1
|
García-Montero C, Fraile-Martinez O, Cobo-Prieto D, De Leon-Oliva D, Boaru DL, De Castro-Martinez P, Pekarek L, Gragera R, Hernández-Fernández M, Guijarro LG, Toledo-Lobo MDV, López-González L, Díaz-Pedrero R, Monserrat J, Álvarez-Mon M, Saez MA, Ortega MA. Abnormal Histopathological Expression of Klotho, Ferroptosis, and Circadian Clock Regulators in Pancreatic Ductal Adenocarcinoma: Prognostic Implications and Correlation Analyses. Biomolecules 2024; 14:947. [PMID: 39199335 PMCID: PMC11353028 DOI: 10.3390/biom14080947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 07/27/2024] [Accepted: 07/29/2024] [Indexed: 09/01/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an extremely lethal tumor with increasing incidence, presenting numerous clinical challenges. The histopathological examination of novel, unexplored biomarkers offers a promising avenue for research, with significant translational potential for improving patient outcomes. In this study, we evaluated the prognostic significance of ferroptosis markers (TFRC, ALOX-5, ACSL-4, and GPX-4), circadian clock regulators (CLOCK, BMAL1, PER1, PER2), and KLOTHO in a retrospective cohort of 41 patients deceased by PDAC. Immunohistochemical techniques (IHC) and multiple statistical analyses (Kaplan-Meier curves, correlograms, and multinomial linear regression models) were performed. Our findings reveal that ferroptosis markers are directly associated with PDAC mortality, while circadian regulators and KLOTHO are inversely associated. Notably, TFRC emerged as the strongest risk marker associated with mortality (HR = 35.905), whereas CLOCK was identified as the most significant protective marker (HR = 0.01832). Correlation analyses indicate that ferroptosis markers are positively correlated with each other, as are circadian regulators, which also positively correlate with KLOTHO expression. In contrast, KLOTHO and circadian regulators exhibit inverse correlations with ferroptosis markers. Among the clinical variables examined, only the presence of chronic pathologies showed an association with the expression patterns of several proteins studied. These findings underscore the complexity of PDAC pathogenesis and highlight the need for further research into the specific molecular mechanisms driving disease progression.
Collapse
Affiliation(s)
- Cielo García-Montero
- Department of Medicine and Medical Specialities (CIBEREHD), Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (C.G.-M.); (O.F.-M.); (D.C.-P.); (D.D.L.-O.); (D.L.B.); (P.D.C.-M.); (L.P.); (R.G.); (J.M.); (M.Á.-M.); (M.A.S.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (L.G.G.); (M.D.V.T.-L.); (L.L.-G.); (R.D.-P.)
| | - Oscar Fraile-Martinez
- Department of Medicine and Medical Specialities (CIBEREHD), Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (C.G.-M.); (O.F.-M.); (D.C.-P.); (D.D.L.-O.); (D.L.B.); (P.D.C.-M.); (L.P.); (R.G.); (J.M.); (M.Á.-M.); (M.A.S.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (L.G.G.); (M.D.V.T.-L.); (L.L.-G.); (R.D.-P.)
| | - David Cobo-Prieto
- Department of Medicine and Medical Specialities (CIBEREHD), Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (C.G.-M.); (O.F.-M.); (D.C.-P.); (D.D.L.-O.); (D.L.B.); (P.D.C.-M.); (L.P.); (R.G.); (J.M.); (M.Á.-M.); (M.A.S.)
- Immune System Diseases-Rheumatology Service, Central University Hospital of Defence-UAH Madrid, 28801 Alcala de Henares, Spain
| | - Diego De Leon-Oliva
- Department of Medicine and Medical Specialities (CIBEREHD), Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (C.G.-M.); (O.F.-M.); (D.C.-P.); (D.D.L.-O.); (D.L.B.); (P.D.C.-M.); (L.P.); (R.G.); (J.M.); (M.Á.-M.); (M.A.S.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (L.G.G.); (M.D.V.T.-L.); (L.L.-G.); (R.D.-P.)
| | - Diego Liviu Boaru
- Department of Medicine and Medical Specialities (CIBEREHD), Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (C.G.-M.); (O.F.-M.); (D.C.-P.); (D.D.L.-O.); (D.L.B.); (P.D.C.-M.); (L.P.); (R.G.); (J.M.); (M.Á.-M.); (M.A.S.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (L.G.G.); (M.D.V.T.-L.); (L.L.-G.); (R.D.-P.)
| | - Patricia De Castro-Martinez
- Department of Medicine and Medical Specialities (CIBEREHD), Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (C.G.-M.); (O.F.-M.); (D.C.-P.); (D.D.L.-O.); (D.L.B.); (P.D.C.-M.); (L.P.); (R.G.); (J.M.); (M.Á.-M.); (M.A.S.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (L.G.G.); (M.D.V.T.-L.); (L.L.-G.); (R.D.-P.)
| | - Leonel Pekarek
- Department of Medicine and Medical Specialities (CIBEREHD), Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (C.G.-M.); (O.F.-M.); (D.C.-P.); (D.D.L.-O.); (D.L.B.); (P.D.C.-M.); (L.P.); (R.G.); (J.M.); (M.Á.-M.); (M.A.S.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (L.G.G.); (M.D.V.T.-L.); (L.L.-G.); (R.D.-P.)
| | - Raquel Gragera
- Department of Medicine and Medical Specialities (CIBEREHD), Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (C.G.-M.); (O.F.-M.); (D.C.-P.); (D.D.L.-O.); (D.L.B.); (P.D.C.-M.); (L.P.); (R.G.); (J.M.); (M.Á.-M.); (M.A.S.)
| | - Mauricio Hernández-Fernández
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain;
| | - Luis G. Guijarro
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (L.G.G.); (M.D.V.T.-L.); (L.L.-G.); (R.D.-P.)
- Unit of Biochemistry and Molecular Biology, Department of System Biology (CIBEREHD), University of Alcalá, 28801 Alcala de Henares, Spain
| | - María Del Val Toledo-Lobo
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (L.G.G.); (M.D.V.T.-L.); (L.L.-G.); (R.D.-P.)
- Department of Biomedicine and Biotechnology, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain
| | - Laura López-González
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (L.G.G.); (M.D.V.T.-L.); (L.L.-G.); (R.D.-P.)
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain;
| | - Raul Díaz-Pedrero
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (L.G.G.); (M.D.V.T.-L.); (L.L.-G.); (R.D.-P.)
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain;
| | - Jorge Monserrat
- Department of Medicine and Medical Specialities (CIBEREHD), Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (C.G.-M.); (O.F.-M.); (D.C.-P.); (D.D.L.-O.); (D.L.B.); (P.D.C.-M.); (L.P.); (R.G.); (J.M.); (M.Á.-M.); (M.A.S.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (L.G.G.); (M.D.V.T.-L.); (L.L.-G.); (R.D.-P.)
| | - Melchor Álvarez-Mon
- Department of Medicine and Medical Specialities (CIBEREHD), Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (C.G.-M.); (O.F.-M.); (D.C.-P.); (D.D.L.-O.); (D.L.B.); (P.D.C.-M.); (L.P.); (R.G.); (J.M.); (M.Á.-M.); (M.A.S.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (L.G.G.); (M.D.V.T.-L.); (L.L.-G.); (R.D.-P.)
- Immune System Diseases-Rheumatology and Internal Medicine Service, University Hospital Prince of Asturias, Networking Research Center on for Liver and Digestive Diseases (CIBEREHD), 28806 Alcala de Henares, Spain
| | - Miguel A. Saez
- Department of Medicine and Medical Specialities (CIBEREHD), Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (C.G.-M.); (O.F.-M.); (D.C.-P.); (D.D.L.-O.); (D.L.B.); (P.D.C.-M.); (L.P.); (R.G.); (J.M.); (M.Á.-M.); (M.A.S.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (L.G.G.); (M.D.V.T.-L.); (L.L.-G.); (R.D.-P.)
- Pathological Anatomy Service, University Hospital Gómez-Ulla, 28806 Alcala de Henares, Spain
| | - Miguel A. Ortega
- Department of Medicine and Medical Specialities (CIBEREHD), Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (C.G.-M.); (O.F.-M.); (D.C.-P.); (D.D.L.-O.); (D.L.B.); (P.D.C.-M.); (L.P.); (R.G.); (J.M.); (M.Á.-M.); (M.A.S.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (L.G.G.); (M.D.V.T.-L.); (L.L.-G.); (R.D.-P.)
| |
Collapse
|
2
|
Ferdous KU, Tesfay MZ, Cios A, Shelton RS, Hartupee C, Urbaniak A, Chamcheu JC, Mavros MN, Giorgakis E, Mustafa B, Simoes CC, Miousse IR, Basnakian AG, Moaven O, Post SR, Cannon MJ, Kelly T, Nagalo BM. Enhancing Neoadjuvant Virotherapy's Effectiveness by Targeting Stroma to Improve Resectability in Pancreatic Cancer. Biomedicines 2024; 12:1596. [PMID: 39062169 PMCID: PMC11275208 DOI: 10.3390/biomedicines12071596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 07/15/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024] Open
Abstract
About one-fourth of patients with pancreatic ductal adenocarcinoma (PDAC) are categorized as borderline resectable (BR) or locally advanced (LA). Chemotherapy and radiation therapy have not yielded the anticipated outcomes in curing patients with BR/LA PDAC. The surgical resection of these tumors presents challenges owing to the unpredictability of the resection margin, involvement of vasculature with the tumor, the likelihood of occult metastasis, a higher ratio of positive lymph nodes, and the relatively larger size of tumor nodules. Oncolytic virotherapy has shown promising activity in preclinical PDAC models. Unfortunately, the desmoplastic stroma within the PDAC tumor microenvironment establishes a barrier, hindering the infiltration of oncolytic viruses and various therapeutic drugs-such as antibodies, adoptive cell therapy agents, and chemotherapeutic agents-in reaching the tumor site. Recently, a growing emphasis has been placed on targeting major acellular components of tumor stroma, such as hyaluronic acid and collagen, to enhance drug penetration. Oncolytic viruses can be engineered to express proteolytic enzymes that cleave hyaluronic acid and collagen into smaller polypeptides, thereby softening the desmoplastic stroma, ultimately leading to increased viral distribution along with increased oncolysis and subsequent tumor size regression. This approach may offer new possibilities to improve the resectability of patients diagnosed with BR and LA PDAC.
Collapse
Affiliation(s)
- Khandoker Usran Ferdous
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (K.U.F.); (M.Z.T.); (A.C.); (C.C.S.); (S.R.P.); (T.K.)
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (M.N.M.); (M.J.C.)
| | - Mulu Z. Tesfay
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (K.U.F.); (M.Z.T.); (A.C.); (C.C.S.); (S.R.P.); (T.K.)
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (M.N.M.); (M.J.C.)
| | - Aleksandra Cios
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (K.U.F.); (M.Z.T.); (A.C.); (C.C.S.); (S.R.P.); (T.K.)
| | - Randal S. Shelton
- College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA;
| | - Conner Hartupee
- Division of Surgical Oncology, Department of Surgery, Louisiana State University (LSU) Health, New Orleans, LA 70112, USA; (C.H.); (O.M.)
| | - Alicja Urbaniak
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (A.U.); (I.R.M.)
| | - Jean Christopher Chamcheu
- Department of Biological Sciences and Chemistry, Southern University and A&M College, Baton Rouge, LA 70813, USA;
- Division of Biotechnology and Molecular Medicine, Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Michail N. Mavros
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (M.N.M.); (M.J.C.)
- College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA;
- Department of Surgery, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA;
| | - Emmanouil Giorgakis
- Department of Surgery, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA;
| | - Bahaa Mustafa
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA;
| | - Camila C. Simoes
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (K.U.F.); (M.Z.T.); (A.C.); (C.C.S.); (S.R.P.); (T.K.)
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (M.N.M.); (M.J.C.)
| | - Isabelle R. Miousse
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (A.U.); (I.R.M.)
| | - Alexei G. Basnakian
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA;
- Central Arkansas Veterans Healthcare System, John L. McClellan Memorial VA Hospital, Little Rock, AR 72205, USA
| | - Omeed Moaven
- Division of Surgical Oncology, Department of Surgery, Louisiana State University (LSU) Health, New Orleans, LA 70112, USA; (C.H.); (O.M.)
- Department of Interdisciplinary Oncology, Louisiana Cancer Research Center, Louisiana State University (LSU) Health, New Orleans, LA 70112, USA
| | - Steven R. Post
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (K.U.F.); (M.Z.T.); (A.C.); (C.C.S.); (S.R.P.); (T.K.)
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (M.N.M.); (M.J.C.)
| | - Martin J. Cannon
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (M.N.M.); (M.J.C.)
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Thomas Kelly
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (K.U.F.); (M.Z.T.); (A.C.); (C.C.S.); (S.R.P.); (T.K.)
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (M.N.M.); (M.J.C.)
| | - Bolni Marius Nagalo
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (K.U.F.); (M.Z.T.); (A.C.); (C.C.S.); (S.R.P.); (T.K.)
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (M.N.M.); (M.J.C.)
| |
Collapse
|
3
|
Amoah AS, Pestov NB, Korneenko TV, Prokhorenko IA, Kurakin GF, Barlev NA. Lipoxygenases at the Intersection of Infection and Carcinogenesis. Int J Mol Sci 2024; 25:3961. [PMID: 38612771 PMCID: PMC11011848 DOI: 10.3390/ijms25073961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 03/08/2024] [Accepted: 03/28/2024] [Indexed: 04/14/2024] Open
Abstract
The persisting presence of opportunistic pathogens like Pseudomonas aeruginosa poses a significant threat to many immunocompromised cancer patients with pulmonary infections. This review highlights the complexity of interactions in the host's defensive eicosanoid signaling network and its hijacking by pathogenic bacteria to their own advantage. Human lipoxygenases (ALOXs) and their mouse counterparts are integral elements of the innate immune system, mostly operating in the pro-inflammatory mode. Taking into account the indispensable role of inflammation in carcinogenesis, lipoxygenases have counteracting roles in this process. In addition to describing the structure-function of lipoxygenases in this review, we discuss their roles in such critical processes as cancer cell signaling, metastases, death of cancer and immune cells through ferroptosis, as well as the roles of ALOXs in carcinogenesis promoted by pathogenic infections. Finally, we discuss perspectives of novel oncotherapeutic approaches to harness lipoxygenase signaling in tumors.
Collapse
Affiliation(s)
- Abdul-Saleem Amoah
- Institute of Biomedical Chemistry, Moscow 119121, Russia; (A.-S.A.); (N.A.B.)
- Laboratory of Molecular Oncology, Phystech School of Biological and Medical Physics, Moscow Institute of Physics and Technology, Dolgoprudny 141701, Russia
| | - Nikolay B. Pestov
- Institute of Biomedical Chemistry, Moscow 119121, Russia; (A.-S.A.); (N.A.B.)
