cytochrome c plays crucial roles in apoptosis and the immune response. We previously demonstrated that cathepsin B from Apostichopus japonicus (AjCTSB) induces coelomocyte apoptosis. However, the mechanistic explanation and the regulation of this process have not been investigated. In the present study, we identified three cytochrome c cDNAs from A. japonicus (designated Ajcytc1, Ajcytc-1, and Ajcytc-2) using expressed sequence tag- (EST) and RACE-based approaches. The deduced amino acid sequences of the three cytochrome isoforms contained conserved CXXCH motifs, which are involved in binding heme and maintaining proteolytic activity. Time course expression analysis in vitro and in vivo revealed that the three cytochrome isoforms were induced upon pathogen challenge and LPS exposure. More importantly, AjCTSB knockdown by siRNA dramatically increased mitochondrial membrane potential (ΔΨm) in a time-dependent manner based on JC-1 fluorescent probe staining. AjCTSB knockdown also resulted in decreased expression of these three cytochromes 24 h after siAjCTSB transfection. Functional analysis using isoform-specific siRNAs revealed that Ajcytc-1, but not Ajcytc1 or Ajcytc-2, is involved in coelomocyte apoptosis. Moreover, the transcript level of Ajcaspase-3, an apoptosis executioner, was also consistently down-regulated upon silencing of Ajcytc-1 but not Ajcytc1 or Ajcytc-2. Collectively, these results indicate that Ajcytc1, Ajcytc-1, and Ajcytc-2 play distinct roles in mediating the immune response to bacteria according to AjCTSB expression. Moreover, Ajcytc-1 could be released upon dissipation of the ΔΨm, which could further trigger coelomocyte apoptosis through the activation of Ajcaspase-3.
Collapse
Affiliation(s)
- Huahui Chen
- School of Marine Sciences, Ningbo University, PR China
| | - Miao Lv
- School of Marine Sciences, Ningbo University, PR China
| | - Zhimeng Lv
- School of Marine Sciences, Ningbo University, PR China
| | - Chenghua Li
- School of Marine Sciences, Ningbo University, PR China.
| | - Weiwei Zhang
- School of Marine Sciences, Ningbo University, PR China
| | - Xuelin Zhao
- School of Marine Sciences, Ningbo University, PR China
| | - Xuemei Duan
- School of Marine Sciences, Ningbo University, PR China
| | - Chunhua Jin
- School of Marine Sciences, Ningbo University, PR China
| | - Jinbo Xiong
- School of Marine Sciences, Ningbo University, PR China
| | - Feng Xu
- School of Marine Sciences, Ningbo University, PR China
| | - Ye Li
- School of Marine Sciences, Ningbo University, PR China
| |
Collapse
|
14
|
Sun H, Huang M, Yao N, Hu J, Li Y, Chen L, Hu N, Ye W, Chi-Shing Tai W, Zhang D, Chen S. The cycloartane triterpenoid ADCX impairs autophagic degradation through Akt overactivation and promotes apoptotic cell death in multidrug-resistant HepG2/ADM cells. Biochem Pharmacol 2017; 146:87-100. [PMID: 29074104 DOI: 10.1016/j.bcp.2017.10.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2017] [Accepted: 10/20/2017] [Indexed: 12/26/2022]
Abstract
Multidrug resistance is the main obstacle in cancer chemotherapy. Emerging evidence demonstrates the important role of autophagy in cancer cell resistance to chemotherapy. Therefore, autophagy inhibition by natural compounds may be a promising strategy for overcoming drug resistance in liver cancer cells. Here, we found that ADCX, a natural cycloartane triterpenoid extracted from the traditional Chinese medicine (TCM) source Cimicifugae rhizoma (Shengma), impaired autophagic degradation by suppressing lysosomal cathepsin B (CTSB) expression in multidrug-resistant liver cancer HepG2/ADM cells, thereby leading to autophagic flux inhibition. Moreover, impairing autophagic flux promoted ADCX-induced apoptotic cell death in HepG2/ADM cells. Interestingly, Akt was overactivated by ADCX treatment, which downregulated CTSB and inhibited autophagic flux. Together, our results provide the first demonstration that an active TCM constituent can overcome multidrug resistance in liver cancer cells via Akt-mediated inhibition of autophagic degradation.
