1
|
Kaffe E, Tisi A, Magkrioti C, Aidinis V, Mehal WZ, Flavell RA, Maccarrone M. Bioactive signalling lipids as drivers of chronic liver diseases. J Hepatol 2024; 80:140-154. [PMID: 37741346 DOI: 10.1016/j.jhep.2023.08.029] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 08/23/2023] [Accepted: 08/28/2023] [Indexed: 09/25/2023]
Abstract
Lipids are important in multiple cellular functions, with most having structural or energy storage roles. However, a small fraction of lipids exert bioactive roles through binding to G protein-coupled receptors and induce a plethora of processes including cell proliferation, differentiation, growth, migration, apoptosis, senescence and survival. Bioactive signalling lipids are potent modulators of metabolism and energy homeostasis, inflammation, tissue repair and malignant transformation. All these events are involved in the initiation and progression of chronic liver diseases. In this review, we focus specifically on the roles of bioactive lipids derived from phospholipids (lyso-phospholipids) and poly-unsaturated fatty acids (eicosanoids, pro-resolving lipid mediators and endocannabinoids) in prevalent chronic liver diseases (alcohol-associated liver disease, non-alcoholic fatty liver disease, viral hepatitis and hepatocellular carcinoma). We discuss the balance between pathogenic and beneficial bioactive lipids as well as potential therapeutic targets related to the agonism or antagonism of their receptors.
Collapse
Affiliation(s)
- Eleanna Kaffe
- Department of Immunobiology, Yale University School of Medicine, 06511, New Haven, CT, USA.
| | - Annamaria Tisi
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100, L'Aquila, Italy
| | | | - Vassilis Aidinis
- Biomedical Sciences Research Center Alexander Fleming, 16672, Athens, Greece
| | - Wajahat Z Mehal
- Department of Internal Medicine, Section of Digestive Diseases, Yale University, New Haven, CT, 06520, USA; Veterans Affairs Medical Center, West Haven, CT, 06516, USA
| | - Richard A Flavell
- Department of Immunobiology, Yale University School of Medicine, 06511, New Haven, CT, USA; Howard Hughes Medical Institute, Yale School of Medicine, New Haven, CT, 06519, USA
| | - Mauro Maccarrone
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100, L'Aquila, Italy; Laboratory of Lipid Neurochemistry, European Center for Brain Research (CERC), Santa Lucia Foundation IRCCS, 00143 Rome, Italy.
| |
Collapse
|
2
|
Hösch NG, Martins BB, Alcantara QA, Bufalo MC, Neto BS, Chudzinki-Tavassi AM, Santa-Cecilia FV, Cury Y, Zambelli VO. Wnt signaling is involved in crotalphine-induced analgesia in a rat model of neuropathic pain. Eur J Pharmacol 2023; 959:176058. [PMID: 37739305 DOI: 10.1016/j.ejphar.2023.176058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 09/13/2023] [Accepted: 09/14/2023] [Indexed: 09/24/2023]
Abstract
The aberrant activation of Wnt/β-catenin and atypical Wnt/Ryk signaling pathways in the spinal cord is critical for the development and maintenance of neuropathic pain. Crotalphine is a structural analog to a peptide first identified in Crotalus durissus terrificus snake venom, which induces antinociception by activating kappa-opioid and CB2 cannabinoid receptors. Consistent with previous data, we showed that the protein levels of the canonical Wnt/β-catenin and the atypical Wnt/Ryk signaling pathways are increased in neuropathic rats. Importantly, the administration of crotalphine downregulates these protein levels, including its downstream cascades, such as TCF4 from the canonical pathway and NR2B glutamatergic receptor and Ca2+-dependent signals, via the Ryk receptor. The CB2 receptor antagonist, AM630, abolished the crotalphine-induced atypical Wnt/Ryk signaling pathway activation. However, the selective CB2 agonist affects both canonical and non-canonical Wnt signaling in the spinal cord. Next, we showed that crotalphine blocked hypersensitivity and significantly decreased the concentration of IL-1ɑ, IL-1β, IL-6, IL-10, IL-18, TNF-ɑ, MIP-1ɑ and MIP-2 induced by intrathecal injection of exogenous Wnt-3a agonist. Taken together, our findings show that crotalphine induces analgesia in a neuropathic pain model by down-regulating the canonical Wnt/β-catenin and the atypical Wnt/Ryk signaling pathways and, consequently controlling neuroinflammation. This effect is, at least in part, mediated by CB2 receptor activation. These results open a perspective for new approaches that can be used to target Wnt signaling in the context of chronic pain. PERSPECTIVE: Our work identified that crotalphine-induced activation of CB2 receptors plays a critical role in the impairment of Wnt signaling during neuropathic pain. This work suggests that drugs with opioid/cannabinoid activity may be a useful strategy to target Wnt signaling in the context of chronic pain.