- Group of Cross-Linking Enzymes, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow 117997, Russia; (T.V.K.); (I.A.P.)
- Laboratory of Tick-Borne Encephalitis and Other Viral Encephalitides, Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products, Moscow 108819, Russia
- Vavilov Institute of General Genetics, Moscow 119991, Russia
| | - Tatyana V. Korneenko
- Group of Cross-Linking Enzymes, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow 117997, Russia; (T.V.K.); (I.A.P.)
| | - Igor A. Prokhorenko
- Group of Cross-Linking Enzymes, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow 117997, Russia; (T.V.K.); (I.A.P.)
| | - Georgy F. Kurakin
- Department of Biochemistry, Pirogov Russian National Research Medical University, Moscow 117513, Russia;
| | - Nickolai A. Barlev
- Institute of Biomedical Chemistry, Moscow 119121, Russia; (A.-S.A.); (N.A.B.)
- Laboratory of Tick-Borne Encephalitis and Other Viral Encephalitides, Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products, Moscow 108819, Russia
| |
Collapse
|
4
|
Li K, Deng Z, Lei C, Ding X, Li J, Wang C. The Role of Oxidative Stress in Tumorigenesis and Progression. Cells 2024; 13:441. [PMID: 38474405 PMCID: PMC10931308 DOI: 10.3390/cells13050441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 02/20/2024] [Accepted: 02/29/2024] [Indexed: 03/14/2024] Open
Abstract
Oxidative stress refers to the imbalance between the production of reactive oxygen species (ROS) and the endogenous antioxidant defense system. Its involvement in cell senescence, apoptosis, and series diseases has been demonstrated. Advances in carcinogenic research have revealed oxidative stress as a pivotal pathophysiological pathway in tumorigenesis and to be involved in lung cancer, glioma, hepatocellular carcinoma, leukemia, and so on. This review combs the effects of oxidative stress on tumorigenesis on each phase and cell fate determination, and three features are discussed. Oxidative stress takes part in the processes ranging from tumorigenesis to tumor death via series pathways and processes like mitochondrial stress, endoplasmic reticulum stress, and ferroptosis. It can affect cell fate by engaging in the complex relationships between senescence, death, and cancer. The influence of oxidative stress on tumorigenesis and progression is a multi-stage interlaced process that includes two aspects of promotion and inhibition, with mitochondria as the core of regulation. A deeper and more comprehensive understanding of the effects of oxidative stress on tumorigenesis is conducive to exploring more tumor therapies.
Collapse
Affiliation(s)
| | | | | | | | | | - Changshan Wang
- Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Science, Inner Mongolia University, 49 Xilingol South Road, Yu Quan District, Hohhot 010020, China; (K.L.); (Z.D.); (C.L.); (X.D.); (J.L.)
| |
Collapse
|
5
|
Wang N, Gao E, Cui C, Wang F, Ren H, Xu C, Ning C, Zheng Y, Liu Q, Yu Q, Zhang G. The combined anticancer of peanut skin procyanidins and resveratrol to CACO-2 colorectal cancer cells. Food Sci Nutr 2023; 11:6483-6497. [PMID: 37831732 PMCID: PMC10563709 DOI: 10.1002/fsn3.3590] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 07/19/2023] [Accepted: 07/23/2023] [Indexed: 10/15/2023] Open
Abstract
Colorectal cancer is one of the leading causes of cancer deaths worldwide. Currently, chemotherapy is the primary way for colorectal cancer, but with severe side effects. Therefore, it is urgent to find safer and more effective adjuvant treatment methods. At present, natural active substances are promising alternatives, as numerous studies have demonstrated possible synergistic anticancer effects in plant-active polyphenols. In the present study, the combined effect of procyanidins (PC) (from peanut skin) and resveratrol (RES) (from peanut buds) on the synergistic anticancer potential was investigated. CACO-2 and HCT-8 cells were served as colorectal cancer models, and HEPG-2 and HUH-7 cells were served as liver cancer models to observe the effects of PC and RES alone or in combination on the growth and proliferation of these four types of cancer cells. The results revealed that both PC and RES could inhibit the cells' proliferation in a manner with concentration-dependent, but they exerted synergistic anticancer effects only on CACO-2 cells. PC and RES could synergistically inhibit CACO-2 cell clone formation, inducing apoptosis of CACO-2 cells and blocking their cell cycle in G0/G1 phase. Additionally, as observed by the results of Western blot assay, the combined effect of PC and RES also inhibited the phosphorylation of Thr308, Ser473, and ERK and promoted the phosphorylation of IKBα and NF-κB in CACO-2 cells. These findings collectively indicate that PC combined with RES might exert synergistic anticancer effects by regulating AKT, ERK, and NF-κB signaling pathways.
Collapse
Affiliation(s)
- Na Wang
- College of Food Science and TechnologyHenan Agricultural UniversityZhengzhouChina
- College of Animal MedicineHenan Agricultural UniversityZhengzhouChina
- Key Laboratory of Nutrition and Healthy Food of ZhengzhouZhengzhouChina
- International Joint Research Center for Animal ImmunologyZhengzhouChina
- Longhu Laboratory of Advanced ImmunologyZhengzhouChina
| | - Enguang Gao
- College of Food Science and TechnologyHenan Agricultural UniversityZhengzhouChina
- Key Laboratory of Nutrition and Healthy Food of ZhengzhouZhengzhouChina
- International Joint Research Center for Animal ImmunologyZhengzhouChina
| | - Chenxu Cui
- College of Food Science and TechnologyHenan Agricultural UniversityZhengzhouChina
- Key Laboratory of Nutrition and Healthy Food of ZhengzhouZhengzhouChina
- International Joint Research Center for Animal ImmunologyZhengzhouChina
| | - Fan Wang
- College of Food Science and TechnologyHenan Agricultural UniversityZhengzhouChina
- Key Laboratory of Nutrition and Healthy Food of ZhengzhouZhengzhouChina
| | - Hongtao Ren
- College of Food Science and TechnologyHenan Agricultural UniversityZhengzhouChina
- Key Laboratory of Nutrition and Healthy Food of ZhengzhouZhengzhouChina
- Longhu Laboratory of Advanced ImmunologyZhengzhouChina
| | - Chao Xu
- College of Food Science and TechnologyHenan Agricultural UniversityZhengzhouChina
- Key Laboratory of Nutrition and Healthy Food of ZhengzhouZhengzhouChina
| | - Cancan Ning
- College of Food Science and TechnologyHenan Agricultural UniversityZhengzhouChina
- Key Laboratory of Nutrition and Healthy Food of ZhengzhouZhengzhouChina
- International Joint Research Center for Animal ImmunologyZhengzhouChina
| | - Yuru Zheng
- College of Food Science and TechnologyHenan Agricultural UniversityZhengzhouChina
- Key Laboratory of Nutrition and Healthy Food of ZhengzhouZhengzhouChina
- International Joint Research Center for Animal ImmunologyZhengzhouChina
| | - Qingqing Liu
- College of Food Science and TechnologyHenan Agricultural UniversityZhengzhouChina
- Key Laboratory of Nutrition and Healthy Food of ZhengzhouZhengzhouChina
- International Joint Research Center for Animal ImmunologyZhengzhouChina
| | - Qiuying Yu
- College of Food Science and TechnologyHenan Agricultural UniversityZhengzhouChina
- Key Laboratory of Nutrition and Healthy Food of ZhengzhouZhengzhouChina
- International Joint Research Center for Animal ImmunologyZhengzhouChina
- Longhu Laboratory of Advanced ImmunologyZhengzhouChina
| | - Gaiping Zhang
- College of Animal MedicineHenan Agricultural UniversityZhengzhouChina
- International Joint Research Center for Animal ImmunologyZhengzhouChina
- Longhu Laboratory of Advanced ImmunologyZhengzhouChina
- School of Advanced Agricultural SciencesPeking UniversityBeijingChina
| |
Collapse
|
6
|
Xu C, Wang M, Zandieh Doulabi B, Sun Y, Liu Y. Paradox: Curcumin, a Natural Antioxidant, Suppresses Osteosarcoma Cells via Excessive Reactive Oxygen Species. Int J Mol Sci 2023; 24:11975. [PMID: 37569346 PMCID: PMC10418684 DOI: 10.3390/ijms241511975] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 07/18/2023] [Accepted: 07/25/2023] [Indexed: 08/13/2023] Open
Abstract
Osteosarcoma (OS) is an aggressive tumor with a rare incidence. Extended surgical resections are the prevalent treatment for OS, which may cause critical-size bone defects. These bone defects lead to dysfunction, weakening the post-surgical quality of patients' life. Hence, an ideal therapeutic agent for OS should simultaneously possess anti-cancer and bone repair capacities. Curcumin (CUR) has been reported in OS therapy and bone regeneration. However, it is not clear how CUR suppresses OS development. Conventionally, CUR is considered a natural antioxidant in line with its capacity to promote the nuclear translocation of a nuclear transcription factor, nuclear factor erythroid 2 (NRF2). After nuclear translocation, NRF2 can activate the transcription of some antioxidases, thereby circumventing excess reactive oxygen species (ROS) that are deleterious to cells. Intriguingly, this research demonstrated that, in vitro, 10 and 20 μM CUR increased the intracellular ROS in MG-63 cells, damaged cells' DNA, and finally caused apoptosis of MG-63 cells, although increased NRF2 protein level and the expression of NRF2-regulated antioxidase genes were identified in those two groups.
Collapse
Affiliation(s)
| | | | | | | | - Yuelian Liu
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), Vrije Universiteit Amsterdam and University of Amsterdam, 1081 LA Amsterdam, The Netherlands; (C.X.); (M.W.); (B.Z.D.); (Y.S.)
| |
Collapse
|
7
|
Moloudi K, Abrahamse H, George BP. Photodynamic therapy induced cell cycle arrest and cancer cell synchronization: review. Front Oncol 2023; 13:1225694. [PMID: 37503319 PMCID: PMC10369002 DOI: 10.3389/fonc.2023.1225694] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 06/21/2023] [Indexed: 07/29/2023] Open
Abstract
Cell cycle arrest (CCA) is seen as a prime candidate for effective cancer therapy. This mechanism can help researchers to create new treatments to target cancer cells at particular stages of the cell cycle (CC). The CCA is a characteristic of various therapeutic modalities, including radiation (RT) and chemotherapy (CT), which synchronizes the cells and facilitates the standardization of radio-chemotherapy protocols. Although it was discovered that photodynamic treatment (PDT) had a biological effect on CCA in cancer cells, the mechanism remains unclear. Furthermore, besides conventional forms of cell death such as apoptosis, autophagy, and necrosis, various unconventional types of cell death including pyroptosis, mitotic catastrophe, paraptosis, ferroptosis, necroptosis, and parthanatos after PDT have been reported. Thus, a variety of elements, such as oxygen, the tumor's microenvironment, the characteristics of light, and photosensitizer (PS), influence the effectiveness of the PDT treatment, which have not yet been studied clearly. This review focuses on CCA induced by PDT for a variety of PSs agents on various cell lines. The CCA by PDT can be viewed as a remarkable effect and instructive for the management of the PDT protocol. Regarding the relationship between the quantity of reactive oxygen species (ROS) and its biological consequences, we have proposed two mathematical models in PDT. Finally, we have gathered recent in vitro and in vivo studies about CCA post-PDT at various stages and made suggestions about how it can standardize, potentiate, and customize the PDT methodology.
Collapse
|
8
|
Allegra A, Murdaca G, Mirabile G, Gangemi S. Redox Signaling Modulates Activity of Immune Checkpoint Inhibitors in Cancer Patients. Biomedicines 2023; 11:biomedicines11051325. [PMID: 37238995 DOI: 10.3390/biomedicines11051325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 04/23/2023] [Accepted: 04/28/2023] [Indexed: 05/28/2023] Open
Abstract
Although immunotherapy is already a staple of cancer care, many patients may not benefit from these cutting-edge treatments. A crucial field of research now focuses on figuring out how to improve treatment efficacy and assess the resistance mechanisms underlying this uneven response. For a good response, immune-based treatments, in particular immune checkpoint inhibitors, rely on a strong infiltration of T cells into the tumour microenvironment. The severe metabolic environment that immune cells must endure can drastically reduce effector activity. These immune dysregulation-related tumour-mediated perturbations include oxidative stress, which can encourage lipid peroxidation, ER stress, and T regulatory cells dysfunction. In this review, we have made an effort to characterize the status of immunological checkpoints, the degree of oxidative stress, and the part that latter plays in determining the therapeutic impact of immunological check point inhibitors in different neoplastic diseases. In the second section of the review, we will make an effort to assess new therapeutic possibilities that, by affecting redox signalling, may modify the effectiveness of immunological treatment.
Collapse
Affiliation(s)
- Alessandro Allegra
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood "Gaetano Barresi", University of Messina, 98125 Messina, Italy
| | - Giuseppe Murdaca
- Department of Internal Medicine, Ospedale Policlinico San Martino IRCCS, University of Genova, Viale Benedetto XV, n. 6, 16132 Genova, Italy
| | - Giuseppe Mirabile
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood "Gaetano Barresi", University of Messina, 98125 Messina, Italy
| | - Sebastiano Gangemi
- Allergy and Clinical Immunology Unit, Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy
| |
Collapse
|
9
|
Wang H, Tian RF, Liang X, Fan J, Duan ZC, Fan XY, Zhang JJ, Yao DS, Chen ZN, Li L. A four oxidative stress gene prognostic model and integrated immunity-analysis in pancreatic adenocarcinoma. Front Oncol 2023; 12:1015042. [PMID: 36713541 PMCID: PMC9880292 DOI: 10.3389/fonc.2022.1015042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 12/28/2022] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND AND AIMS Pancreatic adenocarcinoma (PAAD) is highly aggressive and characterized by a poor prognosis. Oxidative stress has great impacts on the occurrence and development of tumors. However, the predictive role of oxidative stress related genes on PAAD patients' prognosis remains unclear. In this study, we aimed to construct a prognostic model for PAAD based on oxidative stress genes and to evaluate its predictive value. METHODS The Cancer Genome Atlas (TCGA) and three Gene Expression Omnibus (GEO) datasets were used to identify differentially expressed oxidative stress genes. Univariate Cox regression, Kaplan-Meier and multivariate Cox regression analysis were used to select genes and to construct a prognosis model. According to the median value of the model's risk score, patients were divided into high and low risk groups, and gene set enrichment analysis (GSEA), immune infiltration and immunotherapy effect, drug resistance and the expression of immune checkpoint related genes and synthetic driver genes of T cell proliferation were analyzed. Finally, the mRNA and protein levels of four genes in PAAD were verified by the clinical proteomic tumor analysis consortium (CPTAC) database and the immunostaining of patients' tissue. RESULTS 55 differentially expressed oxidative stress genes were identified, and four genes including MET, FYN, CTTN and CDK1 were selected to construct a prognosis model. GESA indicated that immune related pathways, metabolic pathways and DNA repair pathways were significantly enriched in the high risk group as compared to the low risk group. The frequency of genetic mutations was also significantly higher in high risk groups than that in low risk groups. Moreover, the infiltration level of 23 immune cells as well as the expression of immune checkpoint related and synthetic driver genes of T cell proliferation were significantly altered, with the better immunotherapy effect occurring in low risk group. In patient PAAD tissues, the mRNA and protein levels of these four genes were up-regulated. CONCLUSION We have successfully constructed a four oxidative stress gene prognostic model that has important predictive value for PAAD patients, and this model might be a promising guidance for prognostic prediction and efficacy monitoring in clinical individualized therapy.