Collapse
Affiliation(s)
- Haiyan Sun
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; State Key Laboratory of Chinese Medicine and Molecular Pharmacology, Hong Kong Polytechnic University, Hong Kong, China
| | - Maohua Huang
- College of Pharmacy, Jinan University, Guangzhou, China; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Guangzhou, China
| | - Nan Yao
- College of Pharmacy, Jinan University, Guangzhou, China; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Guangzhou, China
| | - Jianyang Hu
- College of Pharmacy, Jinan University, Guangzhou, China; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Guangzhou, China
| | - Yingjie Li
- College of Pharmacy, Jinan University, Guangzhou, China; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Guangzhou, China
| | - Liping Chen
- College of Pharmacy, Jinan University, Guangzhou, China; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Guangzhou, China
| | - Nan Hu
- College of Pharmacy, Jinan University, Guangzhou, China; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Guangzhou, China
| | - Wencai Ye
- College of Pharmacy, Jinan University, Guangzhou, China; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Guangzhou, China
| | - William Chi-Shing Tai
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology, Hong Kong Polytechnic University, Hong Kong, China
| | - Dongmei Zhang
- College of Pharmacy, Jinan University, Guangzhou, China; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Guangzhou, China.
| | - Sibao Chen
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; State Key Laboratory of Chinese Medicine and Molecular Pharmacology, Hong Kong Polytechnic University, Hong Kong, China.
| |
Collapse
|
15
|
Martens S, Jeong M, Tonnus W, Feldmann F, Hofmans S, Goossens V, Takahashi N, Bräsen JH, Lee EW, Van der Veken P, Joossens J, Augustyns K, Fulda S, Linkermann A, Song J, Vandenabeele P. Sorafenib tosylate inhibits directly necrosome complex formation and protects in mouse models of inflammation and tissue injury. Cell Death Dis 2017; 8:e2904. [PMID: 28661484 PMCID: PMC5520944 DOI: 10.1038/cddis.2017.298] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 05/23/2017] [Accepted: 05/25/2017] [Indexed: 12/17/2022]
Abstract
Necroptosis contributes to the pathophysiology of several inflammatory, infectious and degenerative disorders. TNF-induced necroptosis involves activation of the receptor-interacting protein kinases 1 and 3 (RIPK1/3) in a necrosome complex, eventually leading to the phosphorylation and relocation of mixed lineage kinase domain like protein (MLKL). Using a high-content screening of small compounds and FDA-approved drug libraries, we identified the anti-cancer drug Sorafenib tosylate as a potent inhibitor of TNF-dependent necroptosis. Interestingly, Sorafenib has a dual activity spectrum depending on its concentration. In murine and human cell lines it induces cell death, while at lower concentrations it inhibits necroptosis, without affecting NF-κB activation. Pull down experiments with biotinylated Sorafenib show that it binds independently RIPK1, RIPK3 and MLKL. Moreover, it inhibits RIPK1 and RIPK3 kinase activity. In vivo Sorafenib protects against TNF-induced systemic inflammatory response syndrome (SIRS) and renal ischemia–reperfusion injury (IRI). Altogether, we show that Sorafenib can, next to the reported Braf/Mek/Erk and VEGFR pathways, also target the necroptotic pathway and that it can protect in an acute inflammatory RIPK1/3-mediated pathology.
Collapse
Affiliation(s)
- Sofie Martens
- VIB-UGent Center for Inflammation Research (IRC), Ghent, Belgium.,Department of Biomedical Molecular Biology (DBMB), Ghent University, Ghent, Belgium
| | - Manhyung Jeong
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | - Wulf Tonnus
- Department of Internal Medicine III, Division of Nephrology, University Hospital Carl Gustav Carus at Technische Universität Dresden, Dresden, Germany
| | - Friederike Feldmann
- Institute for Experimental Cancer Research in Pediatrics, Goethe-University, Frankfurt, Germany
| | - Sam Hofmans
- Laboratory of Medicinal Chemistry, University of Antwerp, Antwerp, Belgium
| | - Vera Goossens
- VIB-UGent Center for Inflammation Research (IRC), Ghent, Belgium.,Department of Biomedical Molecular Biology (DBMB), Ghent University, Ghent, Belgium
| | - Nozomi Takahashi
- VIB-UGent Center for Inflammation Research (IRC), Ghent, Belgium.,Department of Biomedical Molecular Biology (DBMB), Ghent University, Ghent, Belgium
| | | | - Eun-Woo Lee
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, South Korea
| | | | - Jurgen Joossens