Collapse
Affiliation(s)
- Natália G Hösch
- Laboratory of Pain and Signaling, Butantan Institute, Av. Vital Brazil, 1500, 05503-900, São Paulo, SP, Brazil; Department of Pharmacology, Institute of Biomedical Science, University of São Paulo, Av. Prof. Lineu Prestes, 1524, 05508-900, São Paulo, Brazil
| | - Bárbara B Martins
- Laboratory of Pain and Signaling, Butantan Institute, Av. Vital Brazil, 1500, 05503-900, São Paulo, SP, Brazil
| | - Queren A Alcantara
- Laboratory of Pain and Signaling, Butantan Institute, Av. Vital Brazil, 1500, 05503-900, São Paulo, SP, Brazil; Department of Biochemistry, University of Utah, Salt Lake City, UT, 84112, USA
| | - Michelle Cristiane Bufalo
- Laboratory of Pain and Signaling, Butantan Institute, Av. Vital Brazil, 1500, 05503-900, São Paulo, SP, Brazil; Center of Excellence in New Target Discovery (CENTD), Butantan Institute, Av. Vital Brazil, 1500, 05503-900, São Paulo, SP, Brazil
| | - Beatriz S Neto
- Laboratory of Pain and Signaling, Butantan Institute, Av. Vital Brazil, 1500, 05503-900, São Paulo, SP, Brazil
| | - Ana Marisa Chudzinki-Tavassi
- Center of Excellence in New Target Discovery (CENTD), Butantan Institute, Av. Vital Brazil, 1500, 05503-900, São Paulo, SP, Brazil; Innovation and Development Laboratory, Innovation and Development Center, Butantan Institute, Av. Vital Brazil, 1500, 05503-900, São Paulo, SP, Brazil
| | - Flávia V Santa-Cecilia
- Laboratory of Pain and Signaling, Butantan Institute, Av. Vital Brazil, 1500, 05503-900, São Paulo, SP, Brazil
| | - Yara Cury
- Laboratory of Pain and Signaling, Butantan Institute, Av. Vital Brazil, 1500, 05503-900, São Paulo, SP, Brazil
| | - Vanessa O Zambelli
- Laboratory of Pain and Signaling, Butantan Institute, Av. Vital Brazil, 1500, 05503-900, São Paulo, SP, Brazil.
| |
Collapse
|
3
|
Tripathy M, Bui A, Henderson J, Sun J, Woods CR, Somani S, Doan T, Louis Sam Titus ASC, Mohan C. FAAH inhibition ameliorates breast cancer in a murine model. Oncotarget 2023; 14:910-918. [PMID: 37921652 PMCID: PMC10624203 DOI: 10.18632/oncotarget.28534] [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: 05/23/2023] [Accepted: 09/21/2023] [Indexed: 11/04/2023] Open
Abstract
Breast cancer is the leading cancer among females worldwide. Disease outcome depends on the hormonal status of the cancer and whether or not it is metastatic, but there is a need for more efficacious therapeutic strategies where first line treatment fails. In this study, Fatty Acid Amide Hydrolase (FAAH) inhibition and endocannabinoids were examined as therapeutic alternatives. FAAH is an integral membrane enzyme that hydrolyzes endocannabinoids, rendering them inactive, and FAAH inhibition is predicted to increase cancer cell death. To test this, breast cancer cells were probed for FAAH expression using Western blot analysis, treated with FAAH inhibitors, exogenous endocannabinoids, and combinations of the two treatments, and assessed for viability. High levels of FAAH were observed in different breast cancer cell lines. FAAH inhibition was more effective than exogenous endocannabinoid treatment, and the combination of FAAH inhibitors and endocannabinoids was the most effective in inducing apoptosis of breast cancer cells in vitro. In addition, in vivo FAAH inhibition reduced breast cancer growth in immunodeficient mice. FAAH inhibition is a promising approach, and tremendous progress has been made in the field to validate this mechanism as an alternative to chemotherapy. Further research exploring the therapeutic potential and impact of FAAH expression on cancer cells is warranted.
