1
|
Puente-Cobacho B, Varela-López A, Quiles JL, Vera-Ramirez L. Involvement of redox signalling in tumour cell dormancy and metastasis. Cancer Metastasis Rev 2023; 42:49-85. [PMID: 36701089 PMCID: PMC10014738 DOI: 10.1007/s10555-022-10077-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Accepted: 12/27/2022] [Indexed: 01/27/2023]
Abstract
Decades of research on oncogene-driven carcinogenesis and gene-expression regulatory networks only started to unveil the complexity of tumour cellular and molecular biology. This knowledge has been successfully implemented in the clinical practice to treat primary tumours. In contrast, much less progress has been made in the development of new therapies against metastasis, which are the main cause of cancer-related deaths. More recently, the role of epigenetic and microenviromental factors has been shown to play a key role in tumour progression. Free radicals are known to communicate the intracellular and extracellular compartments, acting as second messengers and exerting a decisive modulatory effect on tumour cell signalling. Depending on the cellular and molecular context, as well as the intracellular concentration of free radicals and the activation status of the antioxidant system of the cell, the signalling equilibrium can be tilted either towards tumour cell survival and progression or cell death. In this regard, recent advances in tumour cell biology and metastasis indicate that redox signalling is at the base of many cell-intrinsic and microenvironmental mechanisms that control disseminated tumour cell fate and metastasis. In this manuscript, we will review the current knowledge about redox signalling along the different phases of the metastatic cascade, including tumour cell dormancy, making emphasis on metabolism and the establishment of supportive microenvironmental connections, from a redox perspective.
Collapse
Affiliation(s)
- Beatriz Puente-Cobacho
- Department of Genomic Medicine, GENYO, Centre for Genomics and Oncology, Pfizer-University of Granada and Andalusian Regional Government, PTS, Granada, Spain
| | - Alfonso Varela-López
- Department of Physiology, Institute of Nutrition and Food Technology "José Mataix Verdú", Biomedical Research Center, University of Granada, Granada, Spain
| | - José L Quiles
- Department of Physiology, Institute of Nutrition and Food Technology "José Mataix Verdú", Biomedical Research Center, University of Granada, Granada, Spain
| | - Laura Vera-Ramirez
- Department of Genomic Medicine, GENYO, Centre for Genomics and Oncology, Pfizer-University of Granada and Andalusian Regional Government, PTS, Granada, Spain. .,Department of Physiology, Institute of Nutrition and Food Technology "José Mataix Verdú", Biomedical Research Center, University of Granada, Granada, Spain.
| |
Collapse
|
2
|
Morais KLP, Ciccone L, Stura E, Alvarez-Flores MP, Mourier G, Driessche MV, Sciani JM, Iqbal A, Kalil SP, Pereira GJ, Marques-Porto R, Cunegundes P, Juliano L, Servent D, Chudzinski-Tavassi AM. Structural and functional properties of the Kunitz-type and C-terminal domains of Amblyomin-X supporting its antitumor activity. Front Mol Biosci 2023; 10:1072751. [PMID: 36845546 PMCID: PMC9948614 DOI: 10.3389/fmolb.2023.1072751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 01/12/2023] [Indexed: 02/11/2023] Open
Abstract
Amblyomin-X is a Kunitz-type FXa inhibitor identified through the transcriptome analysis of the salivary gland from Amblyomma sculptum tick. This protein consists of two domains of equivalent size, triggers apoptosis in different tumor cell lines, and promotes regression of tumor growth, and reduction of metastasis. To study the structural properties and functional roles of the N-terminal (N-ter) and C-terminal (C-ter) domains of Amblyomin-X, we synthesized them by solid-phase peptide synthesis, solved the X-Ray crystallographic structure of the N-ter domain, confirming its Kunitz-type signature, and studied their biological properties. We show here that the C-ter domain is responsible for the uptake of Amblyomin-X by tumor cells and highlight the ability of this domain to deliver intracellular cargo by the strong enhancement of the intracellular detection of molecules with low cellular-uptake efficiency (p15) after their coupling with the C-ter domain. In contrast, the N-ter Kunitz domain of Amblyomin-X is not capable of crossing through the cell membrane but is associated with tumor cell cytotoxicity when it is microinjected into the cells or fused to TAT cell-penetrating peptide. Additionally, we identify the minimum length C-terminal domain named F2C able to enter in the SK-MEL-28 cells and induces dynein chains gene expression modulation, a molecular motor that plays a role in the uptake and intracellular trafficking of Amblyomin-X.
Collapse
Affiliation(s)
- K. L. P. Morais
- Center of Excellence in New Target Discovery (CENTD), Butantan Institute, São Paulo, Brazil,Laboratory of Development and Innovation, Butantan Institute, São Paulo, Brazil,Department of Biochemistry, Federal University of São Paulo, São Paulo, Brazil
| | - L. Ciccone
- Département Médicaments et Technologies pour la Santé (DMTS), Université Paris-Saclay, CEA SIMoS, Gif-sur-Yvette, France,Department of Pharmacy, University of Pisa, Pisa, Italy
| | - E. Stura
- Département Médicaments et Technologies pour la Santé (DMTS), Université Paris-Saclay, CEA SIMoS, Gif-sur-Yvette, France
| | - M. P. Alvarez-Flores
- Center of Excellence in New Target Discovery (CENTD), Butantan Institute, São Paulo, Brazil
| | - G. Mourier
- Département Médicaments et Technologies pour la Santé (DMTS), Université Paris-Saclay, CEA SIMoS, Gif-sur-Yvette, France
| | - M. Vanden Driessche
- Département Médicaments et Technologies pour la Santé (DMTS), Université Paris-Saclay, CEA SIMoS, Gif-sur-Yvette, France
| | - J. M. Sciani
- Center of Excellence in New Target Discovery (CENTD), Butantan Institute, São Paulo, Brazil
| | - A. Iqbal
- Center of Excellence in New Target Discovery (CENTD), Butantan Institute, São Paulo, Brazil,Laboratory of Development and Innovation, Butantan Institute, São Paulo, Brazil
| | - S. P. Kalil
- Center of Excellence in New Target Discovery (CENTD), Butantan Institute, São Paulo, Brazil
| | - G. J. Pereira
- Department of Pharmacology, Federal University of São Paulo, São Paulo, Brazil
| | - R. Marques-Porto
- Laboratory of Development and Innovation, Butantan Institute, São Paulo, Brazil
| | - P. Cunegundes
- Center of Excellence in New Target Discovery (CENTD), Butantan Institute, São Paulo, Brazil,Department of Biochemistry, Federal University of São Paulo, São Paulo, Brazil
| | - L. Juliano
- Department of Biophysics, Federal University of São Paulo, São Paulo, Brazil
| | - D. Servent
- Département Médicaments et Technologies pour la Santé (DMTS), Université Paris-Saclay, CEA SIMoS, Gif-sur-Yvette, France,*Correspondence: D. Servent, ; A. M. Chudzinski-Tavassi,
| | - A. M. Chudzinski-Tavassi
- Center of Excellence in New Target Discovery (CENTD), Butantan Institute, São Paulo, Brazil,Laboratory of Development and Innovation, Butantan Institute, São Paulo, Brazil,*Correspondence: D. Servent, ; A. M. Chudzinski-Tavassi,
| |
Collapse
|
3
|
Jmel MA, Voet H, Araújo RN, Tirloni L, Sá-Nunes A, Kotsyfakis M. Tick Salivary Kunitz-Type Inhibitors: Targeting Host Hemostasis and Immunity to Mediate Successful Blood Feeding. Int J Mol Sci 2023; 24:1556. [PMID: 36675071 PMCID: PMC9865953 DOI: 10.3390/ijms24021556] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 12/07/2022] [Accepted: 12/08/2022] [Indexed: 01/15/2023] Open
Abstract
Kunitz domain-containing proteins are ubiquitous serine protease inhibitors with promising therapeutic potential. They target key proteases involved in major cellular processes such as inflammation or hemostasis through competitive inhibition in a substrate-like manner. Protease inhibitors from the Kunitz superfamily have a low molecular weight (18-24 kDa) and are characterized by the presence of one or more Kunitz motifs consisting of α-helices and antiparallel β-sheets stabilized by three disulfide bonds. Kunitz-type inhibitors are an important fraction of the protease inhibitors found in tick saliva. Their roles in inhibiting and/or suppressing host homeostatic responses continue to be shown to be additive or synergistic with other protease inhibitors such as cystatins or serpins, ultimately mediating successful blood feeding for the tick. In this review, we discuss the biochemical features of tick salivary Kunitz-type protease inhibitors. We focus on their various effects on host hemostasis and immunity at the molecular and cellular level and their potential therapeutic applications. In doing so, we highlight that their pharmacological properties can be exploited for the development of novel therapies and vaccines.
Collapse
Affiliation(s)
- Mohamed Amine Jmel
- Laboratory of Genomics and Proteomics of Disease Vectors, Institute of Parasitology, Biology Centre, Czech Academy of Sciences, 37005 Ceske Budejovice, Czech Republic
| | - Hanne Voet
- Laboratory of Genomics and Proteomics of Disease Vectors, Institute of Parasitology, Biology Centre, Czech Academy of Sciences, 37005 Ceske Budejovice, Czech Republic
| | - Ricardo N. Araújo
- Laboratory of Hematophagous Arthropods, Department of Parasitology, Federal University of Minas Gerais, Belo Horizonte 31270-901, MG, Brazil
- National Institute of Science and Technology in Molecular Entomology, National Council for Scientific and Technological Development (INCT-EM/CNPq), Rio de Janeiro 21941-902, RJ, Brazil
| | - Lucas Tirloni
- Tick-Pathogen Transmission Unit, Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA
| | - Anderson Sá-Nunes
- National Institute of Science and Technology in Molecular Entomology, National Council for Scientific and Technological Development (INCT-EM/CNPq), Rio de Janeiro 21941-902, RJ, Brazil
- Laboratory of Experimental Immunology, Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo 05508-000, SP, Brazil
| | - Michail Kotsyfakis
- Laboratory of Genomics and Proteomics of Disease Vectors, Institute of Parasitology, Biology Centre, Czech Academy of Sciences, 37005 Ceske Budejovice, Czech Republic
| |
Collapse
|
4
|
Lobba ARM, Alvarez-Flores MP, Fessel MR, Buri MV, Oliveira DS, Gomes RN, Cunegundes PS, DeOcesano-Pereira C, Cinel VD, Chudzinski-Tavassi AM. A Kunitz-type inhibitor from tick salivary glands: A promising novel antitumor drug candidate. Front Mol Biosci 2022; 9:936107. [PMID: 36052162 PMCID: PMC9424826 DOI: 10.3389/fmolb.2022.936107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 07/05/2022] [Indexed: 11/13/2022] Open
Abstract
Salivary glands are vital structures responsible for successful tick feeding. The saliva of ticks contains numerous active molecules that participate in several physiological processes. A Kunitz-type factor Xa (FXa) inhibitor, similar to the tissue factor pathway inhibitor (TFPI) precursor, was identified in the salivary gland transcriptome of Amblyomma sculptum ticks. The recombinant mature form of this Kunitz-type inhibitor, named Amblyomin-X, displayed anticoagulant, antiangiogenic, and antitumor properties. Amblyomin-X is a protein that inhibits FXa in the blood coagulation cascade and acts via non-hemostatic mechanisms, such as proteasome inhibition. Amblyomin-X selectively induces apoptosis in cancer cells and promotes tumor regression through these mechanisms. Notably, the cytotoxicity of Amblyomin-X seems to be restricted to tumor cells and does not affect non-tumorigenic cells, tissues, and organs, making this recombinant protein an attractive molecule for anticancer therapy. The cytotoxic activity of Amblyomin-X on tumor cells has led to vast exploration into this protein. Here, we summarize the function, action mechanisms, structural features, pharmacokinetics, and biodistribution of this tick Kunitz-type inhibitor recombinant protein as a promising novel antitumor drug candidate.