Collapse
Affiliation(s)
- Hao Wang
- Institutes of Biomedicine and Department of Cell Biology, Jinan University, Guangzhou, China
| | - Ruo-Fei Tian
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi’an, China
| | - Xue Liang
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi’an, China
| | - Jing Fan
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi’an, China
| | - Zi-Chuan Duan
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi’an, China
| | - Xin-Yu Fan
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi’an, China
| | - Jia-Jia Zhang
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi’an, China
| | - Dong-Sheng Yao
- Institutes of Biomedicine and Department of Cell Biology, Jinan University, Guangzhou, China
| | - Zhi-Nan Chen
- Institutes of Biomedicine and Department of Cell Biology, Jinan University, Guangzhou, China
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi’an, China
| | - Ling Li
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi’an, China
| |
Collapse
|
10
|
Zhang X, Zhao H, Man J, Yin X, Zhang T, Yang X, Lu M. Investigating Causal Associations of Diet-Derived Circulating Antioxidants with the Risk of Digestive System Cancers: A Mendelian Randomization Study. Nutrients 2022; 14:3237. [PMID: 35956413 PMCID: PMC9370260 DOI: 10.3390/nu14153237] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 07/31/2022] [Accepted: 08/03/2022] [Indexed: 11/24/2022] Open
Abstract
Molecular mechanisms and observational studies have found that diet-derived antioxidants are associated with digestive system cancers, whereas there is a lack of causal evidence from randomized clinical trials. In this study, we aimed to assess the causality of these associations through a Mendelian randomization (MR) study. Single nucleotide polymorphisms of diet-derived circulating antioxidants (i.e., α- and γ-tocopherol, ascorbate, retinol, β-carotene, lycopene, and urate), accessed by absolute levels and relative metabolite concentrations, were used as genetic instruments. Summary statistics for digestive system cancers were obtained from the UK Biobank and FinnGen studies. Two-sample MR analyses were performed in each of the two outcome databases, followed by a meta-analysis. The inverse-variance weighted MR was adopted as the primary analysis. Five additional MR methods (likelihood-based MR, MR-Egger, weighted median, penalized weighted median, and MR-PRESSO) and replicate MR analyses for outcomes from different sources were used as sensitivity analyses. Genetically determined antioxidants were not significantly associated with five digestive system cancers, after correcting for multiple tests. However, we found suggestive evidence that absolute ascorbate levels were negatively associated with colon cancer in UK Biobank-the odds ratio (OR) per unit increase in ascorbate was 0.774 (95% confidence interval [CI] 0.608-0.985, p = 0.037), which was consistent with the results in FinnGen, and the combined OR was 0.764 (95% CI 0.623-0.936, p = 0.010). Likewise, higher absolute retinol levels suggestively reduced the pancreatic cancer risk in FinnGen-the OR per 10% unit increase in ln-transformed retinol was 0.705 (95% CI 0.529-0.940, p = 0.017), which was consistent with the results in UK Biobank and the combined OR was 0.747 (95% CI, 0.584-0.955, p = 0.020). Sensitivity analyses verified the above suggestive evidence. Our findings suggest that higher levels of antioxidants are unlikely to be a causal protective factor for most digestive system cancers, except for the suggestive protective effects of ascorbate on colon cancer and of retinol on pancreatic cancer.
Collapse
Affiliation(s)
- Xuening Zhang
- Department of Epidemiology and Health Statistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
- Clinical Epidemiology Unit, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Hao Zhao
- Department of Medical Statistics and Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Jinyu Man
- Department of Epidemiology and Health Statistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
- Clinical Epidemiology Unit, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Xiaolin Yin
- Department of Epidemiology and Health Statistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
- Clinical Epidemiology Unit, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Tongchao Zhang
- Clinical Epidemiology Unit, Qilu Hospital of Shandong University, Jinan 250012, China
- Clinical Research Center, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Xiaorong Yang
- Clinical Epidemiology Unit, Qilu Hospital of Shandong University, Jinan 250012, China
- Clinical Research Center, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Ming Lu
- Department of Epidemiology and Health Statistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
- Clinical Epidemiology Unit, Qilu Hospital of Shandong University, Jinan 250012, China
- Clinical Research Center, Qilu Hospital of Shandong University, Jinan 250012, China
| |
Collapse
|
11
|
Hamada S, Matsumoto R, Masamune A. HIF-1 and NRF2; Key Molecules for Malignant Phenotypes of Pancreatic Cancer. Cancers (Basel) 2022; 14:cancers14020411. [PMID: 35053572 PMCID: PMC8773475 DOI: 10.3390/cancers14020411] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 01/13/2022] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Pancreatic cancer progression involves interactions between cancer cells and stromal cells in harsh tumor microenvironments, which are characterized by hypoxia, few nutrients, and oxidative stress. Clinically, cancer cells overcome therapeutic interventions, such as chemotherapy and radiotherapy, to continue to survive. Activation of the adaptation mechanism is required for cancer cell survival under these conditions, and it also contributes to the acquisition of the malignant phenotype. Stromal cells, especially pancreatic stellate cells, play a critical role in the formation of a cancer-promoting microenvironment. We here review the roles of key molecules, hypoxia inducible factor-1 and KEAP1-NRF2, in stress response mechanisms for the adaptation to hypoxia and oxidative stress in pancreatic cancer cells and stellate cells. Various cancer-promoting properties associated with these molecules have been identified, and they might serve as novel therapeutic targets in the future. Abstract Pancreatic cancer is intractable due to early progression and resistance to conventional therapy. Dense fibrotic stroma, known as desmoplasia, is a characteristic feature of pancreatic cancer, and develops through the interactions between pancreatic cancer cells and stromal cells, including pancreatic stellate cells. Dense stroma forms harsh tumor microenvironments characterized by hypoxia, few nutrients, and oxidative stress. Pancreatic cancer cells as well as pancreatic stellate cells survive in the harsh microenvironments through the altered expression of signaling molecules, transporters, and metabolic enzymes governed by various stress response mechanisms. Hypoxia inducible factor-1 and KEAP1-NRF2, stress response mechanisms for hypoxia and oxidative stress, respectively, contribute to the aggressive behaviors of pancreatic cancer. These key molecules for stress response mechanisms are activated, both in pancreatic cancer cells and in pancreatic stellate cells. Both factors are involved in the mutual activation of cancer cells and stellate cells, by inducing cancer-promoting signals and their mediators. Therapeutic interventions targeting these pathways are promising approaches for novel therapies. In this review, we summarize the roles of stress response mechanisms, focusing on hypoxia inducible factor-1 and KEAP1-NRF2, in pancreatic cancer. In addition, we discuss the potential of targeting these molecules for the treatment of pancreatic cancer.
Collapse
|
12
|
Popova NV, Jücker M. The Functional Role of Extracellular Matrix Proteins in Cancer. Cancers (Basel) 2022; 14:238. [PMID: 35008401 PMCID: PMC8750014 DOI: 10.3390/cancers14010238] [Citation(s) in RCA: 105] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/23/2021] [Accepted: 12/27/2021] [Indexed: 02/04/2023] Open
Abstract
The extracellular matrix (ECM) is highly dynamic as it is constantly deposited, remodeled and degraded to maintain tissue homeostasis. ECM is a major structural component of the tumor microenvironment, and cancer development and progression require its extensive reorganization. Cancerized ECM is biochemically different in its composition and is stiffer compared to normal ECM. The abnormal ECM affects cancer progression by directly promoting cell proliferation, survival, migration and differentiation. The restructured extracellular matrix and its degradation fragments (matrikines) also modulate the signaling cascades mediated by the interaction with cell-surface receptors, deregulate the stromal cell behavior and lead to emergence of an oncogenic microenvironment. Here, we summarize the current state of understanding how the composition and structure of ECM changes during cancer progression. We also describe the functional role of key proteins, especially tenascin C and fibronectin, and signaling molecules involved in the formation of the tumor microenvironment, as well as the signaling pathways that they activate in cancer cells.
Collapse
Affiliation(s)
- Nadezhda V. Popova
- Laboratory of Receptor Cell Biology, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho-Maklaya Str., 16/10, 117997 Moscow, Russia;
| | - Manfred Jücker
- Institute of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246 Hamburg, Germany
| |
Collapse
|
13
|
Bi Y, Lei X, Chai N, Linghu E. NOX4: a potential therapeutic target for pancreatic cancer and its mechanism. J Transl Med 2021; 19:515. [PMID: 34930338 PMCID: PMC8686284 DOI: 10.1186/s12967-021-03182-w] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 12/03/2021] [Indexed: 12/18/2022] Open
Abstract
Nicotinamide adenine dinucleotide phosphate (NADPH) oxidase 4 (NOX4) is one of the seven isoforms of NOX family, which is upregulated in pancreatic cancer cell, mouse model of pancreatic cancer and human pancreatic cancer tissue. NOX4 is a constitutively active enzyme that primarily produces hydrogen peroxide, which exhibits completely different properties from other subtypes of NOX family. More importantly, recent studies illuminate that NOX4 promotes pancreatic cancer occurrence and development in different ways. This review summarizes the potential roles and its mechanism of NOX4 in pancreatic cancer and explores NOX4 as the potential therapeutic target in pancreatic cancer.
Collapse
Affiliation(s)
- Yawei Bi
- Department of Gastroenterology and Hepatology, The First Medical Center of PLA General Hospital, Beijing, 100853, China
| | - Xiao Lei
- Senior Department of Oncology, The Fifth Medical Center of PLA General Hospital, Beijing, 100859, China
| | - Ningli Chai
- Department of Gastroenterology and Hepatology, The First Medical Center of PLA General Hospital, Beijing, 100853, China.
| | - Enqiang Linghu
- Department of Gastroenterology and Hepatology, The First Medical Center of PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|
14
|
Sahoo BM, Banik BK, Borah P, Jain A. Reactive Oxygen Species (ROS): Key components in Cancer Therapies. Anticancer Agents Med Chem 2021; 22:215-222. [PMID: 34102991 DOI: 10.2174/1871520621666210608095512] [Citation(s) in RCA: 118] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 03/31/2021] [Accepted: 04/05/2021] [Indexed: 11/22/2022]
Abstract
Reactive oxygen species (ROS) refer to the highly reactive substances, which contain oxygen radicals. Hypochlorous acid, peroxides, superoxide, singlet oxygen, alpha-oxygen and hydroxyl radicals are the major examples of ROS. Generally, the reduction of oxygen (O2) in molecular form produces superoxide (•O2-) anion. ROS are produced during a variety of biochemical reactions within the cell organelles, such as endoplasmic reticulum, mitochondria and peroxisome. Naturally, ROS are also formed as a byproduct of the normal metabolism of oxygen. The production of ROS can be induced by various factors such as heavy metals, tobacco, smoke, drugs, xenobiotics, pollutants and radiation. From various experimental studies, it is reported that ROS act as either tumor suppressing or tumor promoting agent. The elevated levels of ROS can arrest the growth of tumor through the persistent increase in cell cycle inhibition. The increased level of ROS can induce apoptosis by both intrinsic and extrinsic pathways. ROS are considered to be tumor suppressing agent as the production of ROS is due to the use of most of the chemotherapeutic agents in order to activate the cell death. The cytotoxic effect of ROS provides impetus towards apoptosis, but in higher levels, ROS can cause initiation of malignancy that leads to uncontrolled cell death in cancer cells. Whereas, some species of ROS can influence various activities at the cellular level that include cell proliferation. This review highlights the genesis of ROS within cells by various routes and their role in cancer therapies.
Collapse
Affiliation(s)
- Biswa Mohan Sahoo
- Roland Institute of Pharmaceutical Sciences (Biju Patnaik University of Technology Nodal Centre of Research), Berhampur-760010, Odisha, India
| | - Bimal Krishna Banik
- Department of Mathematics and Natural Sciences, College of Sciences and Human Studies, Prince Mohammad Bin Fahd University, Al Khobar, Saudi Arabia
| | | | - Adya Jain
- Department of Chemistry, MRK Educational Institutions, IGU Rewari, Haryana, India
| |
Collapse
|
15
|
Wu Q, Li W, Zhao J, Sun W, Yang Q, Chen C, Xia P, Zhu J, Zhou Y, Huang G, Yong C, Zheng M, Zhou E, Gao K. Apigenin ameliorates doxorubicin-induced renal injury via inhibition of oxidative stress and inflammation. Biomed Pharmacother 2021; 137:111308. [PMID: 33556877 DOI: 10.1016/j.biopha.2021.111308] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 01/10/2021] [Accepted: 01/12/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND AND OBJECTIVE Doxorubicin (DOX) is an anthracycline antitumor antibiotic widely utilized in treating various tumors. Nevertheless, the toxicity of DOX toward normal cells limits its applicability, with nephrotoxicity considered a major dose-limiting adverse effect. Apigenin (APG), a flavonoid widely distributed in natural plants, has been reported to have antioxidant, anti-inflammatory, and mild tumor-suppressive properties. In this study, we investigated the role of APG in DOX-induced nephrotoxicity and chemotherapeutic efficacy. METHODS Male BALB/c mice were administered DOX (11.5 mg/kg) via the tail vein to establish the DOX nephropathy model. After treatment with or without APG (125, 250, and 500 mg/kg) for two weeks, urine, serum, and tissue samples were collected to evaluate proteinuria, serum albumin, serum creatinine (Scr), blood urea nitrogen (BUN), superoxide dismutase (SOD) activity, malondialdehyde (MDA), glutathione (GSH), and pathological changes. Rat renal tubular epithelial cells (NRK52E), murine podocyte cells (MPC5), and murine breast cancer cells (4T1) were utilized to verify the effect of APG on DOX-induced cell injury. An MTT assay was employed to analyze cell viability. Apoptosis was evaluated using a colorimetric TUNEL staining and cleaved caspase-3 protein analysis by western blotting. A reactive oxygen species (ROS)/superoxide (O2-) fluorescence probe was employed to determine oxidative injury. Western blotting was used to analyze nephrin, α-smooth muscle actin (α-SMA), collagen I (Col1), fibronectin (FN), and SOD2 expression. The mRNA levels of tumor necrosis factor-α (TNF-α), interleukin-18 (IL-18), IL-6, NACHT, LRR, PYD domain-containing protein 3 (NLRP3), caspase-1, and IL-1β were tested by reverse transcription-polymerase chain reaction (RT-PCR). RESULTS APG ameliorated DOX-elicited renal injuries in both the glomeruli and tubules. The DOX + APG groups had much lower tissue MDA, IL-6, TNF-α, NLRP3, caspase-1, and IL-1β levels and generation of intracellular ROS, but significantly higher SOD activity and GSH levels compared to those of the DOX group. Additionally, APG attenuated DOX-induced morphological changes, loss of cellular viability, and apoptosis in NRK-52E and MPC-5 cells, but not in 4T1 cells. CONCLUSION APG has a protective role against DOX-induced nephrotoxicity, without weakening DOX cytotoxicity in malignant tumors. Thus, APG may serve as a potential protective agent against renal injury and inflammatory diseases and may be a promising candidate to attenuate renal toxicity in cancer patients treated with DOX.