Collapse
Affiliation(s)
- Mallika Tripathy
- Department Biomedical Engineering, University of Houston, Houston, TX 77204, USA
| | - Amy Bui
- Department Biomedical Engineering, University of Houston, Houston, TX 77204, USA
| | - Jared Henderson
- Department Biomedical Engineering, University of Houston, Houston, TX 77204, USA
| | - Jeffrey Sun
- Department Biomedical Engineering, University of Houston, Houston, TX 77204, USA
| | | | - Soumya Somani
- Department Biomedical Engineering, University of Houston, Houston, TX 77204, USA
| | - Thao Doan
- Department Biomedical Engineering, University of Houston, Houston, TX 77204, USA
| | | | - Chandra Mohan
- Department Biomedical Engineering, University of Houston, Houston, TX 77204, USA
| |
Collapse
|
4
|
Castro MV, Barbero GA, Máscolo P, Villanueva MB, Nsengimana J, Newton-Bishop J, Illescas E, Quezada MJ, Lopez-Bergami P. ROR2 promotes epithelial-mesenchymal transition by hyperactivating ERK in melanoma. J Cell Commun Signal 2023; 17:75-88. [PMID: 35723796 PMCID: PMC10030744 DOI: 10.1007/s12079-022-00683-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 05/23/2022] [Indexed: 12/19/2022] Open
Abstract
Receptor tyrosine kinase-like orphan receptor 2 (ROR2) is a protein with important functions during embryogenesis that is dysregulated in human cancer. An intriguing feature of this receptor is that it plays opposite roles in different tumor types either promoting or inhibiting tumor progression. Understanding the complex role of this receptor requires a more profound exploration of both the altered biological and molecular mechanisms. Here, we describe that ROR2 promotes Epithelial-Mesenchymal Transition (EMT) by inducing cadherin switch and the upregulation of the transcription factors ZEB1, Twist, Slug, Snail, and HIF1A, together with a mesenchymal phenotype and increased migration. We show that ROR2 activates both p38 and ERK mitogen-activated protein kinase pathways independently of Wnt5a. Further, we demonstrated that the upregulation of EMT-related proteins depends on the hyperactivation of the ERK pathway far above the typical high constitutive activity observed in melanoma. In addition, ROR2 also promoted ERK phosphorylation, EMT, invasion, and necrosis in xenotransplanted mice. ROR2 also associates with EMT in tumor samples from melanoma patients where analysis of large cohorts revealed that increased ROR2 levels are linked to EMT signatures. This important role of ROR2 translates into melanoma patient' s prognosis since elevated ROR2 levels reduced overall survival and distant metastasis-free survival of patients with lymph node metastasis. In sum, these results demonstrate that ROR2 contributes to melanoma progression by inducing EMT and necrosis and can be an attractive therapeutic target for melanoma.