Collapse
Affiliation(s)
- Aline R. M. Lobba
- Centre of Excellence in New Target Discovery-CENTD, Butantan Institute, São Paulo, Brazil
- Development and Innovation Centre, Butantan Institute, Butantan Institute, São Paulo, Brazil
| | - Miryam Paola Alvarez-Flores
- Centre of Excellence in New Target Discovery-CENTD, Butantan Institute, São Paulo, Brazil
- Development and Innovation Centre, Butantan Institute, Butantan Institute, São Paulo, Brazil
| | - Melissa Regina Fessel
- Centre of Excellence in New Target Discovery-CENTD, Butantan Institute, São Paulo, Brazil
- Development and Innovation Centre, Butantan Institute, Butantan Institute, São Paulo, Brazil
| | - Marcus Vinicius Buri
- Centre of Excellence in New Target Discovery-CENTD, Butantan Institute, São Paulo, Brazil
- Development and Innovation Centre, Butantan Institute, Butantan Institute, São Paulo, Brazil
| | - Douglas S. Oliveira
- Centre of Excellence in New Target Discovery-CENTD, Butantan Institute, São Paulo, Brazil
- Development and Innovation Centre, Butantan Institute, Butantan Institute, São Paulo, Brazil
- Biochemistry Department, Federal University of São Paulo, São Paulo, Brazil
| | - Renata N. Gomes
- Centre of Excellence in New Target Discovery-CENTD, Butantan Institute, São Paulo, Brazil
- Development and Innovation Centre, Butantan Institute, Butantan Institute, São Paulo, Brazil
| | - Priscila S. Cunegundes
- Centre of Excellence in New Target Discovery-CENTD, Butantan Institute, São Paulo, Brazil
- Development and Innovation Centre, Butantan Institute, Butantan Institute, São Paulo, Brazil
- Biochemistry Department, Federal University of São Paulo, São Paulo, Brazil
| | - Carlos DeOcesano-Pereira
- Centre of Excellence in New Target Discovery-CENTD, Butantan Institute, São Paulo, Brazil
- Development and Innovation Centre, Butantan Institute, Butantan Institute, São Paulo, Brazil
| | - Victor D. Cinel
- Centre of Excellence in New Target Discovery-CENTD, Butantan Institute, São Paulo, Brazil
- Development and Innovation Centre, Butantan Institute, Butantan Institute, São Paulo, Brazil
- Biochemistry Department, Federal University of São Paulo, São Paulo, Brazil
| | - Ana M. Chudzinski-Tavassi
- Centre of Excellence in New Target Discovery-CENTD, Butantan Institute, São Paulo, Brazil
- Development and Innovation Centre, Butantan Institute, Butantan Institute, São Paulo, Brazil
- Biochemistry Department, Federal University of São Paulo, São Paulo, Brazil
- *Correspondence: Ana M. Chudzinski-Tavassi,
| |
Collapse
|
5
|
Ma L, Xuan X, Fan M, Zhang Y, Yuan G, Huang G, Liu Z. A novel 8-hydroxyquinoline derivative induces breast cancer cell death through paraptosis and apoptosis. Apoptosis 2022; 27:577-589. [PMID: 35674852 DOI: 10.1007/s10495-022-01737-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/17/2022] [Indexed: 11/02/2022]
Abstract
Chemotherapy represents one of the main conventional therapies for breast cancer. However, tumor cells develop mechanisms to evade chemotherapeutic-induced apoptosis. Thus, it is of great significance to induce non-apoptotic cell death modes, such as paraptosis, in breast cancer. Herein, a novel 8-hydroxyquinoline derivative, 5,7-dibromo-8-(methoxymethoxy)-2-methylquinoline (HQ-11), was obtained and its potential anti-breast cancer mechanisms were investigated. Our results showed that extensive cytoplasmic vacuoles derived from the endoplasmic reticulum (ER) and mitochondria were appeared in MCF7 and MDA-MB-231 breast cancer cells by HQ-11 incubation, and pretreatment of cycloheximide was able to inhibit this vacuolation and HQ-11-induced cell death, showing the characteristics of paraptosis. ER stress was involved in HQ-11-caused paraptosis evidenced by the increase of glucose-regulated protein 78, C/EBP homologous protein and polyubiquitinated proteins. Molecular docking analysis revealed a favorable binding mode of HQ-11 in the active site of the chymotrypsin-like β5 subunit of the proteasome, indicative of proteasome dysfunction under HQ-11 treatment, which might result in further aggravated ER stress. Furthermore, treatment of HQ-11 resulted in increased phosphorylation of extracellular signal-regulated kinase (ERK) and c-Jun NH2-terminal kinase, and inhibition of ERK with U0126 significantly attenuated HQ-11-induced ER stress and paraptosis. In addition, exposure to HQ-11 also caused apoptosis in breast cancer cells partially through activation of ERK pathway. All these results conclusively indicate that HQ-11 triggers two distinct cell death modes via inhibition of proteasome and activation of ERK pathway in breast cancer cells, providing a promising candidate in future anti-breast cancer therapy.
Collapse
Affiliation(s)
- Liang Ma
- Department of Chemical Biology and Pharmaceutical Engineering, School of Chemistry and Chemical Engineering, Anhui University of Technology, 59 Hudong Road, Ma'anshan, 243002, Anhui, China
| | - Xiaojing Xuan
- Department of Chemical Biology and Pharmaceutical Engineering, School of Chemistry and Chemical Engineering, Anhui University of Technology, 59 Hudong Road, Ma'anshan, 243002, Anhui, China
| | - Minghui Fan
- Department of Chemical Biology and Pharmaceutical Engineering, School of Chemistry and Chemical Engineering, Anhui University of Technology, 59 Hudong Road, Ma'anshan, 243002, Anhui, China
| | - Yumeng Zhang
- Department of Chemical Biology and Pharmaceutical Engineering, School of Chemistry and Chemical Engineering, Anhui University of Technology, 59 Hudong Road, Ma'anshan, 243002, Anhui, China
| | - Guozan Yuan
- Department of Chemical Biology and Pharmaceutical Engineering, School of Chemistry and Chemical Engineering, Anhui University of Technology, 59 Hudong Road, Ma'anshan, 243002, Anhui, China
| | - Guozheng Huang
- Department of Chemical Biology and Pharmaceutical Engineering, School of Chemistry and Chemical Engineering, Anhui University of Technology, 59 Hudong Road, Ma'anshan, 243002, Anhui, China
| | - Zi Liu
- Department of Chemical Biology and Pharmaceutical Engineering, School of Chemistry and Chemical Engineering, Anhui University of Technology, 59 Hudong Road, Ma'anshan, 243002, Anhui, China.
| |
Collapse
|
6
|
Meng X, Cui X, Shao X, Liu Y, Xing Y, Smith V, Xiong S, Macip S, Chen Y. poly(I:C) synergizes with proteasome inhibitors to induce apoptosis in cervical cancer cells. Transl Oncol 2022; 18:101362. [PMID: 35151092 PMCID: PMC8842080 DOI: 10.1016/j.tranon.2022.101362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 01/28/2022] [Indexed: 11/24/2022] Open
|
7
|
Nesari A, Mansouri MT, Khodayar MJ, Rezaei M. Preadministration of high-dose alpha-tocopherol improved memory impairment and mitochondrial dysfunction induced by proteasome inhibition in rat hippocampus. Nutr Neurosci 2021; 24:119-129. [PMID: 31084475 DOI: 10.1080/1028415x.2019.1601888] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Objective: The ubiquitin-proteasome system plays a key role in memory consolidation. Proteasome inhibition and free radical-induced neural damage were implicated in neurodegenerative states. In this study, it was tested whether alpha-tocopherol (αT) in low and high doses could improve the long-term memory impairment induced by proteasome inhibition and protects against hippocampal oxidative stress. Methods: Alpha-tocopherol (αT) (60, 200 mg/kg, i.p. for 5 days) was administered to rats with memory deficit and hippocampal oxidative stress induced by bilateral intra-hippocampal injection of lactacystin (32 ng/μl) and mitochondrial evaluations were performed for improvement assessments. Results: The results showed that lactacystin significantly reduced the passive avoidance memory performance and increased the level of malondialdehyde (MDA), reactive oxygen species (ROS) and diminished the mitochondrial membrane potential (MMP) in the rat hippocampus. Furthermore, Intraperitoneal administration of αT significantly increased the passive avoidance memory, glutathione content and reduced ROS, MDA levels and impaired MMP. Conclusions: The results suggested that αT has neuroprotective effects against lactacystin-induced oxidative stress and memory impairment via the enhancement of hippocampal antioxidant capacity and concomitant mitochondrial sustainability. This finding shows a way to prevent and also to treat neurodegenerative diseases associated with mitochondrial impairment.
Collapse
Affiliation(s)
- Ali Nesari
- Department of Toxicology, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohammad Taghi Mansouri
- Department of Pharmacology, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Department of Anesthesiology, Irving Medical Center, Columbia University, New York, NY, USA
| | - Mohammad Javad Khodayar
- Department of Toxicology, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Toxicology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohsen Rezaei
- Toxicology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Department of Toxicology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
8
|
Jmel MA, Aounallah H, Bensaoud C, Mekki I, Chmelař J, Faria F, M’ghirbi Y, Kotsyfakis M. Insights into the Role of Tick Salivary Protease Inhibitors during Ectoparasite-Host Crosstalk. Int J Mol Sci 2021; 22:E892. [PMID: 33477394 PMCID: PMC7831016 DOI: 10.3390/ijms22020892] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Revised: 01/12/2021] [Accepted: 01/13/2021] [Indexed: 02/07/2023] Open
Abstract
Protease inhibitors (PIs) are ubiquitous regulatory proteins present in all kingdoms. They play crucial tasks in controlling biological processes directed by proteases which, if not tightly regulated, can damage the host organism. PIs can be classified according to their targeted proteases or their mechanism of action. The functions of many PIs have now been characterized and are showing clinical relevance for the treatment of human diseases such as arthritis, hepatitis, cancer, AIDS, and cardiovascular diseases, amongst others. Other PIs have potential use in agriculture as insecticides, anti-fungal, and antibacterial agents. PIs from tick salivary glands are special due to their pharmacological properties and their high specificity, selectivity, and affinity to their target proteases at the tick-host interface. In this review, we discuss the structure and function of PIs in general and those PI superfamilies abundant in tick salivary glands to illustrate their possible practical applications. In doing so, we describe tick salivary PIs that are showing promise as drug candidates, highlighting the most promising ones tested in vivo and which are now progressing to preclinical and clinical trials.
Collapse
Affiliation(s)
- Mohamed Amine Jmel
- Laboratory of Genomics and Proteomics of Disease Vectors, Biology Centre CAS, Institute of Parasitology, Branišovská 1160/31, 37005 České Budějovice, Czech Republic; (M.A.J.); (C.B.); (I.M.)
| | - Hajer Aounallah
- Institut Pasteur de Tunis, Université de Tunis El Manar, LR19IPTX, Service d’Entomologie Médicale, Tunis 1002, Tunisia; (H.A.); (Y.M.)
- Innovation and Development Laboratory, Innovation and Development Center, Instituto Butantan, São Paulo 05503-900, Brazil;
| | - Chaima Bensaoud
- Laboratory of Genomics and Proteomics of Disease Vectors, Biology Centre CAS, Institute of Parasitology, Branišovská 1160/31, 37005 České Budějovice, Czech Republic; (M.A.J.); (C.B.); (I.M.)
| | - Imen Mekki
- Laboratory of Genomics and Proteomics of Disease Vectors, Biology Centre CAS, Institute of Parasitology, Branišovská 1160/31, 37005 České Budějovice, Czech Republic; (M.A.J.); (C.B.); (I.M.)
- Faculty of Science, University of South Bohemia in České Budějovice, 37005 České Budějovice, Czech Republic;
| | - Jindřich Chmelař
- Faculty of Science, University of South Bohemia in České Budějovice, 37005 České Budějovice, Czech Republic;
| | - Fernanda Faria
- Innovation and Development Laboratory, Innovation and Development Center, Instituto Butantan, São Paulo 05503-900, Brazil;
| | - Youmna M’ghirbi
- Institut Pasteur de Tunis, Université de Tunis El Manar, LR19IPTX, Service d’Entomologie Médicale, Tunis 1002, Tunisia; (H.A.); (Y.M.)
| | - Michalis Kotsyfakis
- Laboratory of Genomics and Proteomics of Disease Vectors, Biology Centre CAS, Institute of Parasitology, Branišovská 1160/31, 37005 České Budějovice, Czech Republic; (M.A.J.); (C.B.); (I.M.)
| |
Collapse
|
9
|
Colella F, Scillitani G, Pierri CL. Sweet as honey, bitter as bile: Mitochondriotoxic peptides and other therapeutic proteins isolated from animal tissues, for dealing with mitochondrial apoptosis. Toxicology 2020; 447:152612. [PMID: 33171268 DOI: 10.1016/j.tox.2020.152612] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 10/02/2020] [Accepted: 10/06/2020] [Indexed: 02/06/2023]
Abstract
Mitochondria are subcellular organelles involved in cell metabolism and cell life-cycle. Their role in apoptosis regulation makes them an interesting target of new drugs for dealing with cancer or rare diseases. Several peptides and proteins isolated from animal and plant sources are known for their therapeutic properties and have been tested on cancer cell-lines and xenograft murine models, highlighting their ability in inducing cell-death by triggering mitochondrial apoptosis. Some of those molecules have been even approved as drugs. Conversely, many other bioactive compounds are still under investigation for their proapoptotic properties. In this review we report about a group of peptides, isolated from animal venoms, with potential therapeutic properties related to their ability in triggering mitochondrial apoptosis. This class of compounds is known with different names, such as mitochondriotoxins or mitocans.