- Laboratory of Medicinal Chemistry, University of Antwerp, Antwerp, Belgium
| | - Koen Augustyns
- Laboratory of Medicinal Chemistry, University of Antwerp, Antwerp, Belgium
| | - Simone Fulda
- Institute for Experimental Cancer Research in Pediatrics, Goethe-University, Frankfurt, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany.,German Cancer Consortium (DKTK), Partner Site Frankfurt, Germany
| | - Andreas Linkermann
- Department of Internal Medicine III, Division of Nephrology, University Hospital Carl Gustav Carus at Technische Universität Dresden, Dresden, Germany
| | - Jaewhan Song
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | - Peter Vandenabeele
- VIB-UGent Center for Inflammation Research (IRC), Ghent, Belgium.,Department of Biomedical Molecular Biology (DBMB), Ghent University, Ghent, Belgium
| |
Collapse
|
16
|
Llewellyn KJ, Nalbandian A, Weiss LN, Chang I, Yu H, Khatib B, Tan B, Scarfone V, Kimonis VE. Myogenic differentiation of VCP disease-induced pluripotent stem cells: A novel platform for drug discovery. PLoS One 2017; 12:e0176919. [PMID: 28575052 PMCID: PMC5456028 DOI: 10.1371/journal.pone.0176919] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2016] [Accepted: 04/19/2017] [Indexed: 02/07/2023] Open
Abstract
Valosin Containing Protein (VCP) disease is an autosomal dominant multisystem proteinopathy caused by mutations in the VCP gene, and is primarily associated with progressive muscle weakness, including atrophy of the pelvic and shoulder girdle muscles. Currently, no treatments are available and cardiac and respiratory failures can lead to mortality at an early age. VCP is an AAA ATPase multifunction complex protein and mutations in the VCP gene resulting in disrupted autophagic clearance. Due to the rarity of the disease, the myopathic nature of the disorder, ethical and practical considerations, VCP disease muscle biopsies are difficult to obtain. Thus, disease-specific human induced pluripotent stem cells (hiPSCs) now provide a valuable resource for the research owing to their renewable and pluripotent nature. In the present study, we report the differentiation and characterization of a VCP disease-specific hiPSCs into precursors expressing myogenic markers including desmin, myogenic factor 5 (MYF5), myosin and heavy chain 2 (MYH2). VCP disease phenotype is characterized by high expression of TAR DNA Binding Protein-43 (TDP-43), ubiquitin (Ub), Light Chain 3-I/II protein (LC3-I/II), and p62/SQSTM1 (p62) protein indicating disruption of the autophagy cascade. Treatment of hiPSC precursors with autophagy stimulators Rapamycin, Perifosine, or AT101 showed reduction in VCP pathology markers TDP-43, LC3-I/II and p62/SQSTM1. Conversely, autophagy inhibitors chloroquine had no beneficial effect, and Spautin-1 or MHY1485 had modest effects. Our results illustrate that hiPSC technology provide a useful platform for a rapid drug discovery and hence constitutes a bridge between clinical and bench research in VCP and related diseases.
Collapse
Affiliation(s)
- Katrina J. Llewellyn
- Division of Genetics and Genomic Medicine, Department of Pediatrics, University of California-Irvine School of Medicine, Irvine, California, United States of America
- Sue and Bill Gross Stem Cell Research Center, University of California-Irvine School of Medicine, Irvine, California, United States of America
| | - Angèle Nalbandian
- Division of Genetics and Genomic Medicine, Department of Pediatrics, University of California-Irvine School of Medicine, Irvine, California, United States of America
- Sue and Bill Gross Stem Cell Research Center, University of California-Irvine School of Medicine, Irvine, California, United States of America
| | - Lan N. Weiss
- Division of Genetics and Genomic Medicine, Department of Pediatrics, University of California-Irvine School of Medicine, Irvine, California, United States of America
- Sue and Bill Gross Stem Cell Research Center, University of California-Irvine School of Medicine, Irvine, California, United States of America
| | - Isabela Chang
- Division of Genetics and Genomic Medicine, Department of Pediatrics, University of California-Irvine School of Medicine, Irvine, California, United States of America
| | - Howard Yu
- Division of Genetics and Genomic Medicine, Department of Pediatrics, University of California-Irvine School of Medicine, Irvine, California, United States of America
| | - Bibo Khatib
- Division of Genetics and Genomic Medicine, Department of Pediatrics, University of California-Irvine School of Medicine, Irvine, California, United States of America