Collapse
Affiliation(s)
- María Victoria Castro
- Centro de Estudios Biomédicos, Básicos, Aplicados y Desarrollo (CEBBAD), Universidad Maimónides, Hidalgo 775, 6th Floor, Lab 602., 1405, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), 1425, Buenos Aires, Argentina
| | - Gastón Alexis Barbero
- Centro de Estudios Biomédicos, Básicos, Aplicados y Desarrollo (CEBBAD), Universidad Maimónides, Hidalgo 775, 6th Floor, Lab 602., 1405, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), 1425, Buenos Aires, Argentina
| | - Paula Máscolo
- Centro de Estudios Biomédicos, Básicos, Aplicados y Desarrollo (CEBBAD), Universidad Maimónides, Hidalgo 775, 6th Floor, Lab 602., 1405, Buenos Aires, Argentina
| | - María Belén Villanueva
- Centro de Estudios Biomédicos, Básicos, Aplicados y Desarrollo (CEBBAD), Universidad Maimónides, Hidalgo 775, 6th Floor, Lab 602., 1405, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), 1425, Buenos Aires, Argentina
| | - Jérémie Nsengimana
- Biostatistics Research Group, Population Health Sciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | | | - Edith Illescas
- Centro de Estudios Biomédicos, Básicos, Aplicados y Desarrollo (CEBBAD), Universidad Maimónides, Hidalgo 775, 6th Floor, Lab 602., 1405, Buenos Aires, Argentina
| | - María Josefina Quezada
- Centro de Estudios Biomédicos, Básicos, Aplicados y Desarrollo (CEBBAD), Universidad Maimónides, Hidalgo 775, 6th Floor, Lab 602., 1405, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), 1425, Buenos Aires, Argentina
| | - Pablo Lopez-Bergami
- Centro de Estudios Biomédicos, Básicos, Aplicados y Desarrollo (CEBBAD), Universidad Maimónides, Hidalgo 775, 6th Floor, Lab 602., 1405, Buenos Aires, Argentina.
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), 1425, Buenos Aires, Argentina.
| |
Collapse
|
5
|
Disorders of cancer metabolism: The therapeutic potential of cannabinoids. Biomed Pharmacother 2023; 157:113993. [PMID: 36379120 DOI: 10.1016/j.biopha.2022.113993] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 11/07/2022] [Accepted: 11/07/2022] [Indexed: 11/13/2022] Open
Abstract
Abnormal energy metabolism, as one of the important hallmarks of cancer, was induced by multiple carcinogenic factors and tumor-specific microenvironments. It comprises aerobic glycolysis, de novo lipid biosynthesis, and glutamine-dependent anaplerosis. Considering that metabolic reprogramming provides various nutrients for tumor survival and development, it has been considered a potential target for cancer therapy. Cannabinoids have been shown to exhibit a variety of anticancer activities by unclear mechanisms. This paper first reviews the recent progress of related signaling pathways (reactive oxygen species (ROS), AMP-activated protein kinase (AMPK), mitogen-activated protein kinases (MAPK), phosphoinositide 3-kinase (PI3K), hypoxia-inducible factor-1alpha (HIF-1α), and p53) mediating the reprogramming of cancer metabolism (including glucose metabolism, lipid metabolism, and amino acid metabolism). Then we comprehensively explore the latest discoveries and possible mechanisms of the anticancer effects of cannabinoids through the regulation of the above-mentioned related signaling pathways, to provide new targets and insights for cancer prevention and treatment.
Collapse
|
6
|
Cherkasova V, Wang B, Gerasymchuk M, Fiselier A, Kovalchuk O, Kovalchuk I. Use of Cannabis and Cannabinoids for Treatment of Cancer. Cancers (Basel) 2022; 14:5142. [PMID: 36291926 PMCID: PMC9600568 DOI: 10.3390/cancers14205142] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 10/03/2022] [Accepted: 10/17/2022] [Indexed: 07/26/2023] Open
Abstract
The endocannabinoid system (ECS) is an ancient homeostasis mechanism operating from embryonic stages to adulthood. It controls the growth and development of many cells and cell lineages. Dysregulation of the components of the ECS may result in uncontrolled proliferation, adhesion, invasion, inhibition of apoptosis and increased vascularization, leading to the development of various malignancies. Cancer is the disease of uncontrolled cell division. In this review, we will discuss whether the changes to the ECS are a cause or a consequence of malignization and whether different tissues react differently to changes in the ECS. We will discuss the potential use of cannabinoids for treatment of cancer, focusing on primary outcome/care-tumor shrinkage and eradication, as well as secondary outcome/palliative care-improvement of life quality, including pain, appetite, sleep, and many more factors. Finally, we will complete this review with the chapter on sex- and gender-specific differences in ECS and response to cannabinoids, and equality of the access to treatments with cannabinoids.