Collapse
Affiliation(s)
- Qijing Wu
- Division of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China
| | - Wei Li
- Division of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China
| | - Jing Zhao
- Division of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China
| | - Wei Sun
- Division of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China.
| | - Qianqian Yang
- Center for Kidney Disease, 2nd Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Chong Chen
- Division of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China
| | - Ping Xia
- Division of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China
| | - Jingjing Zhu
- Division of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China
| | - Yiceng Zhou
- Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, Jiangsu Province, China
| | - Guoshun Huang
- Division of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China
| | - Chen Yong
- Division of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China
| | - Min Zheng
- Division of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China
| | - Enchao Zhou
- Division of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China
| | - Kun Gao
- Division of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China.
| |
Collapse
|
16
|
Sala M, Ros M, Saltel F. A Complex and Evolutive Character: Two Face Aspects of ECM in Tumor Progression. Front Oncol 2020; 10:1620. [PMID: 32984031 PMCID: PMC7485352 DOI: 10.3389/fonc.2020.01620] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 07/27/2020] [Indexed: 12/24/2022] Open
Abstract
Tumor microenvironment, including extracellular matrix (ECM) and stromal cells, is a key player during tumor development, from initiation, growth and progression to metastasis. During all of these steps, remodeling of matrix components occurs, changing its biochemical and physical properties. The global and basic cancer ECM model is that tumors are surrounded by activated stromal cells, that remodel physiological ECM to evolve into a stiffer and more crosslinked ECM than in normal conditions, thereby increasing invasive capacities of cancer cells. In this review, we show that this too simple model does not consider the complexity, specificity and heterogeneity of each organ and tumor. First, we describe the general ECM in context of cancer. Then, we go through five invasive and most frequent cancers from different origins (breast, liver, pancreas, colon, and skin), and show that each cancer has its own specific matrix, with different stromal cells, ECM components, biochemical properties and activated signaling pathways. Furthermore, in these five cancers, we describe the dual role of tumor ECM: as a protective barrier against tumor cell proliferation and invasion, and as a major player in tumor progression. Indeed, crosstalk between tumor and stromal cells induce changes in matrix organization by remodeling ECM through invadosome formation in order to degrade it, promoting tumor progression and cell invasion. To sum up, in this review, we highlight the specificities of matrix composition in five cancers and the necessity not to consider the ECM as one general and simple entity, but one complex, dynamic and specific entity for each cancer type and subtype.
Collapse
|
17
|
Hadden M, Mittal A, Samra J, Zreiqat H, Sahni S, Ramaswamy Y. Mechanically stressed cancer microenvironment: Role in pancreatic cancer progression. Biochim Biophys Acta Rev Cancer 2020; 1874:188418. [PMID: 32827581 DOI: 10.1016/j.bbcan.2020.188418] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 07/21/2020] [Accepted: 08/12/2020] [Indexed: 02/06/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal solid malignancies in the world due to its insensitivity to current therapies and its propensity to metastases from the primary tumor mass. This is largely attributed to its complex microenvironment composed of unique stromal cell populations and extracellular matrix (ECM). The recruitment and activation of these cell populations cause an increase in deposition of ECM components, which highly influences the behavior of malignant cells through disrupted forms of signaling. As PDAC progresses from premalignant lesion to invasive carcinoma, this dynamic landscape shields the mass from immune defenses and cytotoxic intervention. This microenvironment influences an invasive cell phenotype through altered forms of mechanical signaling, capable of enacting biochemical changes within cells through activated mechanotransduction pathways. The effects of altered mechanical cues on malignant cell mechanotransduction have long remained enigmatic, particularly in PDAC, whose microenvironment significantly changes over time. A more complete and thorough understanding of PDAC's physical surroundings (microenvironment), mechanosensing proteins, and mechanical properties may help in identifying novel mechanisms that influence disease progression, and thus, provide new potential therapeutic targets.
Collapse
Affiliation(s)
- Matthew Hadden
- School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, NSW 2006, Australia
| | - Anubhav Mittal
- Northern Clinical School, Faculty of Medicine and Health, University of Sydney, Australia; Kolling Institute of Medical Research, University of Sydney, Australia; Australian Pancreatic Centre, St Leonards, Sydney, Australia
| | - Jaswinder Samra
- Northern Clinical School, Faculty of Medicine and Health, University of Sydney, Australia; Kolling Institute of Medical Research, University of Sydney, Australia; Australian Pancreatic Centre, St Leonards, Sydney, Australia
| | - Hala Zreiqat
- School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, NSW 2006, Australia; ARC Training Centre for Innovative Bioengineering, The University of Sydney, NSW 2006, Australia; The University of Sydney Nano Institute, The University of Sydney, Sydney, NSW 2006, Australia
| | - Sumit Sahni
- Northern Clinical School, Faculty of Medicine and Health, University of Sydney, Australia; Kolling Institute of Medical Research, University of Sydney, Australia; Australian Pancreatic Centre, St Leonards, Sydney, Australia.
| | - Yogambha Ramaswamy
- School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, NSW 2006, Australia; The University of Sydney Nano Institute, The University of Sydney, Sydney, NSW 2006, Australia.
| |
Collapse
|
18
|
Roife D, Fleming JB, Gomer RH. Fibrocytes in the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1224:79-85. [PMID: 32036606 PMCID: PMC7212529 DOI: 10.1007/978-3-030-35723-8_6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Tumors have long been compared to chronic wounds that do not heal, since they share many of the same molecular and cellular processes. In normal wounds, healing processes lead to restoration of cellular architecture, while in malignant tumors, these healing processes become dysregulated and contribute to growth and invasion of neoplastic cells into the surrounding tissues. Fibrocytes are fibroblast-like cells that differentiate from bone marrow-derived CD14+ circulating monocytes and aid wound healing. Although most monocytes will differentiate into macrophages after extravasating into a tissue, signals present in a wound environment can cause some monocytes to differentiate into fibrocytes. The fibrocytes secrete matrix proteins and inflammatory cytokines, activate local fibroblasts to proliferate and increase extracellular matrix production, and promote angiogenesis, and because fibrocytes are contractile, they also help wound contraction. There is now emerging evidence that fibrocytes are present in the tumor microenvironment, attracted by the chronic tissue damage and cytokines from both cancer cells and other immune cells. Fibrocytes may aid in the survival and spread of neoplastic cells, so these wound-healing cells may be a promising target for anticancer research in future studies.
Collapse
Affiliation(s)
- David Roife
- Department of Surgery, University of South Florida Morsani College of Medicine, Tampa, FL, USA
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Jason B Fleming
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Richard H Gomer
- Department of Biology/ILSB, Texas A&M University, College Station, TX, USA.
| |
Collapse
|
19
|
Shah IA, Bhat GA, Rafiq R, Nissa N, Muzaffar M, Rasool MT, Lone MM, Lone GN, Boffetta P, Dar NA. Strenuous occupational physical activity: Potential association with esophageal squamous cell carcinoma risk. PROCEEDINGS OF SINGAPORE HEALTHCARE 2019; 28:232-242. [DOI: 10.1177/2010105819860860] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023] Open
Abstract
Objective:The impact of recreational physical activity (RPA) on cancer risk has been extensively studied. However, the association of occupational physical activity (OPA), which differs in dose and intensity from RPA, with different cancers including esophageal squamous cell carcinoma (ESCC), has received less attention.Materials and methods:We conducted a hospital-based case–control study in Kashmir, India, majorly a rural population, to evaluate the association of OPA with ESCC risk. Histopathologically confirmed 703 ESCC cases and 1664 controls, individually matched to the respective cases for age, sex and district of residence, were recruited.Main outcome measures:Information on type, duration and intensity of physical activity was obtained in face-to-face interviews with participants using a structured questionnaire. Conditional logistic regression models were used to calculate odds ratios (ORs) and 95% confidence intervals (95% CIs). Body mass index was unable to be accounted for in the analysis.Results:A high level of OPA was associated with increased ESCC risk (OR = 2.17, 95% CI; 1.41–3.32), compared to subjects with moderate OPA. The association with ESCC risk was stronger in strenuous workers (OR = 3.64, 95% CI; 2.13–6.20). The association of strenuous OPA with ESCC risk persisted only in subjects that were involved in strenuous activities for equal to or greater than five days/week.Conclusions:Our study suggests a possible association of strenuous OPA with ESCC risk. Although our results were adjusted for multiple factors, including indicators of socioeconomic status, more replicative occupational epidemiological studies are needed to rule out any residual confounding.
Collapse
Affiliation(s)
- Idrees Ayoub Shah
- Department of Biochemistry, University of Kashmir, Srinagar, India
- Department of Human Genetics, Punjabi University Patiala, India
| | | | - Rumaisa Rafiq
- Department of Biochemistry, University of Kashmir, Srinagar, India
| | - Najma Nissa
- Department of Biochemistry, University of Kashmir, Srinagar, India
| | - Mansha Muzaffar
- Department of Biochemistry, University of Kashmir, Srinagar, India
| | - Malik Tariq Rasool
- Department of Radiation Oncology, SK Institute of Medical Sciences, Srinagar, India
| | - Mohd Maqbool Lone
- Department of Radiation Oncology, SK Institute of Medical Sciences, Srinagar, India
| | - Ghulam Nabi Lone
- Department of CVTS, SK Institute of Medical Sciences, Srinagar, India
| | - Paolo Boffetta
- The Tisch Cancer Institute, Mount Sinai School of Medicine, New York, USA
| | - Nazir Ahmad Dar
- Department of Biochemistry, University of Kashmir, Srinagar, India
| |
Collapse
|
20
|
Pandya AD, Jäger E, Bagheri Fam S, Höcherl A, Jäger A, Sincari V, Nyström B, Štěpánek P, Skotland T, Sandvig K, Hrubý M, Mælandsmo GM. Paclitaxel-loaded biodegradable ROS-sensitive nanoparticles for cancer therapy. Int J Nanomedicine 2019; 14:6269-6285. [PMID: 31496685 PMCID: PMC6689768 DOI: 10.2147/ijn.s208938] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Accepted: 06/13/2019] [Indexed: 12/15/2022] Open
Abstract
Background Reactive oxygen species (ROS), such as hydrogen peroxide and superoxide, trigger biodegradation of polymer-based nanoparticles (NPs) bearing pinacol-type boronic ester groups. These NPs may selectively release their cargo, in this case paclitaxel (PTX), at the high levels of ROS present in the intracellular environment of inflamed tissues and most tumors. Purpose The main objective was to determine anti-tumor efficacy of PTX-loaded ROS-sensitive NPs and to examine whether macrophage infiltration had any impact on treatment efficacy. Methods NPs were synthesized and their characteristics in the presence of H2O2 were demonstrated. Both confocal microscopy as well as flow cytometry approaches were used to determine degradation of ROS-sensitive NPs. HeLa cells were cultured in vitro and used to establish tumor xenografts in nude mice. In vivo experiments were performed to understand toxicity, biodistribution and anti-tumor efficacy of the NPs. Moreover, we performed immunohistochemistry on tumor sections to study infiltration of M1 and M2 subsets of macrophages. Results We demonstrated that PTX delivered in NPs containing a ROS-sensitive polymer exhibits a better anti-tumor efficacy than PTX in NPs containing ROS-non-sensitive polymer, free PTX or Abraxane® (nab-PTX). The biodistribution revealed that ROS-sensitive NPs exhibit retention in liver, spleen and lungs, suggesting a potential to target cancer metastasizing to these organs. Finally, we demonstrated a correlation between infiltrated macrophage subsets and treatment efficacy, possibly contributing to the efficient anti-tumor effects. Conclusion Treatment with ROS-sensitive NPs containing PTX gave an improved therapeutic effect in HeLa xenografts than their counterpart, free PTX or nab-PTX. Our data revealed a correlation between macrophage infiltration and efficiency of the different antitumor treatments, as the most effective NPs resulted in the highest infiltration of the anti-tumorigenic M1 macrophages.
Collapse
Affiliation(s)
- Abhilash D Pandya
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway
| | - Eliézer Jäger
- Institute of Macromolecular Chemistry v.v.i, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Shahla Bagheri Fam
- Department of Radiation Biology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway
| | - Anita Höcherl
- Institute of Macromolecular Chemistry v.v.i, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Alessandro Jäger
- Institute of Macromolecular Chemistry v.v.i, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Vladimir Sincari
- Institute of Macromolecular Chemistry v.v.i, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Bo Nyström
- Department of Chemistry, University of Oslo, Oslo, Norway
| | - Petr Štěpánek
- Institute of Macromolecular Chemistry v.v.i, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Tore Skotland
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway
| | - Kirsten Sandvig
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway.,Department of Biosciences, University of Oslo, Oslo, Norway
| | - Martin Hrubý
- Institute of Macromolecular Chemistry v.v.i, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Gunhild M Mælandsmo
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway.,Institute of Medical Biology, Faculty of Health Sciences, The Arctic University of Norway - University of Tromsø, Tromsø, Norway
| |
Collapse
|
21
|
Edderkaoui M, Chheda C, Soufi B, Zayou F, Hu RW, Krishnan Ramanujan V, Pan X, Boros LG, Tajbakhsh J, Madhav A, Bhowmick NA, Wang Q, Lewis M, Tuli R, Habtezion A, Murali R, Pandol SJ. An Inhibitor of GSK3B and HDACs Kills Pancreatic Cancer Cells and Slows Pancreatic Tumor Growth and Metastasis in Mice. Gastroenterology 2018; 155:1985-1998.e5. [PMID: 30144430 PMCID: PMC6328046 DOI: 10.1053/j.gastro.2018.08.028] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 06/14/2018] [Accepted: 08/05/2018] [Indexed: 12/17/2022]
Abstract
BACKGROUND & AIMS Growth, progression, and drug resistance of pancreatic ductal adenocarcinomas (PDACs) have been associated with increased levels and activity of glycogen synthase kinase 3 beta (GSK3B) and histone deacetylases (HDACs). We designed and synthesized molecules that simultaneously inhibit the activities of both enzymes. We tested the effects of one of these molecules, Metavert, in pancreatic cancer cells and mice with pancreatic tumors. METHODS We tested the ability of Metavert to bind GSK3B and HDACs using surface plasmon resonance. MIA PaCa-2, Bx-PC3, HPAF-II, and HPDE6 cell lines were incubated with different concentrations of Metavert, with or without paclitaxel or gemcitabine, or with other inhibitors of GSK3B and HDACs; cells were analyzed for apoptosis and migration and by immunoblotting, immunofluorescence, and real-time polymerase chain reaction. Krasþ/LSLG12D;Trp53þ/LSLR172H;Pdx-1-Cre (KPC) mice (2 months old) were given injections of Metavert (5 mg/kg, 3 times/week) or vehicle (control). B6.129J mice with tumors grown from UN-KPC961-Luc cells were given injections of Metavert or vehicle. Tumors and metastases were counted and pancreata were analyzed by immunohistochemistry. Glucose metabolism was measured using 13C-glucose tracer and mass spectroscopy and flow cytometry. Cytokine levels in blood samples were measured using multiplexing enzyme-linked immunosorbent assay. RESULTS Metavert significantly reduced survival of PDAC cells but not nontransformed cells; the agent reduced markers of the epithelial-to-mesenchymal transition and stem cells in PDAC cell lines. Cells incubated with Metavert in combination with irradiation and paclitaxel or gemcitabine had reduced survival compared with cells incubated with either agent alone; Metavert increased killing of drug-resistant PDAC cells by paclitaxel and gemcitabine. PDAC cells incubated with Metavert acquired normalized glucose metabolism. Administration of Metavert (alone or in combination with gemcitibine) to KPC mice or mice with syngeneic tumors significantly increased their survival times, slowed tumor growth, prevented tumor metastasis, decreased tumor infiltration by tumor-associated macrophages, and decreased blood levels of cytokines. CONCLUSIONS In studies of PDAC cells and 2 mouse models of PDAC, we found a dual inhibitor of GSK3B and HDACs (Metavert) to induce cancer cell apoptosis, reduce migration and expression of stem cell markers, and slow growth of tumors and metastases. Metavert had synergistic effects with gemcitabine.