- Sue and Bill Gross Stem Cell Research Center, University of California-Irvine School of Medicine, Irvine, California, United States of America
| | - Baichang Tan
- Division of Genetics and Genomic Medicine, Department of Pediatrics, University of California-Irvine School of Medicine, Irvine, California, United States of America
| | - Vanessa Scarfone
- Sue and Bill Gross Stem Cell Research Center, University of California-Irvine School of Medicine, Irvine, California, United States of America
| | - Virginia E. Kimonis
- Division of Genetics and Genomic Medicine, Department of Pediatrics, University of California-Irvine School of Medicine, Irvine, California, United States of America
- Sue and Bill Gross Stem Cell Research Center, University of California-Irvine School of Medicine, Irvine, California, United States of America
| |
Collapse
|
17
|
Noguchi S, Shibutani S, Fukushima K, Mori T, Igase M, Mizuno T. Bosutinib, an SRC inhibitor, induces caspase-independent cell death associated with permeabilization of lysosomal membranes in melanoma cells. Vet Comp Oncol 2017; 16:69-76. [DOI: 10.1111/vco.12312] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 02/15/2017] [Accepted: 03/07/2017] [Indexed: 12/14/2022]
Affiliation(s)
- S. Noguchi
- Laboratory of Veterinary Radiology, Graduate School of Life and Environmental Sciences; Osaka Prefecture University; Osaka Japan
- Biomedical Science Center for Translational Research, The United Graduate School of Veterinary Science; Yamaguchi University; Yamaguchi Japan
| | - S. Shibutani
- Laboratory of Veterinary Hygiene, Joint Faculty of Veterinary Medicine; Yamaguchi University; Yamaguchi Japan
| | - K. Fukushima
- Laboratory of Molecular Diagnostics and Therapeutics, Joint Faculty of Veterinary Medicine; Yamaguchi University; Yamaguchi Japan
| | - T. Mori
- Laboratory of Veterinary Clinical Oncology, Faculty of Applied Biological Sciences; Gifu University; Gifu Japan
| | - M. Igase
- The United Graduate School of Veterinary Science; Yamaguchi University; Yamaguchi Japan
| | - T. Mizuno
- Biomedical Science Center for Translational Research, The United Graduate School of Veterinary Science; Yamaguchi University; Yamaguchi Japan
- Laboratory of Molecular Diagnostics and Therapeutics, Joint Faculty of Veterinary Medicine; Yamaguchi University; Yamaguchi Japan
| |
Collapse
|
18
|
Chen H, Lv M, Lv Z, Li C, Xu W, Zhang W, Zhao X, Duan X, Jin C. Molecular cloning and functional characterization of cathepsin B from the sea cucumber Apostichopus japonicus. FISH & SHELLFISH IMMUNOLOGY 2017; 60:447-457. [PMID: 27847342 DOI: 10.1016/j.fsi.2016.11.033] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 11/08/2016] [Accepted: 11/12/2016] [Indexed: 06/06/2023]
Abstract
Cathepsin B (CTSB), a member of lysosomal cysteine protease, is involved in multiple levels of physiological and biological processes, and also plays crucial roles in host immune defense against pathogen infection in vertebrates. However, the function of CTSB within the innate immune system of invertebrates, particularly in marine echinoderms, has been poorly documented. In this study, the immune function of CTSB in Apostichopus japonicus (designated as AjCTSB), a commercially important and disease vulnerable aquaculture specie, was investigated by integrated molecular and protein approaches. A 2153 bp cDNA representing the full-length of AjCTSB was cloned via overlapping ESTs and RACE fragments. AjCTSB contained an open reading frame of 999 bp encoding a secreted protein of 332 amino acid residues with a predicted molecular mass of 36.8 kDa. The deduced amino acid of AjCTSB shared a typical activity center containing three conserved amino acid residues (Cys108, His277 and Asn297). Phylogenetic tree analysis also supported that AjCTSB was a new member of CTSB family with clustering firstly with invertebrate CTSBs. Quantitative real time PCR analysis revealed that AjCTSB was ubiquitously expressed in all examined tissues with the highest levels in intestine. The Vibrio splendidus challenged sea cucumber and LPS-exposed coelomocytes could both significantly boost the expression of AjCTSB. Moreover, the purified recombinant AjCTSB exhibited dose-dependent CTSB activities at the concentration ranged from 0 to 0.24 μg μL-1. Further functional analysis indicated that coelomocytes apoptosis was significantly inhibited by 0.16-fold in vivo and the apoptosis execution Ajcaspase 3 was extremely reduced in Apostichopus japonicus coelomocytes treated with specific AjCTSB siRNA. Collectively, all these results suggested that AjCTSB was an important immune factor and might be served as apoptosis enhancers in pathogen challenged sea cucumber.