Collapse
Affiliation(s)
- Viktoriia Cherkasova
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada
| | - Bo Wang
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada
| | - Marta Gerasymchuk
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada
| | - Anna Fiselier
- Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Olga Kovalchuk
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada
| | - Igor Kovalchuk
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada
| |
Collapse
|
7
|
Iozzo M, Sgrignani G, Comito G, Chiarugi P, Giannoni E. Endocannabinoid System and Tumour Microenvironment: New Intertwined Connections for Anticancer Approaches. Cells 2021; 10:cells10123396. [PMID: 34943903 PMCID: PMC8699381 DOI: 10.3390/cells10123396] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 11/24/2021] [Accepted: 11/30/2021] [Indexed: 01/01/2023] Open
Abstract
The tumour microenvironment (TME) is now recognised as a hallmark of cancer, since tumour:stroma crosstalk supports the key steps of tumour growth and progression. The dynamic co-evolution of the tumour and stromal compartments may alter the surrounding microenvironment, including the composition in metabolites and signalling mediators. A growing number of evidence reports the involvement of the endocannabinoid system (ECS) in cancer. ECS is composed by a complex network of ligands, receptors, and enzymes, which act in synergy and contribute to several physiological but also pathological processes. Several in vitro and in vivo evidence show that ECS deregulation in cancer cells affects proliferation, migration, invasion, apoptosis, and metastatic potential. Although it is still an evolving research, recent experimental evidence also suggests that ECS can modulate the functional behaviour of several components of the TME, above all the immune cells, endothelial cells and stromal components. However, the role of ECS in the tumour:stroma interplay remains unclear and research in this area is particularly intriguing. This review aims to shed light on the latest relevant findings of the tumour response to ECS modulation, encouraging a more in-depth analysis in this field. Novel discoveries could be promising for novel anti-tumour approaches, targeting the microenvironmental components and the supportive tumour:stroma crosstalk, thereby hindering tumour development.
Collapse
|
8
|
Castro MV, Barbero GA, Villanueva MB, Grumolato L, Nsengimana J, Newton-Bishop J, Illescas E, Quezada MJ, Lopez-Bergami P. ROR2 has a protective role in melanoma by inhibiting Akt activity, cell-cycle progression, and proliferation. J Biomed Sci 2021; 28:76. [PMID: 34774050 PMCID: PMC8590781 DOI: 10.1186/s12929-021-00776-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 11/07/2021] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Receptor tyrosine kinase-like orphan receptor 2 (ROR2) is a Wnt5a receptor aberrantly expressed in cancer that was shown to either suppress or promote carcinogenesis in different tumor types. Our goal was to study the role of ROR2 in melanoma. METHODS Gain and loss-of-function strategies were applied to study the biological function of ROR2 in melanoma. Proliferation assays, flow cytometry, and western blotting were used to evaluate cell proliferation and changes in expression levels of cell-cycle and proliferation markers. The role of ROR2 in tumor growth was assessed in xenotransplantation experiments followed by immunohistochemistry analysis of the tumors. The role of ROR2 in melanoma patients was assessed by analysis of clinical data from the Leeds Melanoma Cohort. RESULTS Unlike previous findings describing ROR2 as an oncogene in melanoma, we describe that ROR2 prevents tumor growth by inhibiting cell-cycle progression and the proliferation of melanoma cells. The effect of ROR2 is mediated by inhibition of Akt phosphorylation and activity which, in turn, regulates the expression, phosphorylation, and localization of major cell-cycle regulators including cyclins (A, B, D, and E), CDK1, CDK4, RB, p21, and p27. Xenotransplantation experiments demonstrated that ROR2 also reduces proliferation in vivo, resulting in inhibition of tumor growth. In agreement with these findings, a higher ROR2 level favors thin and non-ulcerated primary melanomas with reduced mitotic rate and better prognosis. CONCLUSION We conclude that the expression of ROR2 slows down the growth of primary tumors and contributes to prolonging melanoma survival. Our results demonstrate that ROR2 has a far more complex role than originally described.