Collapse
Affiliation(s)
- Mouad Edderkaoui
- Departments of Medicine, Biomedical Sciences, Radiation Oncology, and Surgery, Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, Los Angeles, California; Department of Pediatrics, University of California at Los Angeles, Los Angeles, California.
| | - Chintan Chheda
- Departments of Medicine, Biomedical Sciences, Radiation Oncology and Surgery, Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, Los Angeles, California
| | - Badr Soufi
- Departments of Medicine, Biomedical Sciences, Radiation Oncology and Surgery, Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, Los Angeles, California
| | - Fouzia Zayou
- Departments of Medicine, Biomedical Sciences, Radiation Oncology and Surgery, Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, Los Angeles, California
| | - Robert W. Hu
- Departments of Medicine, Biomedical Sciences, Radiation Oncology and Surgery, Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, Los Angeles, California
| | - V. Krishnan Ramanujan
- Departments of Medicine, Biomedical Sciences, Radiation Oncology and Surgery, Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, Los Angeles, California
| | - Xinlei Pan
- Departments of Medicine, Biomedical Sciences, Radiation Oncology and Surgery, Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, Los Angeles, California
| | - Laszlo G. Boros
- Department of Pediatrics, University of California at Los Angeles, California
| | - Jian Tajbakhsh
- Departments of Medicine, Biomedical Sciences, Radiation Oncology and Surgery, Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, Los Angeles, California
| | - Anisha Madhav
- Departments of Medicine, Biomedical Sciences, Radiation Oncology and Surgery, Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, Los Angeles, California
| | - Neil A. Bhowmick
- Departments of Medicine, Biomedical Sciences, Radiation Oncology and Surgery, Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, Los Angeles, California
| | - Qiang Wang
- Departments of Medicine, Biomedical Sciences, Radiation Oncology and Surgery, Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, Los Angeles, California
| | | | - Richard Tuli
- Departments of Medicine, Biomedical Sciences, Radiation Oncology and Surgery, Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, Los Angeles, California
| | - Aida Habtezion
- Division of Gastroenterology and Hepatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA
| | - Ramachandran Murali
- Departments of Medicine, Biomedical Sciences, Radiation Oncology and Surgery, Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, Los Angeles, California
| | - Stephen J. Pandol
- Departments of Medicine, Biomedical Sciences, Radiation Oncology and Surgery, Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, Los Angeles, California,Department of Pediatrics, University of California at Los Angeles, California
| |
Collapse
|
22
|
Tang CT, Gao YJ, Ge ZZ. NOX4, a new genetic target for anti-cancer therapy in digestive system cancer. J Dig Dis 2018; 19:578-585. [PMID: 30058122 DOI: 10.1111/1751-2980.12651] [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: 05/05/2018] [Revised: 07/19/2018] [Accepted: 07/24/2018] [Indexed: 12/11/2022]
Abstract
Oxidative stress has been implicated as an important factor in tumorigenesis and tumor progression. The nicotinamide adenine dinucleotide phosphate (NADPH) oxidase subunit 4 (NOX4), a substrate of NADPH that can generate H2 O2 reactive oxygen species, has been reported to be highly expressed in gastrointestinal tumors. In this review we summarize the available evidence on the biological function of NOX4 in digestive system tumors by focusing on its correlation with classical cell signaling pathways, including VEGF, MAPK and PI3K/AKT, and with biochemical mediators, such as nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), activator protein (AP)-1 and transforming growth factor (TGF)-β. According to the clinical and database studies on tumors of the digestive system, such as colorectal, gastric and pancreatic cancer, there are significant associations between NOX4 expression and tumor prognosis as well as patient's survival. Animal studies using NOX4 inhibitors such as diphenylene iodonium and GKT137831, which selectively block NOX4, indicate their potential as therapeutic agents for targeting cancer cells.
Collapse
Affiliation(s)
- Chao Tao Tang
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai, China
| | - Yun Jie Gao
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai, China
| | - Zhi Zheng Ge
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai, China
| |
Collapse
|
23
|
The Extracellular Matrix and Pancreatic Cancer: A Complex Relationship. Cancers (Basel) 2018; 10:cancers10090316. [PMID: 30200666 PMCID: PMC6162452 DOI: 10.3390/cancers10090316] [Citation(s) in RCA: 189] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 08/31/2018] [Accepted: 09/02/2018] [Indexed: 12/18/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has an extraordinarily dense fibrotic stroma that impedes tumor perfusion and delivery of anticancer drugs. Since the extracellular matrix (ECM) comprises the bulk of the stroma, it is primarily responsible for the increased interstitial tissue pressure and stiff mechanical properties of the stroma. Besides its mechanical influence, the ECM provides important biochemical and physical cues that promote survival, proliferation, and metastasis. By serving as a nutritional source, the ECM also enables PDAC cells to survive under the nutrient-poor conditions. While therapeutic strategies using stroma-depleting drugs have yielded disappointing results, an increasing body of research indicates the ECM may offer a variety of potential therapeutic targets. As preclinical studies of ECM-targeted drugs have shown promising effects, a number of clinical trials are currently investigating agents with the potential to advance the future treatment of PDAC. Thus, the present review seeks to give an overview of the complex relationship between the ECM and PDAC.
Collapse
|
24
|
Role of Oxidative and Nitro-Oxidative Damage in Silver Nanoparticles Cytotoxic Effect against Human Pancreatic Ductal Adenocarcinoma Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:8251961. [PMID: 30186549 PMCID: PMC6116403 DOI: 10.1155/2018/8251961] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Revised: 06/26/2018] [Accepted: 07/05/2018] [Indexed: 01/04/2023]
Abstract
Pancreatic ductal adenocarcinoma is one of the most aggressive human malignancies, where the 5-year survival rate is less than 4% worldwide. Successful treatment of pancreatic cancer is a challenge for today's oncology. Several studies showed that increased levels of oxidative stress may cause cancer cells damage and death. Therefore, we hypothesized that oxidative as well as nitro-oxidative stress is one of the mechanisms inducing pancreatic cancer programmed cell death. We decided to use silver nanoparticles (AgNPs) (2.6 and 18 nm) as a key factor triggering the reactive oxygen species (ROS) and reactive nitrogen species (RNS) in pancreatic ductal adenocarcinoma cells (PANC-1). Previously, we have found that AgNPs induced PANC-1 cells death. Furthermore, it is known that AgNPs may induce an accumulation of ROS and alteration of antioxidant systems in different type of tumors, and they are indicated as promising agents for cancer therapy. Then, the aim of our study was to evaluate the implication of oxidative and nitro-oxidative stress in this cytotoxic effect of AgNPs against PANC-1 cells. We determined AgNP-induced increase of ROS level in PANC-1 cells and pancreatic noncancer cell (hTERT-HPNE) for comparison purposes. We found that the increase was lower in noncancer cells. Reduction of mitochondrial membrane potential and changes in the cell cycle were also observed. Additionally, we determined the increase in RNS level: nitric oxide (NO) and nitric dioxide (NO2) in PANC-1 cells, together with increase in family of nitric oxide synthases (iNOS, eNOS, and nNOS) at protein and mRNA level. Disturbance of antioxidant enzymes: superoxide dismutase (SOD1, SOD2, and SOD3), glutathione peroxidase (GPX-4) and catalase (CAT) were proved at protein and mRNA level. Moreover, we showed cells ultrastructural changes, characteristic for oxidative damage. Summarizing, oxidative and nitro-oxidative stress and mitochondrial disruption are implicated in AgNPs-mediated death in human pancreatic ductal adenocarcinoma cells.
Collapse
|
25
|
Vallée A, Lecarpentier Y. Crosstalk Between Peroxisome Proliferator-Activated Receptor Gamma and the Canonical WNT/β-Catenin Pathway in Chronic Inflammation and Oxidative Stress During Carcinogenesis. Front Immunol 2018; 9:745. [PMID: 29706964 PMCID: PMC5908886 DOI: 10.3389/fimmu.2018.00745] [Citation(s) in RCA: 264] [Impact Index Per Article: 37.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Accepted: 03/26/2018] [Indexed: 12/19/2022] Open
Abstract
Inflammation and oxidative stress are common and co-substantial pathological processes accompanying, promoting, and even initiating numerous cancers. The canonical WNT/β-catenin pathway and peroxisome proliferator-activated receptor gamma (PPARγ) generally work in opposition. If one of them is upregulated, the other one is downregulated and vice versa. WNT/β-catenin signaling is upregulated in inflammatory processes and oxidative stress and in many cancers, although there are some exceptions for cancers. The opposite is observed with PPARγ, which is generally downregulated during inflammation and oxidative stress and in many cancers. This helps to explain in part the opposite and unidirectional profile of the canonical WNT/β-catenin signaling and PPARγ in these three frequent and morbid processes that potentiate each other and create a vicious circle. Many intracellular pathways commonly involved downstream will help maintain and amplify inflammation, oxidative stress, and cancer. Thus, many WNT/β-catenin target genes such as c-Myc, cyclin D1, and HIF-1α are involved in the development of cancers. Nuclear factor-kappaB (NFκB) can activate many inflammatory factors such as TNF-α, TGF-β, interleukin-6 (IL-6), IL-8, MMP, vascular endothelial growth factor, COX2, Bcl2, and inducible nitric oxide synthase. These factors are often associated with cancerous processes and may even promote them. Reactive oxygen species (ROS), generated by cellular alterations, stimulate the production of inflammatory factors such as NFκB, signal transducer and activator transcription, activator protein-1, and HIF-α. NFκB inhibits glycogen synthase kinase-3β (GSK-3β) and therefore activates the canonical WNT pathway. ROS activates the phosphatidylinositol 3 kinase/protein kinase B (PI3K/Akt) signaling in many cancers. PI3K/Akt also inhibits GSK-3β. Many gene mutations of the canonical WNT/β-catenin pathway giving rise to cancers have been reported (CTNNB1, AXIN, APC). Conversely, a significant reduction in the expression of PPARγ has been observed in many cancers. Moreover, PPARγ agonists promote cell cycle arrest, cell differentiation, and apoptosis and reduce inflammation, angiogenesis, oxidative stress, cell proliferation, invasion, and cell migration. All these complex and opposing interactions between the canonical WNT/β-catenin pathway and PPARγ appear to be fairly common in inflammation, oxidative stress, and cancers.
Collapse
Affiliation(s)
| | - Yves Lecarpentier
- Centre de Recherche Clinique, Grand Hôpital de l'Est Francilien (GHEF), Meaux, France
| |
Collapse
|
26
|
Eibl G, Cruz-Monserrate Z, Korc M, Petrov MS, Goodarzi MO, Fisher WE, Habtezion A, Lugea A, Pandol SJ, Hart PA, Andersen DK. Diabetes Mellitus and Obesity as Risk Factors for Pancreatic Cancer. J Acad Nutr Diet 2018; 118:555-567. [PMID: 28919082 PMCID: PMC5845842 DOI: 10.1016/j.jand.2017.07.005] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 07/10/2017] [Indexed: 02/06/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is among the deadliest types of cancer. The worldwide estimates of its incidence and mortality in the general population are eight cases per 100,000 person-years and seven deaths per 100,000 person-years, and they are significantly higher in the United States than in the rest of the world. The incidence of this disease in the United States is more than 50,000 new cases in 2017. Indeed, total deaths due to PDAC are projected to increase dramatically to become the second leading cause of cancer-related deaths before 2030. Considering the failure to date to efficiently treat existing PDAC, increased effort should be undertaken to prevent this disease. A better understanding of the risk factors leading to PDAC development is of utmost importance to identify and formulate preventive strategies. Large epidemiologic and cohort studies have identified risk factors for the development of PDAC, including obesity and type 2 diabetes mellitus. This review highlights the current knowledge of obesity and type 2 diabetes as risk factors for PDAC development and progression, their interplay and underlying mechanisms, and the relation to diet. Research gaps and opportunities to address this deadly disease are also outlined.
Collapse
|
27
|
Semba T, Sugihara E, Kamoshita N, Ueno S, Fukuda K, Yoshino M, Takao K, Yoshikawa K, Izuhara K, Arima Y, Suzuki M, Saya H. Periostin antisense oligonucleotide suppresses bleomycin-induced formation of a lung premetastatic niche for melanoma. Cancer Sci 2018; 109:1447-1454. [PMID: 29498146 PMCID: PMC5980369 DOI: 10.1111/cas.13554] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 02/19/2018] [Accepted: 02/22/2018] [Indexed: 01/12/2023] Open
Abstract
Metastasis is the leading cause of cancer death. A tumor‐supportive microenvironment, or premetastatic niche, at potential secondary tumor sites plays an important role in metastasis, especially in tumor cell colonization. Although a fibrotic milieu is known to promote tumorigenesis and metastasis, the underlying molecular contributors to this effect have remained unclear. Here we show that periostin, a component of the extracellular matrix that functions in tissue remodeling, has a key role in formation of a fibrotic environment that promotes tumor metastatic colonization. We found that periostin was widely expressed in fibrotic lesions of mice with bleomycin‐induced lung fibrosis, and that up‐regulation of periostin expression coincided with activation of myofibroblasts positive for α‐smooth muscle actin. We established a lung metastasis model for B16 murine melanoma cells and showed that metastatic colonization of the lung by these cells was markedly promoted by bleomycin‐induced lung fibrosis. Inhibition of periostin expression by giving an intratracheal antisense oligonucleotide targeting periostin mRNA was found to suppress bleomycin‐induced lung fibrosis and thereby to attenuate metastatic colonization of the lung by melanoma cells. Our results indicate that periostin is a key player in the development of bleomycin‐induced fibrosis and consequent enhancement of tumor cell colonization in the lung. Our results therefore implicate periostin as a potential target for prevention or treatment of lung metastasis.