Collapse
Affiliation(s)
- Francesco Colella
- Laboratory of Biochemistry, Molecular and Structural Biology, Department of Biosciences, Biotechnologies, Biopharmaceutics, University of Bari, Via E. Orabona, 4, 70125, Bari, Italy
| | | | - Ciro Leonardo Pierri
- Laboratory of Biochemistry, Molecular and Structural Biology, Department of Biosciences, Biotechnologies, Biopharmaceutics, University of Bari, Via E. Orabona, 4, 70125, Bari, Italy; BROWSer S.r.l. (https://browser-bioinf.com/) c/o Department of Biosciences, Biotechnologies, Biopharmaceutics, University "Aldo Moro" of Bari, Via E. Orabona, 4, 70126, Bari, Italy.
| |
Collapse
|
10
|
Lichtenstein F, Iqbal A, de Lima Will SEA, Bosch RV, DeOcesano-Pereira C, Goldfeder MB, Chammas R, Trufen CEM, Morais KLP, de Souza JG, Natalino RJM, de Azevedo IJ, Nishiyama Junior MY, Oliveira U, Alves FIA, Araujo JM, Lobba ARM, Chudzinski-Tavassi AM. Modulation of stress and immune response by Amblyomin-X results in tumor cell death in a horse melanoma model. Sci Rep 2020; 10:6388. [PMID: 32286411 PMCID: PMC7156751 DOI: 10.1038/s41598-020-63275-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 03/17/2020] [Indexed: 02/08/2023] Open
Abstract
We have investigated Amblyomin-X-treated horse melanomas to better understand its mode of action through transcriptome analysis and the in vivo model. Amblyomin-X is a Kunitz-type homologous protein that selectively leads to the death of tumor cells via ER stress and apoptosis, currently under investigation as a new drug candidate for cancer treatment. Melanomas are immunogenic tumors, and a better understanding of the immune responses is warranted. Equine melanomas are spontaneous and not so aggressive as human melanomas are, as this study shows that the in vivo treatment of encapsulated horse melanoma tumors led to a significant reduction in the tumor size or even the complete disappearance of the tumor mass through intratumoral injections of Amblyomin-X. Transcriptome analysis identified ER- and mitochondria-stress, modulation of the innate immune system, apoptosis, and possibly immunogenic cell death activation. Interactome analysis showed that Amblyomin-X potentially interacts with key elements found in transcriptomics. Taken together, Amblyomin-X modulated the tumor immune microenvironment in different ways, at least contributing to induce tumor cell death.
Collapse
Affiliation(s)
- Flavio Lichtenstein
- Laboratory of Molecular Biology, Butantan Institute, São Paulo, SP, Brazil.,CENTD, Centre of Excellence in New Target Discovery, Butantan Institute, São Paulo, Brazil
| | - Asif Iqbal
- Laboratory of Molecular Biology, Butantan Institute, São Paulo, SP, Brazil.,CENTD, Centre of Excellence in New Target Discovery, Butantan Institute, São Paulo, Brazil
| | - Sonia Elisabete Alves de Lima Will
- Laboratory of Molecular Biology, Butantan Institute, São Paulo, SP, Brazil.,CENTD, Centre of Excellence in New Target Discovery, Butantan Institute, São Paulo, Brazil
| | - Rosemary Viola Bosch
- Laboratory of Molecular Biology, Butantan Institute, São Paulo, SP, Brazil.,CENTD, Centre of Excellence in New Target Discovery, Butantan Institute, São Paulo, Brazil
| | - Carlos DeOcesano-Pereira
- Laboratory of Molecular Biology, Butantan Institute, São Paulo, SP, Brazil.,CENTD, Centre of Excellence in New Target Discovery, Butantan Institute, São Paulo, Brazil
| | - Mauricio Barbugiani Goldfeder
- Laboratory of Molecular Biology, Butantan Institute, São Paulo, SP, Brazil.,CENTD, Centre of Excellence in New Target Discovery, Butantan Institute, São Paulo, Brazil
| | - Roger Chammas
- ICESP, Center for Translational Research in Oncology, Instituto do Câncer do Estado de São Paulo, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Carlos Eduardo Madureira Trufen
- Laboratory of Molecular Biology, Butantan Institute, São Paulo, SP, Brazil.,CENTD, Centre of Excellence in New Target Discovery, Butantan Institute, São Paulo, Brazil
| | - Katia Luciano Pereira Morais
- Laboratory of Molecular Biology, Butantan Institute, São Paulo, SP, Brazil.,CENTD, Centre of Excellence in New Target Discovery, Butantan Institute, São Paulo, Brazil
| | - Jean Gabriel de Souza
- Laboratory of Molecular Biology, Butantan Institute, São Paulo, SP, Brazil.,CENTD, Centre of Excellence in New Target Discovery, Butantan Institute, São Paulo, Brazil
| | - Renato Jose Mendonça Natalino
- ICESP, Center for Translational Research in Oncology, Instituto do Câncer do Estado de São Paulo, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | | | | | - Ursula Oliveira
- Laboratório Especial de Toxinologia Aplicada - CeTICS, Butantan Institute, São Paulo, Brazil
| | - Francisco Ivanio Arruda Alves
- Laboratory of Molecular Biology, Butantan Institute, São Paulo, SP, Brazil.,CENTD, Centre of Excellence in New Target Discovery, Butantan Institute, São Paulo, Brazil
| | - Jaqueline Mayara Araujo
- Laboratory of Molecular Biology, Butantan Institute, São Paulo, SP, Brazil.,CENTD, Centre of Excellence in New Target Discovery, Butantan Institute, São Paulo, Brazil
| | - Aline Ramos Maia Lobba
- Laboratory of Molecular Biology, Butantan Institute, São Paulo, SP, Brazil.,CENTD, Centre of Excellence in New Target Discovery, Butantan Institute, São Paulo, Brazil
| | - Ana Marisa Chudzinski-Tavassi
- Laboratory of Molecular Biology, Butantan Institute, São Paulo, SP, Brazil. .,CENTD, Centre of Excellence in New Target Discovery, Butantan Institute, São Paulo, Brazil.
| |
Collapse
|
11
|
Pavon LF, Capper D, Sibov TT, de Toledo SRC, Thomale UW, de Souza JG, Cabral FR, Berra CM, Silva da Costa MD, Mendonça Niçacio J, Dastoli PA, de Oliveira DM, Malheiros SMF, da Cruz EF, Malheiros JM, de Oliveira SM, Silva NS, Petrilli AS, Cappellano AM, Brunialti MC, Salomão R, de Paiva Neto MA, Chudzinski-Tavassi AM, Cavalheiro S. New therapeutic target for pediatric anaplastic ependymoma control: study of anti-tumor activity by a Kunitz-type molecule, Amblyomin-X. Sci Rep 2019; 9:9973. [PMID: 31292491 PMCID: PMC6620274 DOI: 10.1038/s41598-019-45799-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 05/30/2019] [Indexed: 11/08/2022] Open
Abstract
EPNs comprise a heterogeneous group of neuroepithelial tumors, accounting for about 10% of all intracranial tumors in children and up to 30% of brain tumors in those younger than 3 years. Actually, the pattern therapy for low-grade EPNs includes complete surgical resection followed by radiation therapy. Total surgical excision is often not possible due to tumor location. The aim of this study was to evaluate, for the first time, the anti-tumor activity of Amblyomin-X in 4 primary cultures derived from pediatric anaplastic posterior fossa EPN, Group A (anaplastic, WHO grade III) and one primary culture of a high grade neuroepithelial tumor with MN1 alteration, which was initially misdiagnosed as EPN: i) by in vitro assays: comparisons of temozolomide and cisplatin; ii) by intracranial xenograft model. Amblyomin-X was able to induce cell death in EPN cells in a more significant percentage compared to cisplatin. The cytotoxic effects of Amblyomin-X were not detected on hFSCs used as control, as opposed to cisplatin-treatment, which promoted a substantial effect in the hAFSCs viability. TEM analysis showed ultrastructural alterations related to the process of cell death: mitochondrial degeneration, autophagosomes and aggregate-like structures. MRI and histopathological analyzes demonstrated significant tumor mass regression. Our results suggest that Amblyomin-X has a selective effect on tumor cells by inducing apoptotic cell death and may be a therapeutic option for Group AEPNs.
Collapse
Affiliation(s)
- Lorena Favaro Pavon
- Discipline of Neurosurgery, Federal University of São Paulo, São Paulo, São Paulo, Brazil.
| | - David Capper
- Charité -Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universitätzu Berlin, and Berlin Institute of Health, Department of Neuropathology, Berlin, Germany
- German Cancer Consortium (DKTK), Partner Site Berlin, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Tatiana Tais Sibov
- Discipline of Neurosurgery, Federal University of São Paulo, São Paulo, São Paulo, Brazil
| | - Silvia Regina Caminada de Toledo
- Pediatric Oncology Institute, Grupo de Apoio ao Adolescente e à Criança com Câncer (GRAACC), Federal University of São Paulo, São Paulo, São Paulo, Brazil
| | - Ulrich-W Thomale
- Pediatric Neurosurgery, Campus Virchow Klinikum, Charité Universitätsmedizin, Berlin, Germany
| | - Jean Gabriel de Souza
- Laboratory of Molecular Biology, Butantan Institute, São Paulo, São Paulo, Brazil
- Centre of Excellence in New Target Discovery (CENTD), Butantan Institute, São Paulo, São Paulo, Brazil
| | | | - Carolina Maria Berra
- Department of Pharmacology, Institute of Biomedical Science, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Marcos Devanir Silva da Costa
- Discipline of Neurosurgery, Federal University of São Paulo, São Paulo, São Paulo, Brazil
- Pediatric Oncology Institute, Grupo de Apoio ao Adolescente e à Criança com Câncer (GRAACC), Federal University of São Paulo, São Paulo, São Paulo, Brazil
| | - Jardel Mendonça Niçacio
- Discipline of Neurosurgery, Federal University of São Paulo, São Paulo, São Paulo, Brazil
- Pediatric Oncology Institute, Grupo de Apoio ao Adolescente e à Criança com Câncer (GRAACC), Federal University of São Paulo, São Paulo, São Paulo, Brazil
| | - Patrícia Alessandra Dastoli
- Discipline of Neurosurgery, Federal University of São Paulo, São Paulo, São Paulo, Brazil
- Pediatric Oncology Institute, Grupo de Apoio ao Adolescente e à Criança com Câncer (GRAACC), Federal University of São Paulo, São Paulo, São Paulo, Brazil
| | | | - Suzana M F Malheiros
- Discipline of Neurosurgery, Federal University of São Paulo, São Paulo, São Paulo, Brazil
- Hospital Israelita Albert Einstein, São Paulo, São Paulo, Brazil
| | | | | | | | - Nasjla Saba Silva
- Pediatric Oncology Institute, Grupo de Apoio ao Adolescente e à Criança com Câncer (GRAACC), Federal University of São Paulo, São Paulo, São Paulo, Brazil
| | - Antonio Sérgio Petrilli
- Pediatric Oncology Institute, Grupo de Apoio ao Adolescente e à Criança com Câncer (GRAACC), Federal University of São Paulo, São Paulo, São Paulo, Brazil
| | - Andrea Maria Cappellano
- Pediatric Oncology Institute, Grupo de Apoio ao Adolescente e à Criança com Câncer (GRAACC), Federal University of São Paulo, São Paulo, São Paulo, Brazil
| | - Milena Colò Brunialti
- Laboratory of Immunology and Infectology, Federal University of São Paulo, São Paulo, São Paulo, Brazil
| | - Reinaldo Salomão
- Laboratory of Immunology and Infectology, Federal University of São Paulo, São Paulo, São Paulo, Brazil
| | - Manoel A de Paiva Neto
- Discipline of Neurosurgery, Federal University of São Paulo, São Paulo, São Paulo, Brazil
| | - Ana Marisa Chudzinski-Tavassi
- Laboratory of Molecular Biology, Butantan Institute, São Paulo, São Paulo, Brazil.