Collapse
Affiliation(s)
- María Victoria Castro
- grid.440480.c0000 0000 9361 4204Centro de Estudios Biomédicos, Básicos, Aplicados y Desarrollo (CEBBAD), Universidad Maimónides, 1405 Buenos Aires, Argentina ,grid.423606.50000 0001 1945 2152Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), 1425 Buenos Aires, Argentina
| | - Gastón Alexis Barbero
- grid.440480.c0000 0000 9361 4204Centro de Estudios Biomédicos, Básicos, Aplicados y Desarrollo (CEBBAD), Universidad Maimónides, 1405 Buenos Aires, Argentina ,grid.423606.50000 0001 1945 2152Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), 1425 Buenos Aires, Argentina
| | - María Belén Villanueva
- grid.440480.c0000 0000 9361 4204Centro de Estudios Biomédicos, Básicos, Aplicados y Desarrollo (CEBBAD), Universidad Maimónides, 1405 Buenos Aires, Argentina ,grid.423606.50000 0001 1945 2152Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), 1425 Buenos Aires, Argentina
| | - Luca Grumolato
- grid.10400.350000 0001 2108 3034INSERM U982, Institute for Research and Innovation in Biomedicine, University of Rouen, 76183 Rouen, France
| | - Jérémie Nsengimana
- grid.1006.70000 0001 0462 7212Biostatistics Research Group, Population Health Sciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, NE2 4HH UK
| | | | - Edith Illescas
- grid.440480.c0000 0000 9361 4204Centro de Estudios Biomédicos, Básicos, Aplicados y Desarrollo (CEBBAD), Universidad Maimónides, 1405 Buenos Aires, Argentina
| | - María Josefina Quezada
- grid.440480.c0000 0000 9361 4204Centro de Estudios Biomédicos, Básicos, Aplicados y Desarrollo (CEBBAD), Universidad Maimónides, 1405 Buenos Aires, Argentina ,grid.423606.50000 0001 1945 2152Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), 1425 Buenos Aires, Argentina
| | - Pablo Lopez-Bergami
- Centro de Estudios Biomédicos, Básicos, Aplicados y Desarrollo (CEBBAD), Universidad Maimónides, 1405, Buenos Aires, Argentina. .,Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), 1425, Buenos Aires, Argentina. .,Centro de Estudios Biomédicos, Biotecnológicos, Ambientales y Diagnóstico, Universidad Maimonides, Hidalgo 775, 6th Floor, Lab 602., 1405, Buenos Aires, Argentina.
| |
Collapse
|
9
|
Frampton G, Reddy P, Jefferson B, Ali M, Khan D, McMillin M. Inhibition of thrombospondin-1 reduces glutathione activity and worsens acute liver injury during acetaminophen hepatotoxicity in mice. Toxicol Appl Pharmacol 2020; 409:115323. [PMID: 33176120 PMCID: PMC8364670 DOI: 10.1016/j.taap.2020.115323] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 11/01/2020] [Accepted: 11/04/2020] [Indexed: 01/07/2023]
Abstract
Acetaminophen (N-Acetyl-p-Aminophenol or APAP)-induced hepatotoxicity is the most common cause of acute liver failure in the United States and Western Europe. Previous studies have shown that TGFβ1 is elevated during APAP-induced hepatotoxicity and promotes liver injury by reducing liver regeneration while inducing hepatocyte senescence. At this time, little is known about the role of proteins that activate latent TGFβ1 and their effects during APAP-induced hepatotoxicity. Thrombospondin-1 (TSP1) is a homotrimeric protein that can not only activate latent TGFβ1 but can also interact with other proteins including Nrf2 to induce antioxidant signaling. The aim of the current study was to assess the role of thrombospondin-1 (TSP1) in both TGFβ1 activation and its contribution to APAP-induced liver injury. C57Bl/6 mice or TSP1 null mice (TSP1-/-) were administered 300 mg/kg or 600 mg/kg of APAP. TGFβ1 signaling, TSP1 expression, measures of hepatic injury, Nrf2 expression, measures of oxidative/nitrosative stress and GSH metabolism were assessed. The expression of TGFβ1, TSP1 and phosphorylation of SMAD proteins increased in APAP-treated mice compared to controls. TSP1-/- mice had reduced TGFβ1 expression and phosphorylation of SMAD proteins but increased liver injury. Hepatocyte cell death was increased in TSP1-/- mice and this was associated with decreased Nrf2 activity, decreased GSH levels and increased oxidative stress in comparison to wild-type C57Bl/6 mice. Together, these data demonstrate that elimination of TSP1 protein in APAP-treated mice reduces TGFβ1 signaling but leads to increased liver injury by reducing Nrf2 expression and GSH activity, ultimately resulting in increased cell death.