Collapse
Affiliation(s)
- Huahui Chen
- School of Marine Sciences, Ningbo University, PR China
| | - Miao Lv
- School of Marine Sciences, Ningbo University, PR China
| | - Zhimeng Lv
- School of Marine Sciences, Ningbo University, PR China
| | - Chenghua Li
- School of Marine Sciences, Ningbo University, PR China.
| | - Wei Xu
- Louisiana State University, Agricultural Center, USA
| | - Weiwei Zhang
- School of Marine Sciences, Ningbo University, PR China
| | - Xuelin Zhao
- School of Marine Sciences, Ningbo University, PR China
| | - Xuemei Duan
- School of Marine Sciences, Ningbo University, PR China
| | - Chunhua Jin
- School of Marine Sciences, Ningbo University, PR China
| |
Collapse
|
19
|
A simple method to induce hypoxia-induced vascular endothelial growth factor-A (VEGF-A) expression in T24 human bladder cancer cells. In Vitro Cell Dev Biol Anim 2016; 53:272-276. [PMID: 27752923 DOI: 10.1007/s11626-016-0103-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 09/27/2016] [Indexed: 10/20/2022]
Abstract
Angiogenesis is an essential process for the establishment, development, and dissemination of several malignant tumors including bladder cancer. The hypoxic condition promotes the stabilization of hypoxia-inducible factor 1 alpha (HIF-1α), which translocates to the nucleus to mediate angiogenic factors including the vascular endothelial growth factor A (VEGF-A). AnaeroGen system was developed for microbiology area to create a low oxygen tension required to the growth of anaerobic bacteria. Here, we hypothesized the use of AnaeroGen system to induce hypoxia in T24 human bladder carcinoma cells, in order to promote the overexpression of VEGF-A. T24 cells were cultured in six-well plates containing McCoy medium. Exposures of T24 cells to hypoxia for 1, 8, 24, and 48 h were performed using the Oxoid AnaeroGen system, while T24 cells under normoxia were used as control. The expression of VEGF-A and HIF-1α was analyzed by real-time PCR. ELISA for HIF-1α was carried out. The VEGF-A expression increased significantly by Oxoid AnaeroGen-induced hypoxia in a time-depending manner, reaching the peak in 48 h of hypoxia. Although HIF-1α mRNA was not changed, HIF-1α protein was increased in the presence of hypoxia, reaching a peak at 8 h. These results demonstrated that the Oxoid AnaeroGen system is a simple method to expose T24 cells to hypoxia and efficiently to upregulate VEGF expression in T24 cells.
Collapse
|
20
|
Zhang L, Ge C, Zhao F, Zhang Y, Wang X, Yao M, Li J. NRBP2 Overexpression Increases the Chemosensitivity of Hepatocellular Carcinoma Cells via Akt Signaling. Cancer Res 2016; 76:7059-7071. [PMID: 27634758 DOI: 10.1158/0008-5472.can-16-0937] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Revised: 07/24/2016] [Accepted: 08/08/2016] [Indexed: 12/22/2022]
Abstract
Hepatocellular carcinoma is highly resistant to chemotherapy. Research data supported that cancer stem cells (CSC) may be responsible for the chemoresistance and strategies that suppress CSCs stemness could also inhibit the drug resistance. In this study, we found that nuclear receptor binding protein 2 (NRBP2) expression was downregulated in the CD133+ hepatocellular carcinoma CSCs. Most adjacent noncancerous liver tissue analyzed expressed higher level of NRBP2 compared with cancerous tissue in hepatocellular carcinoma patients, and high NRBP2 expression indicated a better prognosis. Real-time PCR results showed that NRBP2 negatively correlated with stemness-related genes, including Oct3/4, Nanog, Notch1, Ep300, and CD133 mRNA expression. High NRBP2 expression in hepatocellular carcinoma cells downregulated CK19 protein expression, inhibited tumorsphere formation, and tumorigenesis ability, indicating that high NRBP2 expression restrains the hepatocellular carcinoma cell stemness. Overexpression of NRBP2 reduced the IC50 of sorafenib in hepatocellular carcinoma cells, and NRBP2 expression was negatively correlated with hepatocellular carcinoma cell resistance to the chemotherapy agents, including cisplatin and the Akt signaling inhibitor perifosine. Coimmunoprecipitation results showed that NRBP2 could bind with Annexin A2 (ANXA2) and inhibit ANXA2 expression. Coexpression of ANXA2 restored the chemoresistant ability in NRBP2-overexpressing hepatocellular carcinoma cells. Further analysis showed that NRBP2 downregulated Akt and its downstream signaling target Bad phosphorylation level. ANXA2 coexpression partially restored the Akt phosphorylation. Analysis of the expression of Bcl2 family proteins showed that NRBP2 may increase hepatocellular carcinoma cell chemosensitivity by regulating expression of survival proteins involved in the Akt and Bcl2 pathway. These results suggest that NRBP2 plays an important role in the tumor progression and chemotherapeutic resistance of hepatocellular carcinoma. Cancer Res; 76(23); 7059-71. ©2016 AACR.