Collapse
Affiliation(s)
- Takashi Semba
- Division of Gene Regulation, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan.,Department of Thoracic Surgery, Kumamoto University, Kumamoto, Japan
| | - Eiji Sugihara
- Division of Gene Regulation, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan.,Innovation Medical Research Institute, University of Tsukuba, Ibaraki, Japan
| | - Nagisa Kamoshita
- Division of Gene Regulation, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan
| | - Sayaka Ueno
- Division of Gene Regulation, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan
| | - Keitaro Fukuda
- Department of Dermatology, University of Massachusetts Medical School, Worcester, MA, USA
| | | | | | | | - Kenji Izuhara
- Division of Medical Biochemistry, Department of Biomolecular Sciences, Saga Medical School, Saga, Japan
| | - Yoshimi Arima
- Division of Gene Regulation, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan
| | - Makoto Suzuki
- Department of Thoracic Surgery, Kumamoto University, Kumamoto, Japan
| | - Hideyuki Saya
- Division of Gene Regulation, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
28
|
Bałan BJ, Zygmanowska E, Radomska-Leśniewska DM. Disorders noticed during development of pancreatic cancer: potential opportunities for early and effective diagnostics and therapy. Cent Eur J Immunol 2017; 42:377-382. [PMID: 29472816 PMCID: PMC5820973 DOI: 10.5114/ceji.2017.68698] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 08/02/2017] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer, with a total five-year survival rate below 5%, represents a disease with a high level of malignancy. Some of the pancreatic cancer bad prognosis factors are nutrition disorders. Malnutrition, neither recognized nor properly referred to by the healthcare system, leads to well-documented negative health consequences in hospitalized patients including their impaired immunity, delayed post-surgery wound healing, a high risk of infectious complications, morbidity and mortality. There are numerous factors contributing to the development of pancreatic cancer, including telomerases, inflammation, angiogenesis, epigenetics and genetics factors, miRNA, pancreatic cancer stem cells. On the basis of molecular analyses, it has been established that precursor injuries may trigger pancreatic cancer when added to genetic alterations. Perhaps, combination of few presently used methods, like signal transduction modulated by K-ras, STAT3 activation, HMGB1 releasing, presence of oxidative stress and free radicals secretion, genes for proangiogenic growth factors activation or tissue-specific miRNA genes expression - will solve the problem of inadequate diagnostics.
Collapse
Affiliation(s)
- Barbara Joanna Bałan
- Department of Immunology, Biochemistry and Nutrition, Medical University of Warsaw, Poland
| | - Ewa Zygmanowska
- Department of Immunology, Biochemistry and Nutrition, Medical University of Warsaw, Poland
| | | |
Collapse
|
29
|
Acetylsalicylic Acid Produces Different Effects on the Production of Active Oxygen Species by Activated Platelets in Different Inflammatory Diseases. Bull Exp Biol Med 2017; 164:36-40. [PMID: 29124532 DOI: 10.1007/s10517-017-3921-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Indexed: 10/18/2022]
Abstract
We studied the effect of acetylsalicylic acid on ROS generation by platelets in patients after surgical interventions and in patients with bronchial asthma was studied. Platelets stimulated with platelet-activating factor are characterized by weak luminol-enhanced chemiluminescence in healthy people and patients after operations with laparoscopic incisions. Addition of platelet activation factor to platelet samples from patients after open abdominal surgery caused intensive chemiluminescence that was suppressed after platelet incubation with acetylsalicylic acid. At the same time, platelets of patients with aspirin-sensitive asthma did not respond to addition of platelet activating factor, but after incubation with acetylsalicylic acid, an intensive burst of chemiluminescence was detected with a maximum in 5-10 sec after the addition of a platelet-activating factor. In patients with bronchial asthma tolerant to aspirin, platelet activation factor did not induce chemiluminescence irrespective of incubation with acetylsalicylic acid.
Collapse
|
30
|
Huang H, Du W, Brekken RA. Extracellular Matrix Induction of Intracellular Reactive Oxygen Species. Antioxid Redox Signal 2017; 27:774-784. [PMID: 28791881 DOI: 10.1089/ars.2017.7305] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
SIGNIFICANCE The extracellular matrix (ECM) is the noncellular component secreted by cells and is present within all tissues and organs. The ECM provides the structural support required for tissue integrity and also contributes to diseases, including cancer. Many diseases rich in ECM are characterized by changes in reactive oxygen species (ROS) levels that have been shown to have important context-dependent functions. Recent Advances: Many studies have found that the ECM affects ROS production through integrins. The activation of integrins by ECM ligands results in stimulation of multiple pathways that can generate ROS. Furthermore, control of ECM-integrin interaction by matricellular proteins is an underappreciated pathway that functions as an ROS rheostat in remodeling tissues. CRITICAL ISSUES A better understanding of how the ECM affects the generation of intracellular ROS is required for advances in the development of therapeutic strategies that affect or exploit oxidative stress. FUTURE DIRECTIONS Targeting ROS generation can be therapeutic or can promote disease progression in a context-dependent manner. Many ECM proteins can impact ROS generation. However, given the breadth of different proteins that constitute the ECM and the cell surface receptors that interact with ECM proteins, there are likely many tissue and microenvironmental-specific ROS-generating pathways that have yet to be investigated in depth. Identifying canonical pathways of ECM-induced ROS generation should be a priority for the field. Antioxid. Redox Signal. 27, 774-784.
Collapse
Affiliation(s)
- Huocong Huang
- 1 Division of Surgical Oncology, Department of Surgery, Hamon Center for Therapeutic Oncology Research , Dallas, Texas
| | - Wenting Du
- 1 Division of Surgical Oncology, Department of Surgery, Hamon Center for Therapeutic Oncology Research , Dallas, Texas
| | - Rolf A Brekken
- 1 Division of Surgical Oncology, Department of Surgery, Hamon Center for Therapeutic Oncology Research , Dallas, Texas.,2 Department of Pharmacology, UT Southwestern, Dallas, Texas
| |
Collapse
|
31
|
Tomizawa R, Sugiyama H, Sato R, Ohnishi M, Koizumi N. Male-specific pulmonary hemorrhage and cytokine gene expression in golden hamster in early-phase Leptospira interrogans serovar Hebdomadis infection. Microb Pathog 2017; 111:33-40. [PMID: 28811249 DOI: 10.1016/j.micpath.2017.08.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 08/11/2017] [Accepted: 08/11/2017] [Indexed: 01/19/2023]
Abstract
Leptospirosis causes severe clinical signs more frequently in men than in women, but the mechanism underlying the gender differences in leptospirosis remains unclear. In this study, petechial hemorrhage was observed in male but not in female hamster lung tissues infected with Leptospira interrogans serovar Hebdomadis at 120 h pi, demonstrating that male hamsters were more susceptible to the development of a severe disease upon Leptospira infection. No leptospiral DNA was detected in the lung tissues at 120 h pi when pulmonary hemorrhage was observed, indicating that pulmonary hemorrhage is attributable to the immune reactions of the host rather than from the direct effect of leptospires. The upregulation of nitric oxide synthase genes in the hamsters without pulmonary hemorrhage, inos and enos in female hamsters at 96 h pi and enos in male animals without hemorrhage at 120 h pi, may suggest that nitric oxide has a suppressive effect on leptospirosis-associated pulmonary hemorrhage.
Collapse
Affiliation(s)
- Rina Tomizawa
- Graduate School of Bio-Applications & Systems Engineering, Tokyo University of Agriculture and Technology, 2-24-16 Naka-cho, Koganei, Tokyo, 184-8588, Japan; Department of Bacteriology I, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku, Tokyo, 162-8640, Japan.
| | - Hiromu Sugiyama
- Department of Parasitology, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku, Tokyo, 162-8640, Japan.
| | - Ryoichi Sato
- Graduate School of Bio-Applications & Systems Engineering, Tokyo University of Agriculture and Technology, 2-24-16 Naka-cho, Koganei, Tokyo, 184-8588, Japan.
| | - Makoto Ohnishi
- Department of Bacteriology I, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku, Tokyo, 162-8640, Japan.
| | - Nobuo Koizumi
- Department of Bacteriology I, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku, Tokyo, 162-8640, Japan.
| |
Collapse
|
32
|
Coppola S, Carnevale I, Danen EHJ, Peters GJ, Schmidt T, Assaraf YG, Giovannetti E. A mechanopharmacology approach to overcome chemoresistance in pancreatic cancer. Drug Resist Updat 2017; 31:43-51. [PMID: 28867243 DOI: 10.1016/j.drup.2017.07.001] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 07/19/2017] [Indexed: 02/07/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly chemoresistant malignancy. This chemoresistant phenotype has been historically associated with genetic factors. Major biomedical research efforts were concentrated that resulted in the identification of subtypes characterized by specific genetic lesions and gene expression signatures that suggest important biological differences. However, to date, these distinct differences could not be exploited for therapeutic interventions. Apart from these genetic factors, desmoplasia and tumor microenvironment have been recognized as key contributors to PDAC chemoresistance. However, while several strategies targeting tumor-stroma have been explored including drugs against members of the Hedgehog family, they failed to meet the expectations in the clinical setting. These unsatisfactory clinical results suggest that, an important link between genetics and the influence of tumor microenvironment on PDAC chemoresistance remains to be elucidated. In this respect, mechanobiology is an emerging multidisciplinary field that encompasses cell and developmental biology as well as biophysics and bioengineering. Herein we provide a comprehensive overview of the key players in pancreatic cancer chemoresistance from the perspective of mechanobiology, and discuss novel experimental avenues such as elastic micropillar arrays that could provide fresh insights for the development of mechanobiology-targeted therapeutic approaches (know as mechanopharmacology) to overcome anticancer drug resistance in pancreatic cancer.
Collapse
Affiliation(s)
- Stefano Coppola
- Physics of Life Processes, Huygens-Kamerlingh Onnes Laboratory, Leiden University, Leiden, The Netherlands
| | - Ilaria Carnevale
- Department of Medical Oncology, VU University Medical Center Amsterdam, Amsterdam, The Netherlands; Cancer Pharmacology Lab, AIRC Start-Up Unit, University Hospital of Pisa, Pisa, Italy
| | - Erik H J Danen
- Division of Toxicology, LACDR, Leiden University, Leiden, The Netherlands
| | - Godefridus J Peters
- Department of Medical Oncology, VU University Medical Center Amsterdam, Amsterdam, The Netherlands
| | - Thomas Schmidt
- Physics of Life Processes, Huygens-Kamerlingh Onnes Laboratory, Leiden University, Leiden, The Netherlands
| | - Yehuda G Assaraf
- The Fred Wyszkowski Cancer Research Laboratory, Department of Biology, Technion-Israel Institute of Technology, Haifa, Israel
| | - Elisa Giovannetti
- Department of Medical Oncology, VU University Medical Center Amsterdam, Amsterdam, The Netherlands; Cancer Pharmacology Lab, AIRC Start-Up Unit, University Hospital of Pisa, Pisa, Italy; Institute for Nanoscience and Nanotechnologies, CNR-Nano, Pisa.
| |
Collapse
|
33
|
Lachowski D, Cortes E, Pink D, Chronopoulos A, Karim SA, P Morton J, Del Río Hernández AE. Substrate Rigidity Controls Activation and Durotaxis in Pancreatic Stellate Cells. Sci Rep 2017; 7:2506. [PMID: 28566691 PMCID: PMC5451433 DOI: 10.1038/s41598-017-02689-x] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 04/18/2017] [Indexed: 02/06/2023] Open
Abstract
Pancreatic Ductal Adenocarcinoma (PDAC) is an aggressive malignancy characterised by the presence of extensive desmoplasia, thought to be responsible for the poor response of patients to systemic therapies. Pancreatic stellate cells (PSCs) are key mediators in the production of this fibrotic stroma, upon activation transitioning to a myofibroblast-like, high matrix secreting phenotype. Given their importance in disease progression, characterisation of PSC activation has been extensive, however one aspect that has been overlooked is the mechano-sensing properties of the cell. Here, through the use of a physiomimetic system that recapitulates the mechanical microenvironment found within healthy and fibrotic pancreas, we demonstrate that matrix stiffness regulates activation and mechanotaxis in PSCs. We show the ability of PSCs to undergo phenotypic transition solely as a result of changes in extracellular matrix stiffness, whilst observing the ability of PSCs to durotactically respond to stiffness variations within their local environment. Our findings implicate the mechanical microenvironment as a potent contributor to PDAC progression and survival via induction of PSC activation and fibrosis, suggesting that direct mechanical reprogramming of PSCs may be a viable alternative in the treatment of this lethal disease.
Collapse
Affiliation(s)
- Dariusz Lachowski
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London, SW7 2AZ, United Kingdom
| | - Ernesto Cortes
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London, SW7 2AZ, United Kingdom
| | - Daniel Pink
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London, SW7 2AZ, United Kingdom
| | - Antonios Chronopoulos
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London, SW7 2AZ, United Kingdom
| | - Saadia A Karim
- Pancreatic Cancer Research Team, CRUK Beatson Institute, Glasgow, G61 1BD, United Kingdom
| | - Jennifer P Morton
- Pancreatic Cancer Research Team, CRUK Beatson Institute, Glasgow, G61 1BD, United Kingdom
| | - Armando E Del Río Hernández
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London, SW7 2AZ, United Kingdom.
| |
Collapse
|
34
|
Voudouri K, Nikitovic D, Berdiaki A, Kletsas D, Karamanos NK, Tzanakakis GN. IGF-I/EGF and E2 signaling crosstalk through IGF-IR conduit point affects breast cancer cell adhesion. Matrix Biol 2016; 56:95-113. [DOI: 10.1016/j.matbio.2016.06.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Revised: 06/22/2016] [Accepted: 06/22/2016] [Indexed: 12/17/2022]
|
35
|
Durand N, Storz P. Targeting reactive oxygen species in development and progression of pancreatic cancer. Expert Rev Anticancer Ther 2016; 17:19-31. [PMID: 27841037 DOI: 10.1080/14737140.2017.1261017] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
INTRODUCTION Pancreatic ductal adenocarcinoma (PDA) is characterized by expression of oncogenic KRas which drives all aspects of tumorigenesis. Oncogenic KRas induces the formation of reactive oxygen species (ROS) which have been implicated in initiation and progression of PDA. To facilitate tumor promoting levels and to avoid oncogene-induced senescence or cytotoxicity, ROS homeostasis in PDA cells is balanced by additional up-regulation of antioxidant systems. Areas covered: We examine the sources of ROS in PDA, the mechanisms by which ROS homeostasis is maintained, and the biological consequences of ROS in PDA. Additionally, we discuss the potential mechanisms for targeting ROS homoeostasis as a point of therapeutic intervention. An extensive review of the relevant literature as it relates to the topic was conducted using PubMed. Expert commentary: Even though oncogenic mutations in the KRAS gene have been detected in over 95% of human pancreatic adenocarcinoma, targeting its gene product, KRas, has been difficult. The dependency of PDA cells on balancing ROS homeostasis could be an angle for new prevention or treatment strategies. These include use of antioxidants to prevent formation or progression of precancerous lesions, or methods to increase ROS in tumor cells to toxic levels.