- Centre of Excellence in New Target Discovery (CENTD), Butantan Institute, São Paulo, São Paulo, Brazil.
| | - Sérgio Cavalheiro
- Discipline of Neurosurgery, Federal University of São Paulo, São Paulo, São Paulo, Brazil
- Pediatric Oncology Institute, Grupo de Apoio ao Adolescente e à Criança com Câncer (GRAACC), Federal University of São Paulo, São Paulo, São Paulo, Brazil
| |
Collapse
|
12
|
Štibrániová I, Bartíková P, Holíková V, Kazimírová M. Deciphering Biological Processes at the Tick-Host Interface Opens New Strategies for Treatment of Human Diseases. Front Physiol 2019; 10:830. [PMID: 31333488 PMCID: PMC6617849 DOI: 10.3389/fphys.2019.00830] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Accepted: 06/17/2019] [Indexed: 12/14/2022] Open
Abstract
Ticks are obligatory blood-feeding ectoparasites, causing blood loss and skin damage in their hosts. In addition, ticks also transmit a number of various pathogenic microorganisms that cause serious diseases in humans and animals. Ticks evolved a wide array of salivary bioactive compounds that, upon injection into the host skin, inhibit or modulate host reactions such as hemostasis, inflammation and wound healing. Modulation of the tick attachment site in the host skin involves mainly molecules which affect physiological processes orchestrated by cytokines, chemokines and growth factors. Suppressing host defense reactions is crucial for tick survival and reproduction. Furthermore, pharmacologically active compounds in tick saliva have a promising therapeutic potential for treatment of some human diseases connected with disorders in hemostasis and immune system. These disorders are often associated to alterations in signaling pathways and dysregulation or overexpression of specific cytokines which, in turn, affect mechanisms of angiogenesis, cell motility and cytoskeletal regulation. Moreover, tick salivary molecules were found to exert cytotoxic and cytolytic effects on various tumor cells and have anti-angiogenic properties. Elucidation of the mode of action of tick bioactive molecules on the regulation of cell processes in their mammalian hosts could provide new tools for understanding the complex changes leading to immune disorders and cancer. Tick bioactive molecules may also be exploited as new pharmacological inhibitors of the signaling pathways of cytokines and thus help alleviate patient discomfort and increase patient survival. We review the current knowledge about tick salivary peptides and proteins that have been identified and functionally characterized in in vitro and/or in vivo models and their therapeutic perspective.
Collapse
Affiliation(s)
- Iveta Štibrániová
- Biomedical Research Center, Institute of Virology, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Pavlína Bartíková
- Biomedical Research Center, Institute of Virology, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Viera Holíková
- Biomedical Research Center, Institute of Virology, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Mária Kazimírová
- Institute of Zoology, Slovak Academy of Sciences, Bratislava, Slovakia
| |
Collapse
|
13
|
Binoy A, Nedungadi D, Katiyar N, Bose C, Shankarappa SA, Nair BG, Mishra N. Plumbagin induces paraptosis in cancer cells by disrupting the sulfhydryl homeostasis and proteasomal function. Chem Biol Interact 2019; 310:108733. [PMID: 31276663 DOI: 10.1016/j.cbi.2019.108733] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 06/27/2019] [Accepted: 07/01/2019] [Indexed: 12/20/2022]
Abstract
Plumbagin (PLB) is an active secondary metabolite extracted from the roots of Plumbago rosea. In this study, we report that plumbagin effectively induces paraptosis by triggering extensive cytoplasmic vacuolation followed by cell death in triple negative breast cancer cells (MDA-MB-231), cervical cancer cells (HeLa) and non-small lung cancer cells (A549) but not in normal lung fibroblast cells (WI-38). The vacuoles originated from the dilation of the endoplasmic reticulum (ER) and were found to be empty. The cell death induced by plumbagin was neither apoptotic nor autophagic. Plumbagin induced ER stress mainly by inhibiting the chymotrypsin-like activity of 26S proteasome as also evident from the accumulation of polyubiquitinated proteins. The vacuolation and cell death were found to be independent of reactive oxygen species generation but was effectively inhibited by thiol antioxidant suggesting that plumbagin could modify the sulfur homeostasis in the cellular milieu. Plumbagin also resulted in a decrease in mitochondrial membrane potential eventually decreasing the ATP production. This is the first study to show that Plumbagin induces paraptosis through proteasome inhibition and disruption of sulfhydryl homeostasis and thus further opens up the lead molecule to potential therapeutic strategies for apoptosis-resistant cancers.
Collapse
Affiliation(s)
- Anupama Binoy
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Amritapuri, Clappana P.O., Kollam, 690525, Kerala, India
| | - Divya Nedungadi
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Amritapuri, Clappana P.O., Kollam, 690525, Kerala, India
| | - Neeraj Katiyar
- Center for Nanosciences & Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, 682041, Kerala, India
| | - Chinchu Bose
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Amritapuri, Clappana P.O., Kollam, 690525, Kerala, India
| | - Sahadev A Shankarappa
- Center for Nanosciences & Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, 682041, Kerala, India
| | - Bipin G Nair
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Amritapuri, Clappana P.O., Kollam, 690525, Kerala, India
| | - Nandita Mishra
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Amritapuri, Clappana P.O., Kollam, 690525, Kerala, India.
| |
Collapse
|
14
|
ER stress promotes antitumor effects in BRAFi/MEKi resistant human melanoma induced by natural compound 4-nerolidylcathecol (4-NC). Pharmacol Res 2018; 141:63-72. [PMID: 30550954 DOI: 10.1016/j.phrs.2018.12.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Accepted: 12/10/2018] [Indexed: 12/12/2022]
Abstract
Melanoma accounts for only 4% of malignant neoplasms of the skin, but is considered the most serious because it is highly deadly. Mutations in the MAPK (Ras-Raf-MEK-ERK) pathway is closely linked to the lack of control of cell proliferation. Especially in melanoma, this pathway has become a target for the development of oncogene-targeted therapies, such as the potent inhibitors of v-Raf murine sarcoma viral oncogene homolog B (BRAFi) and mitogen-activated protein kinase kinase (MEKi). Very high rates of response have been achieved, but most patients are relapsed due to the development of resistance, justifying the constant search for new therapeutic compounds. Early results from our group indicated that 4-nerolidylcatechol (4-NC), a catechol compound extracted from Pothomorphe umbellata, induces DNA damage, ROS production, increased p53 expression culminating in apoptosis in melanoma but with no data regarding the 4-NC effects in cells resistant to BRAFi or MEKi. Therefore, here we evaluated the role of 4-NC alone or in combination with BRAFi/MEKi in resistant melanoma cells. Double-resistant cells were generated and characterized by MAPK pathway reactivation. 4-NC alone or in combination (30 μM) with MAPK inhibitors was cytotoxic, inhibited colony formation and decreased invasiveness in two and three-dimensional cell culture models of treatment-naïve, BRAFi-resistant and BRAF/MEKi double-resistant melanoma cells. Apoptosis induction was demonstrated in resistant and double-resistant melanoma cell lines after 4-NC treatments. 4-NC showed important ability to induce apoptosis via Endoplasmatic Reticulum (ER) stress and specifically BiP and CHOP that had increased protein expression in all melanoma cell lines proving to be part of the ER stress pathway activation. CHOP knockdown slightly but enough increases cellular viability following 4-NC treatment indicating that apoptosis observed is partially dependent on CHOP. In summary, we show that 4-NC is a compound with activity against cutaneous melanoma, including resistant cells to clinically approved therapies.
Collapse
|
15
|
Maria DA, Will SEAL, Bosch RV, Souza JG, Sciani JM, Goldfeder MB, Rondon GG, Chudzinski-Tavassi AM. Preclinical evaluation of Amblyomin-X, a Kunitz-type protease inhibitor with antitumor activity. Toxicol Rep 2018; 6:51-63. [PMID: 30581760 PMCID: PMC6298944 DOI: 10.1016/j.toxrep.2018.11.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Revised: 11/28/2018] [Accepted: 11/29/2018] [Indexed: 12/24/2022] Open
Abstract
Amblyomin-X is a recombinant protein with selective antitumor activity, causing tumor reduction in vivo. Acute and subchronic toxicity studies shows low toxicity in healthy mice, with reversible adverse effects. Amblyomin-X toxicity profile was defined.
Amblyomin-X, a Kunitz-type protease inhibitor, is a recombinant protein that selectively induces apoptosis in tumor cells and promotes tumor reduction in vivo in melanoma animal models. Furthermore, Amblyomin-X was able to drastically reduce lung metastasis in a mice orthotopic kidney tumor model. Due to its antitumor activity, Amblyomin-X potential to become a new drug is currently under investigation, therefore the aim of the present study was to perform preclinical assays to evaluate Amblyomin-X toxicity in healthy mice. Exploratory toxicity assays have shown that treatment with 512 mg/kg of Amblyomin-X lead to animal mortality, therefore two groups of treatment were evaluated in the present work: in the acute toxicity assay, animals were injected once with doses ranging from 4 to 256 mg/kg of Amblyomin-X, while in the subacute toxicity assay, animals were injected with 0.25, 0.57 and 1 mg/kg of Amblyomin-X daily, during 28 days. Following this treatment regimens, Amblyomin-X did not cause any mortality; moreover, toxicity signs were discrete, reversible and observed only at the higher doses, thus establishing a safety profile for administration in mice, which can be further used to determine the dose translation of this novel drug candidate for treatment in other species.
Collapse
Affiliation(s)
- Durvanei A Maria
- Laboratório de Biologia Molecular, Instituto Butantan, Av. Vital Brasil, 1500, São Paulo, 05503-900, SP, Brazil
| | - Sonia Elisabete A L Will
- Laboratório de Biologia Molecular, Instituto Butantan, Av. Vital Brasil, 1500, São Paulo, 05503-900, SP, Brazil
| | - Rosemary V Bosch
- Laboratório de Biologia Molecular, Instituto Butantan, Av. Vital Brasil, 1500, São Paulo, 05503-900, SP, Brazil
| | - Jean G Souza
- Laboratório de Biologia Molecular, Instituto Butantan, Av. Vital Brasil, 1500, São Paulo, 05503-900, SP, Brazil
| | - Juliana M Sciani
- Laboratório de Biologia Molecular, Instituto Butantan, Av. Vital Brasil, 1500, São Paulo, 05503-900, SP, Brazil
| | - Mauricio B Goldfeder
- Laboratório de Biologia Molecular, Instituto Butantan, Av. Vital Brasil, 1500, São Paulo, 05503-900, SP, Brazil
| | - Giuliana G Rondon
- Laboratório de Biologia Molecular, Instituto Butantan, Av. Vital Brasil, 1500, São Paulo, 05503-900, SP, Brazil
| | - Ana M Chudzinski-Tavassi
- Laboratório de Biologia Molecular, Instituto Butantan, Av. Vital Brasil, 1500, São Paulo, 05503-900, SP, Brazil
| |
Collapse
|
16
|
Schmidt MCB, Morais KLP, Almeida MESD, Iqbal A, Goldfeder MB, Chudzinski-Tavassi AM. Amblyomin-X, a recombinant Kunitz-type inhibitor, regulates cell adhesion and migration of human tumor cells. Cell Adh Migr 2018; 14:129-138. [PMID: 30238848 PMCID: PMC7527229 DOI: 10.1080/19336918.2018.1516982] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
Abstract
In a tumor microenvironment, endothelial cell migration and angiogenesis allow cancer to spread to other organs causing metastasis. Indeed, a number of molecules that are involved in cytoskeleton re-organization and intracellular signaling have been investigated for their effects on tumor cell growth and metastasis. Alongside that, Amblyomin-X, a recombinant Kunitz-type protein, has been shown to reduce metastasis and tumor growth in in vivo experiments. In the present report, we provide a mechanistic insight to these antitumor effects, this is, Amblyomin-X modulates Rho-GTPases and uPAR signaling, and reduces the release of MMPs, leading to disruption of the actin cytoskeleton and decreased cell migration of tumor cell lines. Altogether, our data support a role for Amblyomin-X as a novel potential antitumor drug. ABBREVIATIONS Amb-X: Amblyomin-X; ECGF: endotelial cell growth factor; ECM: extracellular matrix; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; HUVEC: human umbilical vein endothelial cell; LRP1: low-density lipoprotein receptor-related protein; MMP: matrix metalloproteinase; HPI-4: hedgehog pathway inhibitor 4; PAI-1: plasminogen activator inhibitor 1; PMA: phorbol 12-myristate-13-acetate; TFPI: tissue factor pathway inhibitor; uPA: urokinase plasminogen activator; uPAR: uPA receptor.