Collapse
Affiliation(s)
- Lixing Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Chao Ge
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Fangyu Zhao
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yang Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xin Wang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Ming Yao
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jinjun Li
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.
| |
Collapse
|
21
|
Lin MT, Lin CL, Lin TY, Cheng CW, Yang SF, Lin CL, Wu CC, Hsieh YH, Tsai JP. Synergistic effect of fisetin combined with sorafenib in human cervical cancer HeLa cells through activation of death receptor-5 mediated caspase-8/caspase-3 and the mitochondria-dependent apoptotic pathway. Tumour Biol 2015; 37:6987-96. [PMID: 26662956 DOI: 10.1007/s13277-015-4526-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 11/26/2015] [Indexed: 12/21/2022] Open
Abstract
Combining antitumor agents with bioactive compounds is a potential strategy for improving the effect of chemotherapy on cancer cells. The goal of this study was to elucidate the antitumor effect of the flavonoid, fisetin, combined with the multikinase inhibitor, sorafenib, against human cervical cancer cells in vitro and in vivo. The combination of fisetin and sorafenib synergistically induced apoptosis in HeLa cells, which is accompanied by a marked increase in loss of mitochondrial membrane potential. Apoptosis induction was achieved by caspase-3 and caspase-8 activation which increased the ratio of Bax/Bcl-2 and caused the subsequent cleavage of PARP level while disrupting the mitochondrial membrane potential in HeLa cells. Decreased Bax/Bcl-2 ratio level and mitochondrial membrane potential were also observed in siDR5-treated HeLa cells. In addition, in vivo studies revealed that the combined fisetin and sorafenib treatment was clearly superior to sorafenib treatment alone using a HeLa xenograft model. Our study showed that the combination of fisetin and sorafenib exerted better synergistic effects in vitro and in vivo than either agent used alone against human cervical cancer, and this synergism was based on apoptotic potential through a mitochondrial- and DR5-dependent caspase-8/caspase-3 signaling pathway. This combined fisetin and sorafenib treatment represents a novel therapeutic strategy for further clinical developments in advanced cervical cancer.
Collapse
Affiliation(s)
- Ming-Te Lin
- Department of Obstetrics and Gynecology, Chang Bing Show Chwan Memorial Hospital, Lugang Town, Changhua County, Taiwan.,Liberal Arts Center, Da-Yeh University, Changhua, Taiwan
| | - Chia-Liang Lin
- Institute of Biochemistry, Microbiology and Immunology, Chung Shan Medical University, Taichung, Taiwan
| | - Tzu-Yu Lin
- Department of Biochemistry and Molecular Biology, College of Biological Sciences, University of California Davis, Davis, CA, USA
| | - Chun-Wen Cheng
- Institute of Biochemistry, Microbiology and Immunology, Chung Shan Medical University, Taichung, Taiwan
| | - Shun-Fa Yang
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Chu-Liang Lin
- Institute of Biochemistry, Microbiology and Immunology, Chung Shan Medical University, Taichung, Taiwan
| | - Chih-Chien Wu
- Institute of Biochemistry, Microbiology and Immunology, Chung Shan Medical University, Taichung, Taiwan
| | - Yi-Hsien Hsieh
- Institute of Biochemistry, Microbiology and Immunology, Chung Shan Medical University, Taichung, Taiwan. .,Department of Biochemistry, School of Medicine, Chung Shan Medical University, Taichung, Taiwan. .,Clinical Laboratory, Chung Shan Medical University Hospital, Taichung, Taiwan.
| | - Jen-Pi Tsai
- School of Medicine, Tzu Chi University, Hualien, Taiwan. .,Department of Internal Medicine, Division of Nephrology, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi, Taiwan.
| |
Collapse
|