Collapse
Affiliation(s)
- Nisha Durand
- a Department of Cancer Biology , Mayo Clinic , Jacksonville , FL , USA
| | - Peter Storz
- a Department of Cancer Biology , Mayo Clinic , Jacksonville , FL , USA
| |
Collapse
|
36
|
Wu Y, Meitzler JL, Antony S, Juhasz A, Lu J, Jiang G, Liu H, Hollingshead M, Haines DC, Butcher D, Panter MS, Roy K, Doroshow JH. Dual oxidase 2 and pancreatic adenocarcinoma: IFN-γ-mediated dual oxidase 2 overexpression results in H2O2-induced, ERK-associated up-regulation of HIF-1α and VEGF-A. Oncotarget 2016; 7:68412-68433. [PMID: 27637085 PMCID: PMC5340089 DOI: 10.18632/oncotarget.12032] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 09/01/2016] [Indexed: 12/18/2022] Open
Abstract
Several NADPH oxidase family members, including dual oxidase 2 [DUOX2], are expressed in human tumors, particularly gastrointestinal cancers associated with long-standing chronic inflammation. We found previously that exposure of pancreatic ductal adenocarcinoma cells to the pro-inflammatory cytokine IFN-γ increased DUOX2 expression (but not other NADPH oxidases) leading to long-lived H2O2 production. To elucidate the pathophysiology of DUOX2-mediated H2O2 formation in the pancreas further, we demonstrate here that IFN-γ-treated BxPC-3 and CFPAC-1 pancreatic cancer cells (known to increase DUOX2 expression) produce significant levels of intracellular oxidants and extracellular H2O2 which correlate with concomitant up-regulation of VEGF-A and HIF-1α transcription. These changes are not observed in the PANC-1 line that does not increase DUOX2 expression following IFN-γ treatment. DUOX2 knockdown with short interfering RNA significantly decreased IFN-γ-induced VEGF-A or HIF-1α up-regulation, as did treatment of pancreatic cancer cells with the NADPH oxidase inhibitor diphenylene iodonium, the multifunctional reduced thiol N-acetylcysteine, and the polyethylene glycol-modified form of the hydrogen peroxide detoxifying enzyme catalase. Increased DUOX2-related VEGF-A expression appears to result from reactive oxygen-mediated activation of ERK signaling that is responsible for AP-1-related transcriptional effects on the VEGF-A promoter. To clarify the relevance of these observations in vivo, we demonstrate that many human pre-malignant pancreatic intraepithelial neoplasms and frank pancreatic cancers express substantial levels of DUOX protein compared to histologically normal pancreatic tissues, and that expression of both DUOX2 and VEGF-A mRNAs is significantly increased in surgically-resected pancreatic cancers compared to the adjacent normal pancreas.
Collapse
Affiliation(s)
- Yongzhong Wu
- Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | | | - Smitha Antony
- Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Agnes Juhasz
- Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Jiamo Lu
- Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Guojian Jiang
- Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Han Liu
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD, USA
| | - Melinda Hollingshead
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD, USA
| | - Diana C. Haines
- Pathology/Histotechnology Laboratory, Leidos, Inc./Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - Donna Butcher
- Pathology/Histotechnology Laboratory, Leidos, Inc./Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - Michaela S. Panter
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD, USA
| | - Krishnendu Roy
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD, USA
| | - James H. Doroshow
- Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD, USA
| |
Collapse
|
37
|
The proinflammatory LTB4/BLT1 signal axis confers resistance to TGF-β1-induced growth inhibition by targeting Smad3 linker region. Oncotarget 2016; 6:41650-66. [PMID: 26497676 PMCID: PMC4747179 DOI: 10.18632/oncotarget.6146] [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: 05/19/2015] [Accepted: 09/30/2015] [Indexed: 01/07/2023] Open
Abstract
Leukotriene B4 (LTB4) is a potent pro-inflammatory eicosanoid that is derived from arachidonic acid, and its signaling is known to have a tumor-promoting role in several cancer types. In this study, we investigated whether enhanced LTB4 signaling confers resistance to the cytostatic transforming growth factor-β1 (TGF-β1) response. We found that LTB4 pretreatment or ectopic expression of BLT1, a high affinity LTB4 receptor, fully abrogated TGF-β1-induced cell cycle arrest and expression of p15INK4B and p27KIP1. Mechanism study revealed that LTB4-mediated suppression of TGF-β1-induced Smad3 activation and growth inhibition was due to enhanced phosphorylation of Smad3 linker region (pSmad3L) through activation of BLT1-NAD(P)H oxidase (NOX)-reactive oxygen species (ROS)-epidermal growth factor receptor (EGFR)-phosphatidylinositol 3-kinase (PI3-K)-extracellular signal-activated kinase1/2 (ERK1/2)-linked signaling cascade. Furthermore, the LTB4/BLT1 signaling pathway leading to pSmad3L was constitutively activated in breast cancer cells and was correlated with TGF-β1-resistant growth of the cells in vitro and in vivo. In human breast cancer tissues, the expression level of pSmad3L (Thr179) had a positive correlation with BLT1 expression. Collectively, our data demonstrate for the first time that the induction of pSmad3L through BLT1-NOX-ROS-EGFR-PI3K-ERK1/2 signaling pathway is a key mechanism by which LTB4 blocks the anti-proliferative responses of TGF-β1, providing a novel mechanistic insight into the connection between enhanced inflammatory signal and cancer cell growth.
Collapse
|
38
|
The Janus-Faced Role of Antioxidants in Cancer Cachexia: New Insights on the Established Concepts. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:9579868. [PMID: 27642498 PMCID: PMC5013212 DOI: 10.1155/2016/9579868] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Revised: 06/28/2016] [Accepted: 07/17/2016] [Indexed: 12/14/2022]
Abstract
Chronic inflammation and excessive loss of skeletal muscle usually occur during cancer cachexia, leading to functional impairment and delaying the cure of cancer. The release of cytokines by tumor promotes the formation of reactive oxygen species (ROS), which in turn regulate catabolic pathways involved in muscle atrophy. ROS also exert a dual role within tumor itself, as they can either promote proliferation and vascularization or induce senescence and apoptosis. Accordingly, previous studies that used antioxidants to modulate these ROS-dependent mechanisms, in cancer and cancer cachexia, have obtained contradictory results, hence the need to gather the main findings of these studies and draw global conclusions in order to stimulate more oriented research in this field. Based on the literature reviewed in this paper, it appears that antioxidant supplementation is (1) beneficial in cancer cachectic patients with antioxidant deficiencies, (2) most likely harmful in cancer patients with adequate antioxidant status (i.e., lung, gastrointestinal, head and neck, and esophageal), and (3) not recommended when undergoing radiotherapy. At the moment, measuring the blood levels of antioxidants may help to identify patients with systemic deficiencies. This approach is simple to realize but could not be a gold standard method for cachexia, as it does not necessarily reflect the redox state in other organs, like muscle.
Collapse
|
39
|
Prasad S, Gupta SC, Tyagi AK. Reactive oxygen species (ROS) and cancer: Role of antioxidative nutraceuticals. Cancer Lett 2016; 387:95-105. [PMID: 27037062 DOI: 10.1016/j.canlet.2016.03.042] [Citation(s) in RCA: 627] [Impact Index Per Article: 69.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 03/22/2016] [Accepted: 03/23/2016] [Indexed: 12/21/2022]
Abstract
Extensive research over the past half a century indicates that reactive oxygen species (ROS) play an important role in cancer. Although low levels of ROS can be beneficial, excessive accumulation can promote cancer. One characteristic of cancer cells that distinguishes them from normal cells is their ability to produce increased numbers of ROS and their increased dependence on an antioxidant defense system. ROS are produced as a byproduct intracellularly by mitochondria and other cellular elements and exogenously by pollutants, tobacco, smoke, drugs, xenobiotics, and radiation. ROS modulate various cell signaling pathways, which are primarily mediated through the transcription factors NF-κB and STAT3, hypoxia-inducible factor-1α, kinases, growth factors, cytokines and other proteins, and enzymes; these pathways have been linked to cellular transformation, inflammation, tumor survival, proliferation, invasion, angiogenesis, and metastasis of cancer. ROS are also associated with epigenetic changes in genes, which is helpful in diagnosing diseases. This review considers the role of ROS in the various stages of cancer development. Finally, we provide evidence that nutraceuticals derived from Mother Nature are highly effective in eliminating cancer cells.
Collapse
Affiliation(s)
- Sahdeo Prasad
- Cytokine Research Laboratory, Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA.
| | - Subash C Gupta
- Cytokine Research Laboratory, Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Amit K Tyagi
- Cytokine Research Laboratory, Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| |
Collapse
|
40
|
Reactive Oxygen Species and Targeted Therapy for Pancreatic Cancer. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:1616781. [PMID: 26881012 PMCID: PMC4735911 DOI: 10.1155/2016/1616781] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Revised: 11/28/2015] [Accepted: 12/07/2015] [Indexed: 01/03/2023]
Abstract
Pancreatic cancer is the fourth leading cause of cancer-related death in the United States. Reactive oxygen species (ROS) are generally increased in pancreatic cancer cells compared with normal cells. ROS plays a vital role in various cellular biological activities including proliferation, growth, apoptosis, and invasion. Besides, ROS participates in tumor microenvironment orchestration. The role of ROS is a doubled-edged sword in pancreatic cancer. The dual roles of ROS depend on the concentration. ROS facilitates carcinogenesis and cancer progression with mild-to-moderate elevated levels, while excessive ROS damages cancer cells dramatically and leads to cell death. Based on the recent knowledge, either promoting ROS generation to increase the concentration of ROS with extremely high levels or enhancing ROS scavenging ability to decrease ROS levels may benefit the treatment of pancreatic cancer. However, when faced with oxidative stress, the antioxidant programs of cancer cells have been activated to help cancer cells to survive in the adverse condition. Furthermore, ROS signaling and antioxidant programs play the vital roles in the progression of pancreatic cancer and in the response to cancer treatment. Eventually, it may be the novel target for various strategies and drugs to modulate ROS levels in pancreatic cancer therapy.
Collapse
|
41
|
Grefte S, Adjobo-Hermans M, Versteeg E, Koopman W, Daamen W. Impaired primary mouse myotube formation on crosslinked type I collagen films is enhanced by laminin and entactin. Acta Biomater 2016; 30:265-276. [PMID: 26555376 DOI: 10.1016/j.actbio.2015.11.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 10/16/2015] [Accepted: 11/06/2015] [Indexed: 02/05/2023]
Abstract
In skeletal muscle, the stem cell niche is important for controlling the quiescent, proliferation and differentiation states of satellite cells, which are key for skeletal muscle regeneration after wounding. It has been shown that type I collagen, often used as 3D-scaffolds for regenerative medicine purposes, impairs myoblast differentiation. This is most likely due to the absence of specific extracellular matrix proteins providing attachment sites for myoblasts and/or myotubes. In this study we investigated the differentiation capacity of primary murine myoblasts on type I collagen films either untreated or modified with elastin, laminin, type IV collagen, laminin/entactin complex, combinations thereof, and Matrigel as a positive control. Additionally, increased reactive oxygen species (ROS) and ROCK signaling might also be involved. To measure ROS levels with live-cell microscopy, fibronectin-coated glass coverslips were additionally coated with type I collagen and Matrigel onto which myoblasts were differentiated. On type I collagen-coated coverslips, myotube formation was impaired while ROS levels were increased. However, anti-oxidant treatment did not enhance myotube formation. ROCK inhibition, which generally improve cellular attachment to uncoated surfaces or type I collagen, enhanced myoblast attachment to type I collagen-coated coverslips and -films, but slightly enhanced myotube formation. Only modification of type I collagen films by Matrigel and a combination of laminin/entactin significantly improved myotube formation. Our results indicate that type I collagen scaffolds can be modified by satellite cell niche factors of which specifically laminin and entactin enhanced myotube formation. This offers a promising approach for regenerative medicine purposes to heal skeletal muscle wounds. STATEMENT OF SIGNIFICANCE In this manuscript we show for the first time that impaired myotube formation on type I collagen scaffolds can be completely restored by modification with laminin and entactin, two extracellular proteins from the satellite cell niche. This offers a promising approach for regenerative medicine approaches to heal skeletal muscle wounds.
Collapse
|
42
|
Topalovski M, Brekken RA. Matrix control of pancreatic cancer: New insights into fibronectin signaling. Cancer Lett 2015; 381:252-8. [PMID: 26742464 DOI: 10.1016/j.canlet.2015.12.027] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 12/18/2015] [Accepted: 12/19/2015] [Indexed: 12/18/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDA) is a highly metastatic disease that resists most current therapies. A defining characteristic of PDA is an intense fibrotic response that promotes tumor cell invasion and chemoresistance. Efforts to understand the complex relationship between the tumor and its extracellular network and to therapeutically perturb tumor-stroma interactions are ongoing. Fibronectin (FN), a provisional matrix protein abundant in PDA stroma but not normal tissues, supports metastatic spread and chemoresistance of this deadly disease. FN also supports angiogenesis, which is required for even hypovascular tumors such as PDA to develop and progress. Targeting components of the tumor stroma, such as FN, can effectively reduce tumor growth and spread while also enhancing delivery of chemotherapy. Here, we review the molecular mechanisms by which FN drives angiogenesis, metastasis and chemoresistance in PDA. In light of these new findings, we also discuss therapeutic strategies to inhibit FN signaling.