Collapse
Affiliation(s)
- Mariana Costa Braga Schmidt
- Laboratory of Molecular Biology, Butantan Institute , São Paulo, SP, Brazil.,Departament of Biochemistry, Federal University of São Paulo , São Paulo, SP, Brazil
| | - Katia L P Morais
- Laboratory of Molecular Biology, Butantan Institute , São Paulo, SP, Brazil.,Departament of Biochemistry, Federal University of São Paulo , São Paulo, SP, Brazil.,Centre of Excellence in New Target Discovery, Butantan Institute , São Paulo, SP, Brazil
| | - Maíra Estanislau Soares de Almeida
- Laboratory of Molecular Biology, Butantan Institute , São Paulo, SP, Brazil.,Centre of Excellence in New Target Discovery, Butantan Institute , São Paulo, SP, Brazil
| | - Asif Iqbal
- Laboratory of Molecular Biology, Butantan Institute , São Paulo, SP, Brazil.,Centre of Excellence in New Target Discovery, Butantan Institute , São Paulo, SP, Brazil
| | - Mauricio Barbugiani Goldfeder
- Laboratory of Molecular Biology, Butantan Institute , São Paulo, SP, Brazil.,Centre of Excellence in New Target Discovery, Butantan Institute , São Paulo, SP, Brazil
| | - Ana Marisa Chudzinski-Tavassi
- Laboratory of Molecular Biology, Butantan Institute , São Paulo, SP, Brazil.,Departament of Biochemistry, Federal University of São Paulo , São Paulo, SP, Brazil
| |
Collapse
|
17
|
Parizi LF, Ali A, Tirloni L, Oldiges DP, Sabadin GA, Coutinho ML, Seixas A, Logullo C, Termignoni C, DA Silva Vaz I. Peptidase inhibitors in tick physiology. MEDICAL AND VETERINARY ENTOMOLOGY 2018; 32:129-144. [PMID: 29111611 DOI: 10.1111/mve.12276] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 06/23/2017] [Accepted: 10/07/2017] [Indexed: 06/07/2023]
Abstract
Peptidase inhibitors regulate a wide range of physiological processes involved in the interaction between hematophagous parasites and their hosts, including tissue remodeling, the immune response and blood coagulation. In tick physiology, peptidase inhibitors have a crucial role in adaptation to improve parasitism mechanisms, facilitating blood feeding by interfering with defense-related host peptidases. Recently, a larger number of studies on this topic led to the description of several new tick inhibitors displaying interesting novel features, for example a role in pathogen transmission to the host. A comprehensive review discussing these emerging concepts can therefore shed light on peptidase inhibitor functions, their relevance to tick physiology and their potential applications. Here, we summarize and examine the general characteristics, functional diversity and action of tick peptidase inhibitors with known physiological roles in the tick-host-pathogen interaction.
Collapse
Affiliation(s)
- L F Parizi
- Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Faculdade de Veterinária, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - A Ali
- Department of Zoology, Abdul Wali Khan University, Mardan, Pakistan
- Escola de Enfermagem de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brazil
| | - L Tirloni
- Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - D P Oldiges
- Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - G A Sabadin
- Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - M L Coutinho
- Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Faculdade de Veterinária, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - A Seixas
- Departamento de Farmacociências, Universidade Federal de Ciências da Saúde de Porto Alegre, RS, Brazil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Rio de Janeiro, RJ, Brazil
| | - C Logullo
- Laboratório de Química e Função de Proteínas e Peptídeos-CBB and Unidade de Experimentação Animal, Universidade Estadual do Norte Fluminense Darcy Ribeiro, Campos dos Goytacazes, RJ, Brazil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Rio de Janeiro, RJ, Brazil
| | - C Termignoni
- Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Departamento de Bioquímica, Universidade Federal do Rio Grande do Sul, Porto Alegre, Porto Alegre, Brazil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Rio de Janeiro, RJ, Brazil
| | - I DA Silva Vaz
- Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Faculdade de Veterinária, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Rio de Janeiro, RJ, Brazil
| |
Collapse
|
18
|
Huang K, Chen Y, Zhang R, Wu Y, Ma Y, Fang X, Shen S. Honokiol induces apoptosis and autophagy via the ROS/ERK1/2 signaling pathway in human osteosarcoma cells in vitro and in vivo. Cell Death Dis 2018; 9:157. [PMID: 29410403 PMCID: PMC5833587 DOI: 10.1038/s41419-017-0166-5] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 11/16/2017] [Accepted: 11/17/2017] [Indexed: 12/14/2022]
Abstract
Osteosarcoma is the most common primary malignant tumor of bone, the long-term survival of which has stagnated in the past several decades. In the present study, we investigated the anticancer effect of honokiol (HNK), an active component isolated and purified from the magnolia officinalis on human osteosarcoma cells. Our results showed that honokiol caused dose-dependent and time-dependent cell death in human osteosarcoma cells. The types of cell death induced by honokiol were primarily autophagy and apoptosis. Furthermore, honokiol induced G0/G1 phase arrest, elevated the levels of glucose-regulated protein (GRP)-78, an endoplasmic reticular stress (ERS)-associated protein, and increased the production of intracellular reactive oxygen species (ROS). In contrast, reducing production of intracellular ROS using N-acetylcysteine, a scavenger of ROS, concurrently suppressed honokiol-induced cellular apoptosis, autophagy, and cell cycle arrest. Consequently, honokiol stimulated phosphorylation of extracellular signal-regulated kinase (ERK)1/2. Furthermore, pretreatment of osteosarcoma cells with PD98059, an inhibitor of ERK1/2, inhibited honokiol-induced apoptosis and autophagy. Finally, honokiol suppressed tumor growth in the mouse xenograft model. Taken together, our results revealed that honokiol caused G0/G1 phase arrest, induced apoptosis, and autophagy via the ROS/ERK1/2 signaling pathway in human osteosarcoma cells. Honokiol is therefore a promising candidate for development of antitumor drugs targeting osteosarcoma.
Collapse
Affiliation(s)
- Kangmao Huang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, 3 East Qingchun Road, Hangzhou, 310016, China
| | - Yanyan Chen
- Department of Surgical Oncology, First Affiliated Hospital, Medical College of Zhejiang University, Hangzhou, 310003, China
| | - Rui Zhang
- Department of Neurosurgery, Children's hospital of Nanjing Medical University, Nanjing City, China
| | - Yizheng Wu
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, 3 East Qingchun Road, Hangzhou, 310016, China
| | - Yan Ma
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, 3 East Qingchun Road, Hangzhou, 310016, China
| | - Xiangqian Fang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, 3 East Qingchun Road, Hangzhou, 310016, China.
| | - Shuying Shen
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, 3 East Qingchun Road, Hangzhou, 310016, China.
| |
Collapse
|
19
|
Inhibition of PTEN Attenuates Endoplasmic Reticulum Stress and Apoptosis via Activation of PI3K/AKT Pathway in Alzheimer’s Disease. Neurochem Res 2017; 42:3052-3060. [PMID: 28819903 DOI: 10.1007/s11064-017-2338-1] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Revised: 06/14/2017] [Accepted: 06/20/2017] [Indexed: 12/14/2022]
|
20
|
Ittichaicharoen J, Apaijai N, Tanajak P, Sa-Nguanmoo P, Chattipakorn N, Chattipakorn SC. Impaired mitochondria and intracellular calcium transients in the salivary glands of obese rats. Appl Physiol Nutr Metab 2016; 42:420-429. [PMID: 28177730 DOI: 10.1139/apnm-2016-0545] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Long-term consumption of a high-fat diet (HFD) causes not only obese-insulin resistance, but is also associated with mitochondrial dysfunction in several organs. However, the effect of obese-insulin resistance on salivary glands has not been investigated. We hypothesized that obese-insulin resistance induced by HFD impaired salivary gland function by reducing salivation, increasing inflammation, and fibrosis, as well as impairing mitochondrial function and calcium transient signaling. Male Wistar rats (200-220 g) were fed either a ND or an HFD (n = 8/group) for 16 weeks. At the end of week 16, salivary flow rates, metabolic parameters, and plasma oxidative stress were determined. Rats were then sacrificed and submandibular glands were removed to determine inflammation, fibrosis, apoptosis, mitochondrial function and dynamics, and intracellular calcium transient signaling. Long-term consumption of an HFD caused obese-insulin resistance and increased oxidative stress, fibrosis, inflammation, and apoptosis in the salivary glands. In addition, impaired mitochondrial function, as indicated by increased mitochondrial reactive oxygen species, mitochondrial membrane depolarization, and mitochondrial swelling in salivary glands and impaired intracellular calcium regulation, as indicated by a reduced intracellular calcium transient rising rate, decay rates, and amplitude of salivary acinar cells, were observed in HFD-fed rats. However, salivary flow rate and level of aquaporin 5 protein were not different between both groups. Although HFD consumption did not affect salivation, it caused obese-insulin resistance, leading to pathophysiological alteration of salivary glands, including impaired intracellular calcium transients, increased oxidative stress and inflammation, and salivary mitochondrial dysfunction.
Collapse
Affiliation(s)
- Jitjiroj Ittichaicharoen
- a Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Nattayaporn Apaijai
- b Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Pongpan Tanajak
- c Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Department of Physiology, Faculty of Medicine, Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Piangkwan Sa-Nguanmoo
- c Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Department of Physiology, Faculty of Medicine, Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Nipon Chattipakorn
- c Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Department of Physiology, Faculty of Medicine, Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Siriporn C Chattipakorn
- a Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
| |
Collapse
|
21
|
de Souza JG, Morais KL, Anglés-Cano E, Boufleur P, de Mello ES, Maria DA, Origassa CST, Zampolli HDC, Câmara NOS, Berra CM, Bosch RV, Chudzinski-Tavassi AM. Promising pharmacological profile of a Kunitz-type inhibitor in murine renal cell carcinoma model. Oncotarget 2016; 7:62255-62266. [PMID: 27566592 PMCID: PMC5308724 DOI: 10.18632/oncotarget.11555] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 08/13/2016] [Indexed: 12/20/2022] Open
Abstract
Renal cell carcinoma (RCC), also called kidney cancer or renal adenocarcinoma, is highly resistant to current treatments. It has been previously reported that a Kunitz-type inhibitor domain-containing protein, isolated from the salivary glands of the Amblyomma cajennense tick, triggers apoptosis in murine renal adenocarcinoma cells (Renca) by inhibiting the proteasome and endoplasmic reticulum stress. Of note, Amblyomin-X is the corresponding recombinant protein identified in the cDNA library from A. cajennense salivary glands. Herein, using orthotopic kidney tumors in mice, we demonstrate that Amblyomin-X is able to drastically reduce the incidence of lung metastases by inducing cell cycle arrest and apoptosis. The in vitro assays show that Amblyomin-X is capable of reducing the proliferation rate of Renca cells, promoting cell cycle arrest, and down-regulating the expression of crucial proteins (cyclin D1, Ki67 and Pgp) involved in the aggressiveness and resistance of RCC. Regarding non-tumor cells (NIH3T3), Amblyomin-X produced minor effects in the cyclin D1 levels. Interestingly, observing the image assays, the fluorescence-labelled Amblyomin-X was indeed detected in the tumor stroma whereas in healthy animals it was rapidly metabolized and excreted. Taken the findings together, Amblyomin-X can be considered as a potential anti-RCC drug candidate.
Collapse
Affiliation(s)
- Jean Gabriel de Souza
- Biochemistry and Biophysics Laboratory, Butantan Institute, SP, Brazil
- Department of Biochemistry, Federal University of São Paulo, SP, Brazil
- CENTD- Center of Excellence in New Target Discovery, Butantan Institute, SP, Brazil
| | - Katia L.P. Morais
- Biochemistry and Biophysics Laboratory, Butantan Institute, SP, Brazil
- CENTD- Center of Excellence in New Target Discovery, Butantan Institute, SP, Brazil
| | - Eduardo Anglés-Cano
- INSERM UMR_S 1140-Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Pamela Boufleur
- Biochemistry and Biophysics Laboratory, Butantan Institute, SP, Brazil
- Department of Biochemistry, Federal University of São Paulo, SP, Brazil
| | | | | | - Clarice Silvia Taemi Origassa
- Laboratory of Transplantation Immunobiology, Department of Immunology, Institute of Biomedical Sciences IV, University of São Paulo, SP, Brazil
| | | | - Niels Olsen Saraiva Câmara
- Laboratory of Transplantation Immunobiology, Department of Immunology, Institute of Biomedical Sciences IV, University of São Paulo, SP, Brazil
- Nephrology Division, Federal University of São Paulo, SP, Brazil
| | | | | | - Ana Marisa Chudzinski-Tavassi
- Biochemistry and Biophysics Laboratory, Butantan Institute, SP, Brazil
- CENTD- Center of Excellence in New Target Discovery, Butantan Institute, SP, Brazil
| |
Collapse
|
22
|
Branco VG, Iqbal A, Alvarez-Flores MP, Sciani JM, de Andrade SA, Iwai LK, Serrano SMT, Chudzinski-Tavassi AM. Amblyomin-X having a Kunitz-type homologous domain, is a noncompetitive inhibitor of FXa and induces anticoagulation in vitro and in vivo. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2016; 1864:1428-35. [PMID: 27479486 DOI: 10.1016/j.bbapap.2016.07.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 07/22/2016] [Accepted: 07/28/2016] [Indexed: 10/21/2022]
Abstract
BACKGROUND Cancer has long been associated with thrombosis and many of the standard chemotherapeutics used to treat cancer are pro-thrombotic. Thus, the identification of novel selective anticancer drugs that also have antithrombotic properties is of enormous significance. Amblyomin-X is an anticancer protein derived from the salivary glands of the Amblyomma cajennense tick. METHODS In this work, we determined the inhibition profile of Amblyomin-X and its effect on activated partial thromboplastin time (aPTT) and prothrombin time (PT), using various approaches such as, kinetic analyses, amidolytic assays, SDS-PAGE, and mass spectrometry. RESULTS Amblyomin-X inhibited factor Xa, prothrombinase and tenase activities. It was hydrolyzed by trypsin and plasmin. MS/MS data of tryptic hydrolysate of Amblyomin-X suggested the presence of Cys(8)-Cys(59) and Cys(19)-Cys(42) but not Cys(34)-Cys(55) disulfide bond. Instead of Cys(34)-Cys(55), two noncanonical Cys(34)-Cys(74) and Cys(55)-Cys(74) disulfide bonds were identified. Furthermore, when Amblyomin-X (1mg/kg) injected in rabbits, it prolonged aPTT and PT. CONCLUSION Amblyomin-X is a noncompetitive inhibitor (Ki=3.9μM) of factor Xa. It is a substrate for plasmin and trypsin, but not for factor Xa and thrombin. The disulfide Cys(34)-Cys(55) bond probably scrambles with interchain seventh free cysteine residues (Cys(74)) of Amblyomin-X. The prolongation of PT and aPTT is reversible. GENERAL SIGNIFICANCE In term of anticoagulant property, this is structural and functional characterization of Amblyomin-X. All together, these results and previous findings suggest that Amblyomin-X has a potential to become an anticancer drug with antithrombotic property.
Collapse
Affiliation(s)
- Vania G Branco
- Laboratory of Biochemistry and Biophysics, Butantan Institute, Sao Paulo, SP, Brazil
| | - Asif Iqbal
- Laboratory of Biochemistry and Biophysics, Butantan Institute, Sao Paulo, SP, Brazil; Centre of Excellence for New Target Discovery (CENTD), Butantan Institute, São Paulo, SP, Brazil
| | - Miryam P Alvarez-Flores
- Laboratory of Biochemistry and Biophysics, Butantan Institute, Sao Paulo, SP, Brazil; Centre of Excellence for New Target Discovery (CENTD), Butantan Institute, São Paulo, SP, Brazil
| | - Juliana M Sciani
- Laboratory of Biochemistry and Biophysics, Butantan Institute, Sao Paulo, SP, Brazil
| | - Sonia A de Andrade
- Laboratory of Biochemistry and Biophysics, Butantan Institute, Sao Paulo, SP, Brazil
| | - Leo K Iwai
- Special Laboratory of Applied Toxinology, Butantan Institute, São Paulo, SP, Brazil
| | - Solange M T Serrano
- Special Laboratory of Applied Toxinology, Butantan Institute, São Paulo, SP, Brazil
| | - Ana M Chudzinski-Tavassi
- Laboratory of Biochemistry and Biophysics, Butantan Institute, Sao Paulo, SP, Brazil; Centre of Excellence for New Target Discovery (CENTD), Butantan Institute, São Paulo, SP, Brazil.
| |
Collapse
|
23
|
Guo L, Lv G, Qiu L, Yang H, Zhang L, Yu H, Zou M, Lin J. Insights into anticancer activity and mechanism of action of a ruthenium(II) complex in human esophageal squamous carcinoma EC109 cells. Eur J Pharmacol 2016; 786:60-71. [PMID: 27262377 DOI: 10.1016/j.ejphar.2016.05.042] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Revised: 05/30/2016] [Accepted: 05/30/2016] [Indexed: 12/09/2022]
Abstract
A ruthenium(II) complex [Ru(p-cymene)(NHC)Cl2] (NHC=1,3-bis(4-(tert-butyl)benzylimidazol-2-ylidene), referred to as L-4, has been designed and synthesized recently in order to look for new anticancer drugs with high efficacy and low side effects. The anticancer activity and mechanism of action of L-4 in human esophageal squamous carcinoma EC109 cells were systematically investigated. The results revealed that L-4 exerted strong inhibitory effect on the proliferation of EC109 cells, and it arrested EC109 cells at G2/M phase, accompanied with the up-regulation of p53 and p21 and the down-regulation of cyclin D1. The results also showed that the reactive oxygen species (ROS)-dependent apoptosis of EC109 can be induced by L-4 via inhibiting the activity of glutathione reductase (GR), decreasing the ratio of glutathione to oxidized glutathione (GSH/GSSG), and leading to the generation of reactive oxygen species. The mitochondria-mediated apoptosis of EC109 induced by L-4 was also observed from the increase of Bax/Bcl-2 ratio, overload of Ca(2+), disruption of mitochondrial membrane potential (MMP), redistribution of cytochrome c, and activation of caspase-3/-9. However, the effects of L-4 on the cell viability, GR activity, GSH/GSSG ratio, reactive oxygen species level, mitochondria dysfunction and apoptosis induction were remarkably attenuated by adding the reactive oxygen species scavenger, NAC. Therefore, it was concluded that L-4 can inhibit the proliferation of EC109 cells via blocking cell cycle progression and inducing reactive oxygen species-dependent and mitochondria-mediated apoptosis. These findings suggested that the ruthenium(II) complex might be a potential effective chemotherapeutic agent for human esophageal squamous carcinoma (ESCC) and worthy of further investigation.
Collapse
Affiliation(s)
- Liubin Guo
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Gaochao Lv
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Ling Qiu
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China.
| | - Hui Yang
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Li Zhang
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Huixin Yu
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Meifen Zou
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Jianguo Lin
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China.
| |
Collapse
|
24
|
Morais KLP, Pacheco MTF, Berra CM, Bosch RV, Sciani JM, Chammas R, de Freitas Saito R, Iqbal A, Chudzinski-Tavassi AM. Amblyomin-X induces ER stress, mitochondrial dysfunction, and caspase activation in human melanoma and pancreatic tumor cell. Mol Cell Biochem 2016; 415:119-31. [PMID: 27015684 PMCID: PMC4819916 DOI: 10.1007/s11010-016-2683-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 03/12/2016] [Indexed: 01/13/2023]
Abstract
During the last two decades, new insights into proteasome function and its role in several human diseases made it a potential therapeutic target. In this context, Amblyomin-X is a Kunitz-type FXa inhibitor similar to endogenous tissue factor pathway inhibitor (TFPI) and is a novel proteasome inhibitor. Herein, we have demonstrated Amblyomin-X cytotoxicity to different tumor cells lines such as pancreatic (Panc1, AsPC1BxPC3) and melanoma (SK-MEL-5 and SK-MEL-28). Of note, Amblyomin-X was not cytotoxic to normal human fibroblast cells. In addition, Amblyomin-X promoted accumulation of ER stress markers (GRP78 and GADD153) in sensitive (SK-MEL-28) and bortezomib-resistant (Mia-PaCa-2) tumor cells. The intracellular calcium concentration [Ca2+]i was slightly modulated in human tumor cells (SK-MEL-28 and Mia-PaCa-2) after 24 h of Amblyomin-X treatment. Furthermore, Amblyomin-X induced mitochondrial dysfunction, cytochrome-c release, PARP cleavage, and activation of caspase cascade in both human tumor (SK-MEL-28 and Mia-PaCa-2) cells. These investigations might help in further understanding of the antitumor properties of Amblyomin-X.
Collapse
Affiliation(s)
- Katia L P Morais
- Biochemistry and Biophysics Laboratory, Butantan Institute, Av. Vital Brazil, 1500, São Paulo, SP, 05503-900, Brazil.,Department of Biochemistry, Federal University of São Paulo, São Paulo, SP, Brazil
| | | | - Carolina Maria Berra
- Biochemistry Department, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Rosemary V Bosch
- Biochemistry and Biophysics Laboratory, Butantan Institute, Av. Vital Brazil, 1500, São Paulo, SP, 05503-900, Brazil
| | - Juliana Mozer Sciani
- Biochemistry and Biophysics Laboratory, Butantan Institute, Av. Vital Brazil, 1500, São Paulo, SP, 05503-900, Brazil
| | - Roger Chammas
- Experimental Oncology Medical Investigation Laboratory - LIM/24, University of São Paulo School of Medicine, São Paulo, SP, Brazil
| | - Renata de Freitas Saito
- Experimental Oncology Medical Investigation Laboratory - LIM/24, University of São Paulo School of Medicine, São Paulo, SP, Brazil
| | - Asif Iqbal
- Biochemistry and Biophysics Laboratory, Butantan Institute, Av. Vital Brazil, 1500, São Paulo, SP, 05503-900, Brazil
| | - Ana Marisa Chudzinski-Tavassi
- Biochemistry and Biophysics Laboratory, Butantan Institute, Av. Vital Brazil, 1500, São Paulo, SP, 05503-900, Brazil. .,Department of Biochemistry, Federal University of São Paulo, São Paulo, SP, Brazil.
| |
Collapse
|
25
|
Pacheco MTF, Morais KLP, Berra CM, Demasi M, Sciani JM, Branco VG, Bosch RV, Iqbal A, Chudzinski-Tavassi AM. Specific role of cytoplasmic dynein in the mechanism of action of an antitumor molecule, Amblyomin-X. Exp Cell Res 2015; 340:248-58. [PMID: 26748183 DOI: 10.1016/j.yexcr.2015.12.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Revised: 11/28/2015] [Accepted: 12/30/2015] [Indexed: 11/29/2022]
Abstract
The Kunitz-type recombinant protein, Amblyomin-X, is an antitumor recombinant molecule from a cDNA library prepared from the salivary glands of the tick Amblyomma cajennense. The primary target of this protein appears to be the proteasome. Amblyomin-X increased gene and protein expression of distinct subunits of the molecular motor dynein, which plays a key role in the intracellular transport. Herein, Amblyomin-X was specifically taken up by tumor cells through lipid-raft endocytic pathways, but not by fibroblasts. Moreover, dynein inhibitor, ciliobrevin A, decreased Amblyomin-X uptake by tumor cells. Furthermore, incubation of tumor cells with Amblyomin-X inhibited trypsin-like activity of the proteasome, which was restored upon pretreatment with ciliobrevin A. Only in tumor cells treated with Amblyomin-X, we identified proteins bounds to dynein that are related to aggresome formation, autophagy inhibition, and early and recycling endosome markers. In addition, Amblyomin-X was found to interact with dynein, increased Rab11A protein expression and Rab11A co-localization with the light-intermediate chain 2 (LIC2) of dynein. Thereby, the results provide new insights on the antitumor mechanism of Amblyomin-X and reveal an unsuspected role of cytoplasmic dynein in its uptake, intracellular trafficking and pro-apoptotic action.
Collapse
Affiliation(s)
- Mario T F Pacheco
- Biochemistry and Biophysics Laboratory, Butantan Institute, São Paulo, Brazil
| | - Kátia L P Morais
- Biochemistry and Biophysics Laboratory, Butantan Institute, São Paulo, Brazil; Department of Biochemistry, Federal University of São Paulo, São Paulo, Brazil
| | - Carolina M Berra
- Biochemistry Department, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Marilene Demasi
- Biochemistry and Biophysics Laboratory, Butantan Institute, São Paulo, Brazil
| | - Juliana M Sciani
- Biochemistry and Biophysics Laboratory, Butantan Institute, São Paulo, Brazil
| | - Vania G Branco
- Biochemistry and Biophysics Laboratory, Butantan Institute, São Paulo, Brazil
| | - Rosemary V Bosch
- Biochemistry and Biophysics Laboratory, Butantan Institute, São Paulo, Brazil
| | - Asif Iqbal
- Biochemistry and Biophysics Laboratory, Butantan Institute, São Paulo, Brazil
| | | |
Collapse
|
26
|
Chudzinski-Tavassi AM, Morais KLP, Pacheco MTF, Pasqualoto KFM, de Souza JG. Tick salivary gland as potential natural source for the discovery of promising antitumor drug candidates. Biomed Pharmacother 2015; 77:14-9. [PMID: 26796259 DOI: 10.1016/j.biopha.2015.11.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 11/03/2015] [Indexed: 12/31/2022] Open
Abstract
Nowadays, the relationship between cancer blood coagulation is well established. Regarding biodiversity and bioprospection, the tick biology has become quite attractive natural source for coagulation inhibitors, since its saliva has a very rich variety of bioactive molecules. For instance, a Kunitz-type FXa inhibitor, named Amblyomin-X, was found through transcriptome of the salivary gland of the Amblyomma cajennense. tick. This TFPI-like inhibitor, after obtained as recombinant protein, has presented anticoagulant, antigionenic, and antitumor properties. Although its effects on blood coagulation could be relevant for antitumor effect, Amblyomin-X acts by non-hemostatic mechanisms, such as proteasome inhibition and autophagy inhibition. Notably, cytotoxicity was not observed on non-tumor cells treated with this protein, suggesting some selectivity for tumor cells. Considering the current efforts in order to develop effective anticancer therapies, the findings presented in this review strongly suggest Amblyomin-X as a promising novel antitumor drug candidate.