Collapse
Affiliation(s)
- Mary Topalovski
- Hamon Center for Therapeutic Oncology Research and the Division of Surgical Oncology, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Rolf A Brekken
- Hamon Center for Therapeutic Oncology Research and the Division of Surgical Oncology, UT Southwestern Medical Center, Dallas, TX 75390, USA; Departments of Surgery and Pharmacology, UT Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
43
|
Wang M, Topalovski M, Toombs JE, Wright CM, Moore ZR, Boothman DA, Yanagisawa H, Wang H, Witkiewicz A, Castrillon DH, Brekken RA. Fibulin-5 Blocks Microenvironmental ROS in Pancreatic Cancer. Cancer Res 2015; 75:5058-69. [PMID: 26577699 DOI: 10.1158/0008-5472.can-15-0744] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 08/15/2015] [Indexed: 12/16/2022]
Abstract
Elevated oxidative stress is an aberration seen in many solid tumors, and exploiting this biochemical difference has the potential to enhance the efficacy of anticancer agents. Homeostasis of reactive oxygen species (ROS) is important for normal cell function, but excessive production of ROS can result in cellular toxicity, and therefore ROS levels must be balanced finely. Here, we highlight the relationship between the extracellular matrix and ROS production by reporting a novel function of the matricellular protein Fibulin-5 (Fbln5). We used genetically engineered mouse models of pancreatic ductal adenocarcinoma (PDAC) and found that mutation of the integrin-binding domain of Fbln5 led to decreased tumor growth, increased survival, and enhanced chemoresponse to standard PDAC therapies. Through mechanistic investigations, we found that improved survival was due to increased levels of oxidative stress in Fbln5-mutant tumors. Furthermore, loss of the Fbln5-integrin interaction augmented fibronectin signaling, driving integrin-induced ROS production in a 5-lipooxygenase-dependent manner. These data indicate that Fbln5 promotes PDAC progression by functioning as a molecular rheostat that modulates cell-ECM interactions to reduce ROS production, and thus tip the balance in favor of tumor cell survival and treatment-refractory disease.
Collapse
Affiliation(s)
- Miao Wang
- Hamon Center for Therapeutic Oncology Research, UT Southwestern Medical Center, Dallas, Texas
| | - Mary Topalovski
- Hamon Center for Therapeutic Oncology Research, UT Southwestern Medical Center, Dallas, Texas
| | - Jason E Toombs
- Hamon Center for Therapeutic Oncology Research, UT Southwestern Medical Center, Dallas, Texas
| | - Christopher M Wright
- Hamon Center for Therapeutic Oncology Research, UT Southwestern Medical Center, Dallas, Texas
| | - Zachary R Moore
- Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, Texas
| | - David A Boothman
- Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, Texas
| | - Hiromi Yanagisawa
- Department of Molecular Biology, UT Southwestern Medical Center, Dallas, Texas
| | - Huamin Wang
- Department of Pathology, UT MD Anderson Cancer Center, Houston, Texas
| | | | | | - Rolf A Brekken
- Hamon Center for Therapeutic Oncology Research, UT Southwestern Medical Center, Dallas, Texas. Department of Surgery, UT Southwestern Medical Center, Dallas, Texas. Department of Pharmacology, UT Southwestern Medical Center, Dallas, Texas.
| |
Collapse
|
44
|
Moir JAG, Mann J, White SA. The role of pancreatic stellate cells in pancreatic cancer. Surg Oncol 2015; 24:232-8. [PMID: 26080604 DOI: 10.1016/j.suronc.2015.05.002] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Accepted: 05/11/2015] [Indexed: 01/05/2023]
Abstract
BACKGROUND The prognosis of pancreatic cancer remains desperately poor, with little progress made over the past 30 years despite the development of new combination chemotherapy regimens. Stromal activity is especially prominent in the tissue surrounding pancreatic tumours, and has a profound influence in dictating tumour development and dissemination. Pancreatic stellate cells (PaSCs) have a key role in this tumour microenvironment, and have been the subject of much research in the past decade. This review examines the relationship between PaSCs and cancer cells. METHODS A comprehensive literature search was performed of multiple databases up to March 2014, including Medline, Pubmed and Google Scholar. RESULTS A complex bidirectional interplay exists between PaSCs and cancer cells, resulting in a perpetuating loop of increased activity and an overriding pro-tumorigenic effect. This involves a number of signalling pathways that also impacts on other stromal components and vasculature, contributing to chemoresistance. The Reverse Warburg Effect is also introduced as a novel concept in tumour stroma. CONCLUSION This review highlights the pancreatic tumour microenvironment, and in particular PaSCs, as an ideal target for therapeutics. There are a number of cellular processes involving PaSCs which could hold the key to more effectively treating pancreatic cancer. The feasibility of targeting these pathways warrant further in depth investigation, with the aim of reducing the aggressiveness of pancreatic cancer and improving chemodelivery.
Collapse
Affiliation(s)
- John A G Moir
- Freeman Hospital, Department of HPB and Transplant Surgery, Newcastle upon Tyne, United Kingdom; Institute of Cellular Medicine, Fibrosis Lab, Newcastle upon Tyne, United Kingdom.
| | - Jelena Mann
- Institute of Cellular Medicine, Fibrosis Lab, Newcastle upon Tyne, United Kingdom
| | - Steve A White
- Freeman Hospital, Department of HPB and Transplant Surgery, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
45
|
Pandol SJ, Edderkaoui M. What are the macrophages and stellate cells doing in pancreatic adenocarcinoma? Front Physiol 2015; 6:125. [PMID: 26029109 PMCID: PMC4432577 DOI: 10.3389/fphys.2015.00125] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 04/07/2015] [Indexed: 12/11/2022] Open
Abstract
Pancreatic ductal adenocarcinoma is a devastating disease characterized by a dense desmoplastic stroma. Chemo- and radio-therapeutic strategies based on targeting cancer cells have failed in improving the outcome of this cancer suggesting important roles for stroma in therapy resistance. Cells in the tumor stroma have been shown to regulate proliferation, resistance to apoptosis and treatments, epithelial to mesenchymal transition (EMT) and stemness of cancer cells. Stellate cells in their activated state have been thought over the past decade to only have tumor promoting roles. However, recent findings suggest that stellate cells may have protective roles as well. The present review highlights the latest findings on the role of two major components of tumor stroma, pancreatic stellate cells and macrophages, in promoting or inhibiting pancreatic cancer, focused on their effects on EMT and cancer stemness.
Collapse
Affiliation(s)
- Stephen J Pandol
- Departments of Medicine and Biological Sciences, Cedars-Sinai Medical Center Los Angeles, CA, USA ; Veterans Affairs Greater Los Angeles Healthcare System and University of California, Los Angeles Los Angeles, CA, USA
| | - Mouad Edderkaoui
- Departments of Medicine and Biological Sciences, Cedars-Sinai Medical Center Los Angeles, CA, USA ; Veterans Affairs Greater Los Angeles Healthcare System and University of California, Los Angeles Los Angeles, CA, USA
| |
Collapse
|
46
|
Thrower E. Pathologic cellular events in smoking-related pancreatitis. Cancers (Basel) 2015; 7:723-35. [PMID: 25938854 PMCID: PMC4491681 DOI: 10.3390/cancers7020723] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 04/17/2015] [Accepted: 04/21/2015] [Indexed: 12/13/2022] Open
Abstract
Pancreatitis, a debilitating inflammatory disorder, results from pancreatic injury. Alcohol abuse is the foremost cause, although cigarette smoking has recently surfaced as a distinct risk factor. The mechanisms by which cigarette smoke and its toxins initiate pathological cellular events leading to pancreatitis, have not been clearly defined. Although cigarette smoke is composed of more than 4000 compounds, it is mainly nicotine and the tobacco-specific nitrosamine 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK), which have been extensively studied with respect to pancreatic diseases. This review summarizes these research findings and highlights cellular pathways which may be of relevance in initiation and progression of smoking-related pancreatitis.
Collapse
Affiliation(s)
- Edwin Thrower
- Department of Internal Medicine, Section of Digestive Diseases, Yale University School of Medicine, New Haven, CT 06520, USA.
- Veterans Affairs Connecticut Healthcare, West Haven, CT 06516, USA .
| |
Collapse
|
47
|
Byun HG, Lee JK. Chlorella ethanol extract induced phase II enzyme through NFE2L2 (nuclear factor [erythroid-derived] 2-like 2, NRF2) activation and protected ethanol-induced hepatoxicity. J Med Food 2015; 18:182-9. [PMID: 25602788 DOI: 10.1089/jmf.2014.3159] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
In this study, we investigated the hepatoprotective effects of ethanol extracts from Chlorella vulgaris (CH) on animals. We measured its effect on the quinone reductase (QR) activity in Hepa1c1c7 cells, finding that CH induced a significantly higher QR activity in these cells. We isolated the active fraction (CH F4-2) from CH using chromatography methods. CH F4-2 may activate cellular antioxidant enzymes through upregulation of the Nrf2 pathway in hepatocarcinoma cells with CH F4-2 (25.0-200 μg/mL) for 48 h. Furthermore, CH F4-2 increased the expression of NQO1 [ NAD(P)H quinone oxidoreductase, also known as QR], heme oxygenase-1, and glutathione-S-transferase P. Moreover, we found that ethanol-induced hepatic pathological changes-elevations in glutamic oxaloacetic transaminase, glutamic pyruvic transaminase, γ-glutamyltransferase, and lactate dehydrogenase-were significantly decreased. The inhibitory effect of CH on alcohol-induced liver injury was associated with the suppression of alcohol-induced increases in intestinal permeability. The ethanol extract from CH was found to induce QR activation, making it a potentially good candidate for a hepatoprotection agent.
Collapse
Affiliation(s)
- Hee-Guk Byun
- Department of Marine Biotechnology, Gangneung-Wonju National University , Gangneung, Korea
| | | |
Collapse
|
48
|
Powell WS, Rokach J. Biosynthesis, biological effects, and receptors of hydroxyeicosatetraenoic acids (HETEs) and oxoeicosatetraenoic acids (oxo-ETEs) derived from arachidonic acid. Biochim Biophys Acta Mol Cell Biol Lipids 2014; 1851:340-55. [PMID: 25449650 DOI: 10.1016/j.bbalip.2014.10.008] [Citation(s) in RCA: 221] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Revised: 10/10/2014] [Accepted: 10/21/2014] [Indexed: 12/14/2022]
Abstract
Arachidonic acid can be oxygenated by a variety of different enzymes, including lipoxygenases, cyclooxygenases, and cytochrome P450s, and can be converted to a complex mixture of oxygenated products as a result of lipid peroxidation. The initial products in these reactions are hydroperoxyeicosatetraenoic acids (HpETEs) and hydroxyeicosatetraenoic acids (HETEs). Oxoeicosatetraenoic acids (oxo-ETEs) can be formed by the actions of various dehydrogenases on HETEs or by dehydration of HpETEs. Although a large number of different HETEs and oxo-ETEs have been identified, this review will focus principally on 5-oxo-ETE, 5S-HETE, 12S-HETE, and 15S-HETE. Other related arachidonic acid metabolites will also be discussed in less detail. 5-Oxo-ETE is synthesized by oxidation of the 5-lipoxygenase product 5S-HETE by the selective enzyme, 5-hydroxyeicosanoid dehydrogenase. It actions are mediated by the selective OXE receptor, which is highly expressed on eosinophils, suggesting that it may be important in eosinophilic diseases such as asthma. 5-Oxo-ETE also appears to stimulate tumor cell proliferation and may also be involved in cancer. Highly selective and potent OXE receptor antagonists have recently become available and could help to clarify its pathophysiological role. The 12-lipoxygenase product 12S-HETE acts by the GPR31 receptor and promotes tumor cell proliferation and metastasis and could therefore be a promising target in cancer therapy. It may also be involved as a proinflammatory mediator in diabetes. In contrast, 15S-HETE may have a protective effect in cancer. In addition to GPCRs, higher concentration of HETEs and oxo-ETEs can activate peroxisome proliferator-activated receptors (PPARs) and could potentially regulate a variety of processes by this mechanism. This article is part of a Special Issue entitled "Oxygenated metabolism of PUFA: analysis and biological relevance".
Collapse
Affiliation(s)
- William S Powell
- Meakins-Christie Laboratories, Department of Medicine, McGill University, 3626St. Urbain Street, Montreal, Quebec H2X 2P2, Canada.
| | - Joshua Rokach
- Claude Pepper Institute and Department of Chemistry, Florida Institute of Technology, 150 West University Boulevard, Melbourne, FL 32901, USA
| |
Collapse
|
49
|
Yu JH, Kim H. Oxidative stress and cytokines in the pathogenesis of pancreatic cancer. J Cancer Prev 2014; 19:97-102. [PMID: 25337577 PMCID: PMC4204162 DOI: 10.15430/jcp.2014.19.2.97] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Revised: 06/17/2014] [Accepted: 06/17/2014] [Indexed: 12/12/2022] Open
Abstract
Pancreatic cancer is one of the most aggressive, drug-resistant and lethal types of cancer with poor prognosis. Various factors including reactive oxygen species, cytokines, growth factors, and extracellular matrix proteins are reported to be involved in the development of pancreatic cancer. However, the pathogenesis of pancreatic cancer has not been completely elucidated. Oxidative stress has been shown to contribute to the development of pancreatic cancer. Evidences supporting the role of reactive oxygen species and cytokines as a risk for pancreatic cancer and the concept of antioxidant supplementation as a preventive approach for pancreatic cancer have been proposed. Here, we review the literature on oxidative stress, cytokine expression, inflammatory signaling, and natural antioxidant supplementation in relation to pancreatic cancer.
Collapse
Affiliation(s)
- Ji Hoon Yu
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, Korea
| | - Hyeyoung Kim
- Department of Food and Nutrition, Brain Korea 21 PLUS Project, College of Human Ecology, Yonsei University, Seoul, Korea
| |
Collapse
|
50
|
Bhattacharyya A, Chattopadhyay R, Mitra S, Crowe SE. Oxidative stress: an essential factor in the pathogenesis of gastrointestinal mucosal diseases. Physiol Rev 2014; 94:329-54. [PMID: 24692350 DOI: 10.1152/physrev.00040.2012] [Citation(s) in RCA: 1510] [Impact Index Per Article: 137.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Reactive oxygen species (ROS) are generated as by-products of normal cellular metabolic activities. Superoxide dismutase, glutathione peroxidase, and catalase are the enzymes involved in protecting cells from the damaging effects of ROS. ROS are produced in response to ultraviolet radiation, cigarette smoking, alcohol, nonsteroidal anti-inflammatory drugs, ischemia-reperfusion injury, chronic infections, and inflammatory disorders. Disruption of normal cellular homeostasis by redox signaling may result in cardiovascular, neurodegenerative diseases and cancer. ROS are produced within the gastrointestinal (GI) tract, but their roles in pathophysiology and disease pathogenesis have not been well studied. Despite the protective barrier provided by the mucosa, ingested materials and microbial pathogens can induce oxidative injury and GI inflammatory responses involving the epithelium and immune/inflammatory cells. The pathogenesis of various GI diseases including peptic ulcers, gastrointestinal cancers, and inflammatory bowel disease is in part due to oxidative stress. Unraveling the signaling events initiated at the cellular level by oxidative free radicals as well as the physiological responses to such stress is important to better understand disease pathogenesis and to develop new therapies to manage a variety of conditions for which current therapies are not always sufficient.
Collapse
|