Collapse
Affiliation(s)
| | - Katia L P Morais
- Biochemistry and Biophysics Laboratory, Butantan Institute, SP, Brazil; Department of Biochemistry, Federal University of São Paulo, SP, Brazil
| | | | | | - Jean Gabriel de Souza
- Biochemistry and Biophysics Laboratory, Butantan Institute, SP, Brazil; Department of Biochemistry, Federal University of São Paulo, SP, Brazil
| |
Collapse
|
27
|
Saito RDF, Tortelli TC, Jacomassi MD, Otake AH, Chammas R. Emerging targets for combination therapy in melanomas. FEBS Lett 2015; 589:3438-48. [PMID: 26450371 DOI: 10.1016/j.febslet.2015.09.022] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Revised: 09/25/2015] [Accepted: 09/25/2015] [Indexed: 12/21/2022]
Abstract
Cutaneous melanomas are often difficult to treat when diagnosed in advanced stages. Melanoma cells adapt to survive in extreme environmental conditions and are among the tumors with larger genomic instability. Here we discuss some intrinsic and extrinsic mechanisms of resistance of melanoma cells to both conventional and target therapies, such as autophagy, adaptation to endoplasmic reticulum stress, metabolic reprogramming, mechanisms of tumor repopulation and the role of extracellular vesicles in this later phenomenon. These biological processes are potentially targetable and thus provide a platform for research and discovery of new drugs for combination therapy to manage melanoma patient treatment.
Collapse
Affiliation(s)
- Renata de Freitas Saito
- Center for Translational Research in Oncology (LIM24), Dept. of Radiology and Oncology, Faculdade de Medicina da Universidade de São Paulo and Instituto do Câncer do Estado de São Paulo, Brazil
| | - Tharcísio Citrângulo Tortelli
- Center for Translational Research in Oncology (LIM24), Dept. of Radiology and Oncology, Faculdade de Medicina da Universidade de São Paulo and Instituto do Câncer do Estado de São Paulo, Brazil
| | - Mayara D'Auria Jacomassi
- Center for Translational Research in Oncology (LIM24), Dept. of Radiology and Oncology, Faculdade de Medicina da Universidade de São Paulo and Instituto do Câncer do Estado de São Paulo, Brazil
| | - Andréia Hanada Otake
- Center for Translational Research in Oncology (LIM24), Dept. of Radiology and Oncology, Faculdade de Medicina da Universidade de São Paulo and Instituto do Câncer do Estado de São Paulo, Brazil
| | - Roger Chammas
- Center for Translational Research in Oncology (LIM24), Dept. of Radiology and Oncology, Faculdade de Medicina da Universidade de São Paulo and Instituto do Câncer do Estado de São Paulo, Brazil.
| |
Collapse
|
28
|
Post-transcriptional control of Amblyomin-X on secretion of vascular endothelial growth factor and expression of adhesion molecules in endothelial cells. Toxicon 2015; 101:1-10. [PMID: 25912945 DOI: 10.1016/j.toxicon.2015.04.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Revised: 03/20/2015] [Accepted: 04/21/2015] [Indexed: 12/12/2022]
Abstract
Angiogenesis is a pivotal process of homeostasis and tissue repair, but it also favours neovascularisation syndromes and cancer nutrition. The chemical mediation of angiogenesis is complex, involving a balance between serine proteases and their inhibitors. We addressed the mechanisms of action of a Kunitz serine protease inhibitor (KPI) on spontaneous angiogenesis, using Amblyomin-X, a KPI designed from the cDNA library of the Amblyomma cajennense tick. Amblyomin-X treatment (10-1000 ng/10 μL; each 48 h; 3 times) reduced the number of vessels in the subcutaneous dorsal tissue of male Swiss mice, as measured by intravital microscopy, haematoxylin-eosin staining, and PECAM-1 immunofluorescence labeling. Incubation of Amblyomin-X with t-End endothelial cells, a murine endothelial microvascular lineage, did not alter cell proliferation, cell-cycle phases, necrosis and apoptosis, and the production of nitric oxide and prostaglandin E2. Nevertheless, Amblyomin-X treatment reduced t-End migration and adhesion to Matrigel(®), and inhibited the VEGF-A secretion and VCAM-1 and β3 integrin expressions by posttranscriptional pathways. Together, data herein outline novel posttranscriptional mechanisms of KPIs on endothelial cells during angiogenesis and point out the possible application of Amblyomin-X as a local inhibitor to undesired neovascularisation process.
Collapse
|
29
|
Yang W, Zou L, Huang C, Lei Y. Redox regulation of cancer metastasis: molecular signaling and therapeutic opportunities. Drug Dev Res 2015; 75:331-41. [PMID: 25160073 DOI: 10.1002/ddr.21216] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Cancer metastasis is the major cause of cancer-related mortality. Accumulated evidence has shown that high-metastasis potential cancer cells have more reactive oxygen species (ROS) accumulation compared with low-metastasis potential cancer cells. ROS can function as second messengers to regulate multiple cancer metastasis-related signaling pathways via reversible oxidative posttranslational modifications of cysteine in key redox-sensitive proteins, which leads to the structural and functional change of these proteins. Because ROS can promote cancer metastasis, therapeutic strategies aiming at inducing/reducing cellular ROS level or targeting redox sensors involved in metastasis hold great potential in developing new efficient approaches for anticancer therapy. In this review, we summarize recent findings on regulation of tumor metastasis by key redox sensors and describe the potential of targeting redox signaling pathways for cancer therapy.
Collapse
Affiliation(s)
- Wenyong Yang
- Department of Biochemistry and Molecular Biology, and Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, 400016, China; College of Life Sciences, Sichuan University, Chengdu, 610065, China; The State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | | | | | | |
Collapse
|
30
|
Chiu HW, Tseng YC, Hsu YH, Lin YF, Foo NP, Guo HR, Wang YJ. Arsenic trioxide induces programmed cell death through stimulation of ER stress and inhibition of the ubiquitin-proteasome system in human sarcoma cells. Cancer Lett 2015; 356:762-772. [PMID: 25449439 DOI: 10.1016/j.canlet.2014.10.025] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Accepted: 10/23/2014] [Indexed: 01/21/2023]
Abstract
Sarcoma is a rare form of cancer that differs from the much more common carcinomas because it occurs in a distinct type of tissue. Many patients of sarcoma have poor response to chemotherapy and an increased risk for local recurrence. Arsenic trioxide (ATO) is used to treat certain types of leukemia. Recently, data have revealed that ATO induces sarcoma cell death in several types of solid tumor cell lines. In the present study, we investigated whether ATO induces cancer cell death and elucidated the underlying anti-cancer mechanisms. Our results showed that ATO caused concentration- and time-dependent cell death in human osteosarcoma and fibrosarcoma cells. The types of cell death that were induced by ATO were primarily autophagy and apoptosis. Furthermore, ATO activated p38, JNK and AMPK and inhibited the Akt/mTOR signaling pathways. Specifically, we found that ATO induced endoplasmic reticulum (ER) stress and suppressed proteasome activation in two types of sarcoma cell lines. However, the level of proteasome inhibition in osteosarcoma cells was lower than in fibrosarcoma cells. Thus, we used combined treatment with ATO and a proteasome inhibitor to examine the antitumor activity in fibrosarcoma cells. The data indicated showed that the combination treatment of ATO and MG132 (a proteasome inhibitor) resulted in synergistic cytotoxicity. In a fibrosarcoma xenograft mouse model, the combined treatment significantly reduced tumor progression. Immunohistochemical studies revealed that combined treatment induced autophagy and apoptosis. In summary, our results suggest a potential clinical application of ATO in sarcoma therapy and that combined treatment with a proteasome inhibitor can increase the therapeutic efficacy.
Collapse
Affiliation(s)
- Hui-Wen Chiu
- Department of Environmental and Occupational Health, National Cheng Kung University, Tainan, Taiwan; Division of Nephrology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, Taiwan; Graduate Institute of Clinical Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yin-Chiu Tseng
- Department of Environmental and Occupational Health, National Cheng Kung University, Tainan, Taiwan
| | - Yung-Ho Hsu
- Division of Nephrology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, Taiwan
| | - Yuh-Feng Lin
- Division of Nephrology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, Taiwan; Graduate Institute of Clinical Medicine, Taipei Medical University, Taipei, Taiwan
| | - Ning-Ping Foo
- Department of Environmental and Occupational Health, National Cheng Kung University, Tainan, Taiwan; Department of Emergency Medicine, Chi-Mei Medical Center, Liouying, Tainan, Taiwan; Department of Emergency Medicine, China Medical University-An Nan Hospital, Tainan, Taiwan
| | - How-Ran Guo
- Department of Environmental and Occupational Health, National Cheng Kung University, Tainan, Taiwan; Department of Occupational and Environmental Medicine, National Cheng Kung University Hospital, Tainan, Taiwan.
| | - Ying-Jan Wang
- Department of Environmental and Occupational Health, National Cheng Kung University, Tainan, Taiwan; Department of Biomedical Informatics, Asia University, Taichung, Taiwan; Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan.
| |
Collapse
|
31
|
Soliman E, Henderson KL, Danell AS, Van Dross R. Arachidonoyl-ethanolamide activates endoplasmic reticulum stress-apoptosis in tumorigenic keratinocytes: Role of cyclooxygenase-2 and novel J-series prostamides. Mol Carcinog 2015; 55:117-30. [PMID: 25557612 DOI: 10.1002/mc.22257] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Revised: 11/03/2015] [Accepted: 11/05/2015] [Indexed: 11/12/2022]
Abstract
Non-melanoma skin cancer and other epithelial tumors overexpress cyclooxygenase-2 (COX-2), differentiating them from normal cells. COX-2 metabolizes arachidonic acid to prostaglandins including, the J-series prostaglandins, which induce apoptosis by mechanisms including endoplasmic reticulum (ER) stress. Arachidonoyl-ethanolamide (AEA) is a cannabinoid that causes apoptosis in diverse tumor types. Previous studies from our group demonstrated that AEA was metabolized by COX-2 to J-series prostaglandins. Thus, the current study examines the role of COX-2, J-series prostaglandins, and ER stress in AEA-induced apoptosis. In tumorigenic keratinocytes that overexpress COX-2, AEA activated the PKR-like ER kinase (PERK), inositol requiring kinase-1 (IRE1), and activating transcription factor-6 (ATF6) ER stress pathways and the ER stress apoptosis-associated proteins, C/EBP homologous protein-10 (CHOP10), caspase-12, and caspase-3. Using an ER stress inhibitor, it was determined that ER stress was required for AEA-induced apoptosis. To evaluate the role of COX-2 in ER stress-apoptosis, HaCaT keratinocytes with low endogenous COX-2 expression were transfected with COX-2 cDNA or an empty vector and AEA-induced ER stress-apoptosis occurred only in the presence of COX-2. Moreover, LC-MS analysis showed that the novel prostaglandins, 15-deoxyΔ(12,14) PGJ2 -EA and Δ(12) PGJ2 /PGJ2-EA, were synthesized from AEA. These findings suggest that AEA will be selectively toxic in tumor cells that overexpress COX-2 due to the metabolism of AEA by COX-2 to J-series prostaglandin-ethanolamides (prostamides). Hence, AEA may be an ideal topical agent for the elimination of malignancies that overexpress COX-2.
Collapse
Affiliation(s)
- Eman Soliman
- Brody School of Medicine, Pharmacology and Toxicology, East Carolina University, Greenville, NC
| | - Kate L Henderson
- Department of Chemistry, East Carolina University, Greenville, NC
| | - Allison S Danell
- Department of Chemistry, East Carolina University, Greenville, NC
| | - Rukiyah Van Dross
- Brody School of Medicine, Pharmacology and Toxicology, East Carolina University, Greenville, NC
| |
Collapse
|
32
|
de Queiroz EAIF, Akamine EH, de Carvalho MHC, Sampaio SC, Fortes ZB. Metformin reduces the Walker-256 tumor development in obese-MSG rats via AMPK and FOXO3a. Life Sci 2014; 121:78-87. [PMID: 25497710 DOI: 10.1016/j.lfs.2014.11.028] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Revised: 11/11/2014] [Accepted: 11/24/2014] [Indexed: 11/24/2022]
Abstract
AIMS Studies have associated obesity with a wide variety of cancers. Metformin, an anti-diabetic drug, has recently received attention as a potentially useful therapeutic agent for treating cancer. Therefore, the objective of this study was to analyze the mechanisms involved in the increase in tumor development and the reduction of it by metformin in obesity using an experimental breast tumor model. MATERIAL AND METHODS Newborn male Wistar rats were subcutaneously injected with 400mg/kg monosodium glutamate (MSG) (obese) or saline (control) at 2, 3, 4, 5 and 6 days of age. After 16 weeks, 1 × 10(7) Walker-256 tumor cells were subcutaneously injected in the right flank of the rats and concomitantly the treatment with metformin 300 mg/kg/15 days, via gavage, started. The rats were divided into 4 groups: control tumor (CT), control tumor metformin (CTM), obese-MSG tumor (OT) and obese-MSG tumor metformin (OTM). On the 18th week the tumor development and metformin effect were analyzed. KEY FINDINGS Tumor development was higher in OT rats compared with CT rats. Activation of insulin-IR-ERK1/2 pathway and an anti-apoptotic effect might be the mechanisms involved in the higher development of tumor in obesity. The effect of metformin reducing the tumor development in obese rats might involve increased mRNA expression of pRb and p27, increased activity of AMPK and FOXO3a and decreased expression of p-ERK1/2 (Thr202/Tyr204) in Walker-256 tumor. SIGNIFICANCE Our data allow us to suggest that metformin, reducing the stimulatory effect of obesity on tumor development, has a potential role in the management of cancers.
Collapse
Affiliation(s)
- Eveline A I F de Queiroz
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil; Department of Physiological Sciences, State University of Londrina, Londrina, Brazil.
| | - Eliana H Akamine
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Maria Helena C de Carvalho
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Sandra C Sampaio
- Laboratory of Pathophysiology, Butantan Institute, São Paulo, Brazil
| | - Zuleica B Fortes
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil.
| |
Collapse
|
33
|
Chantong B, Kratschmar DV, Lister A, Odermatt A. Inhibition of metabotropic glutamate receptor 5 induces cellular stress through pertussis toxin-sensitive Gi-proteins in murine BV-2 microglia cells. J Neuroinflammation 2014; 11:190. [PMID: 25407356 PMCID: PMC4240888 DOI: 10.1186/s12974-014-0190-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Accepted: 10/30/2014] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND Activation of metabotropic glutamate receptor 5 (mGluR5) by (RS)-2-chloro-5-hydroxyphenylglycine (CHPG) was shown to suppress microglia activation and decrease the release of associated pro-inflammatory mediators. In contrast, the consequences of mGluR5 inhibition are less well understood. Here, we used BV-2 cells, retaining key characteristics of primary mouse microglia, to examine whether mGluR5 inhibition by 2-methyl-6-(phenylethynyl)-pyridine (MPEP) enhances cellular stress and production of inflammatory mediators. METHODS BV-2 cells were treated with MPEP, followed by determination of cellular stress using fluorescent dyes and high-content imaging. The expression of inflammatory mediators, endoplasmic reticulum (ER)-stress markers and phosphorylated AMPKα was analyzed by quantitative PCR, ELISA and Western blotting. Additionally, phospholipase C (PLC) activity, cellular ATP content and changes in intracellular free Ca(2+) ([Ca(2+)]i) were measured using luminescence and fluorescence assays. RESULTS Treatment of BV-2 microglia with 100 μM MPEP increased intracellular reactive oxygen species (ROS), mitochondrial superoxide, mitochondrial mass as well as inducible nitric oxide synthase (iNOS) and IL-6 expression. Furthermore, MPEP reduced cellular ATP and induced AMPKα phosphorylation and the expression of the ER-stress markers CHOP, GRP78 and GRP96. The MPEP-dependent effects were preceded by a rapid concentration-dependent elevation of [Ca(2+)]i, following Ca(2+) release from the ER, mainly via inositol triphosphate-induced receptors (IP3R). The MPEP-induced ER-stress could be blocked by pretreatment with the chemical chaperone 4-phenylbutyrate and the Ca(2+) chelator BAPTA-AM. Pretreatment with the AMPK agonist AICAR partially abolished, whilst the inhibitor compound C potentiated, the MPEP-dependent ER-stress. Importantly, the PLC inhibitor U-73122 and the Gi-protein inhibitor pertussis toxin (PTX) blocked the MPEP-induced increase in [Ca(2+)]i. Moreover, pretreatment of microglia with AICAR, BAPTA-AM, U-73122 and PTX prevented the MPEP-induced generation of oxidative stress and inflammatory mediators, further supporting a role for Gi-protein-mediated activation of PLC. CONCLUSIONS The results emphasize the potential pathophysiological role of mGluR5 antagonism in mediating oxidative stress, ER-stress and inflammation through a Ca(2+)-dependent pathway in microglia. The induction of cellular stress and inflammatory mediators involves PTX-sensitive Gi-proteins and subsequent activation of PLC, IP3R and Ca(2+) release from the ER.
Collapse
Affiliation(s)
- Boonrat Chantong
- Current address: Department of Preclinical Science and Applied Animal Science, Faculty of Veterinary Science, Mahidol University, Phutthamonthon, Nakhonpathom, Thailand.
| | - Denise V Kratschmar
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland.
| | - Adam Lister
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland.
| | - Alex Odermatt
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland.
| |
Collapse
|
34
|
Lee J, Hong SW, Park SE, Rhee EJ, Park CY, Oh KW, Park SW, Lee WY. Exendin-4 attenuates endoplasmic reticulum stress through a SIRT1-dependent mechanism. Cell Stress Chaperones 2014; 19:649-56. [PMID: 24446069 PMCID: PMC4147070 DOI: 10.1007/s12192-013-0490-3] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Revised: 12/18/2013] [Accepted: 12/21/2013] [Indexed: 02/08/2023] Open
Abstract
Accumulation of excess hepatic lipids contributes to insulin resistance and liver disease associated with endoplasmic reticulum (ER) stress. Exendin-4 is an agonist of the glucagon-like peptide 1 receptor and plays a role in improving insulin resistance and liver disease by increasing silent mating type information regulation 2 homolog (SIRT) 1. However, the effects and mechanism of action of exendin-4 on responses to palmitic acid (PA)-induced ER stress in hepatocytes have not been clearly defined. We investigated whether exendin-4 attenuates PA-induced ER stress via SIRT1 in HepG2 cells. PA treatment induced increased expression of PRKR-like endoplasmic reticulum kinase, inositol-requiring kinase 1α (IRE1α), activating transcription factor 6 (ATF6), and C/EBP homologous protein (CHOP) mRNA. Exendin-4 decreased the expression of P-IRE1α, ATF6, X-box binding protein-1 and CHOP, and increased the expression of SERCA2b. A significant decrease in the hepatic expression of PUMA, BAX, cytochrome c, and cleaved caspase-3 were observed in hepatocytes treated with exendin-4. The TUNEL assay consistently showed that exendin-4 reversed hepatocyte apoptosis induced by treatment with PA. Inhibition of SIRT1 by nicotinamide and siRNA significantly increased the expression of ER stress marker genes in cells treated with both PA and exendin-4. In conclusion, increased SIRT1 by exendin-4 attenuates PA-induced ER stress and mitochondrial dysfunction in hepatocytes.
Collapse
Affiliation(s)
- Jinmi Lee
- />Institute of Medical Research, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, 110-746 South Korea
| | - Seok-Woo Hong
- />Institute of Medical Research, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, 110-746 South Korea
| | - Se Eun Park
- />Division of Endocrinology and Metabolism, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, 110-746 South Korea
| | - Eun-Jung Rhee
- />Division of Endocrinology and Metabolism, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, 110-746 South Korea
| | - Cheol-Young Park
- />Division of Endocrinology and Metabolism, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, 110-746 South Korea
| | - Ki-Won Oh
- />Division of Endocrinology and Metabolism, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, 110-746 South Korea
| | - Sung-Woo Park
- />Division of Endocrinology and Metabolism, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, 110-746 South Korea
| | - Won-Young Lee
- />Division of Endocrinology and Metabolism, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, 110-746 South Korea
- />Division of Endocrinology and Metabolism, Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, # 108 Pyung-Dong, Seoul, Jongro-Ku 110-746 South Korea
| |
Collapse
|
35
|
Moon J, Koh SS, Malilas W, Cho IR, Kaewpiboon C, Kaowinn S, Lee K, Jhun BH, Choi YW, Chung YH. Acetylshikonin induces apoptosis of hepatitis B virus X protein-expressing human hepatocellular carcinoma cells via endoplasmic reticulum stress. Eur J Pharmacol 2014; 735:132-40. [PMID: 24769509 DOI: 10.1016/j.ejphar.2014.04.021] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2013] [Revised: 04/05/2014] [Accepted: 04/09/2014] [Indexed: 11/15/2022]
Abstract
Since it has been known that shikonin derived from a medicinal plant possesses anti-cancer activity, we wonder whether acetylshikonin (ASK), a derivate of shikonin, can be used to treat hepatocellular carcinoma cells expressing hepatitis B virus X protein (HBX), an oncoprotein from hepatitis B virus. When ASK was added to Hep3B cells stably expressing HBX, it induced apoptosis in a dose-dependent manner. ASK induced upregulation and export of Nur77 to the cytoplasm and activation of JNK. Likewise, suppression of Nur77 and JNK inactivation protected the cells from ASK-induced apoptosis, indicating that Nur77 upregulation and JNK activation were required for ASK-mediated apoptosis. Furthermore, ASK increased the expression of Bip and ubiquitination levels of cellular proteins, features of endoplasmic reticulum (ER) stress, via the production of reactive oxygen species in a dose-dependent manner. Suppression of reactive oxygen species with N-acetylcysteine reduced levels of Bip protein and ubiquitination levels of cellular proteins during ASK treatment, leading to protection of cells from apoptosis. Cycloheximide treatment reduced ASK-induced ER stress, suggesting that protein synthesis is involved in ASK-induced ER stress. Moreover, we showed using salubrinal, an ER stress inhibitor that reactive oxygen species production, JNK activation, and Nur77 upregulation and its translocation to cytoplasm are necessary for ER-induced stress. Interestingly, we found that JNK inactivation suppresses ASK-induced ER stress, whereas Nur77 siRNA treatment does not, indicating that JNK is required for ASK-induced ER stress. Accordingly, we report that ASK induces ER stress, which is prerequisite for apoptosis of HBX-expressing hepatocellular carcinoma cells.
Collapse
Affiliation(s)
- Jeong Moon
- BK21+, Department of Cogno-Mechatronics Engineering, Pusan National University, Busan 609-735, Republic of Korea
| | - Sang Seok Koh
- Department of Biological Sciences, Dong-A University, Busan 604-020, Republic of Korea
| | - Waraporn Malilas
- BK21+, Department of Cogno-Mechatronics Engineering, Pusan National University, Busan 609-735, Republic of Korea
| | - Il-Rae Cho
- BK21+, Department of Cogno-Mechatronics Engineering, Pusan National University, Busan 609-735, Republic of Korea
| | - Chutima Kaewpiboon
- Program in Biotechnology, Faculty of Science, Chulalongkorn University, Phayathai Road, Bangkok 10330, Thailand
| | - Sirichat Kaowinn
- BK21+, Department of Cogno-Mechatronics Engineering, Pusan National University, Busan 609-735, Republic of Korea
| | - Keesook Lee
- Hormone Research Center, School of Biological Science and Technology, Chonnam National University, 500-757, Republic of Korea
| | - Byung Hak Jhun
- Department of Applied Nanoscience, Pusan National University, Busan 609-735, Republic of Korea
| | - Young Whan Choi
- Department of Horticultural Bioscience, Pusan National University, Miryang 627-706, Republic of Korea
| | - Young-Hwa Chung
- BK21+, Department of Cogno-Mechatronics Engineering, Pusan National University, Busan 609-735, Republic of Korea.
| |
Collapse
|
36
|
Morais KLP, Pasqualoto KFM, Pacheco MTF, Berra CM, Alvarez-Flores MP, Chudzinski-Tavassi AM. Rational development of a novel TFPI-like inhibitor fromAmblyomma cajennensetick. TOXIN REV 2013. [DOI: 10.3109/15569543.2013.845217] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|