Collapse
Affiliation(s)
- Gabriel Frampton
- Central Texas Veterans Health Care System, Austin, TX, United States of America; The University of Texas at Austin Dell Medical School, Department of Internal Medicine, Austin, TX, United States of America
| | - Priyanka Reddy
- Central Texas Veterans Health Care System, Austin, TX, United States of America
| | - Brandi Jefferson
- Central Texas Veterans Health Care System, Austin, TX, United States of America
| | - Malaika Ali
- Central Texas Veterans Health Care System, Austin, TX, United States of America
| | - Durreshahwar Khan
- Central Texas Veterans Health Care System, Austin, TX, United States of America
| | - Matthew McMillin
- Central Texas Veterans Health Care System, Austin, TX, United States of America; The University of Texas at Austin Dell Medical School, Department of Internal Medicine, Austin, TX, United States of America.
| |
Collapse
|
10
|
Ghorbani M, Shahabi P, Karimi P, Soltani-Zangbar H, Morshedi M, Bani S, Jafarzadehgharehziaaddin M, Sadeghzadeh-Oskouei B, Ahmadalipour A. Impacts of epidural electrical stimulation on Wnt signaling, FAAH, and BDNF following thoracic spinal cord injury in rat. J Cell Physiol 2020; 235:9795-9805. [PMID: 32488870 DOI: 10.1002/jcp.29793] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 04/30/2020] [Accepted: 05/05/2020] [Indexed: 12/14/2022]
Abstract
Electrical stimulation (ES) has been shown to improve some of impairments after spinal cord injury (SCI), but the underlying mechanisms remain unclear. The Wnt signaling pathways and the endocannabinoid system appear to be modulated in response to SCI. This study aimed to investigate the effect of ES therapy on the activity of canonical/noncanonical Wnt signaling pathways, brain-derived neurotrophic factor (BDNF), and fatty-acid amide hydrolase (FAAH), which regulate endocannabinoids levels. Forty male Wistar rats were randomly divided into four groups: (a) Sham, (b) laminectomy + epidural subthreshold ES, (c) SCI, and (d) SCI + epidural subthreshold ES. A moderate contusion SCI was performed at the thoracic level (T10). Epidural subthreshold ES was delivered to upper the level of T10 segment every day (1 hr/rat) for 2 weeks. Then, animals were killed and immunoblotting was used to assess spinal cord parameters. Results revealed that ES intervention for 14 days could significantly increase wingless-type3 (Wnt3), Wnt7, β-catenin, Nestin, and cyclin D1 levels, as well as phosphorylation of glycogen synthase kinase 3β and Jun N-terminal kinase. Additionally, SCI reduced BDNF and FAAH levels, and ES increased BDNF and FAAH levels in the injury site. We propose that ES therapy may improve some of impairments after SCI through Wnt signaling pathways. Outcomes also suggest that BDNF and FAAH are important players in the beneficial impacts of ES therapy. However, the precise mechanism of BDNF, FAAH, and Wnt signaling pathways on SCI requires further investigation.
Collapse
Affiliation(s)
- Meysam Ghorbani
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz, Iran.,Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Parviz Shahabi
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz, Iran
| | - Pouran Karimi
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamid Soltani-Zangbar
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Morshedi
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz, Iran.,Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Soheila Bani
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz, Iran
| | | | | | - Ali Ahmadalipour
- Research Center of Psychiatry and Behavioral Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.,Faculty of Medicine, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
11
|
Jefferson B, Ali M, Grant S, Frampton G, Ploof M, Andry S, DeMorrow S, McMillin M. Thrombospondin-1 Exacerbates Acute Liver Failure and Hepatic Encephalopathy Pathology in Mice by Activating Transforming Growth Factor β1. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 190:347-357. [PMID: 31734229 PMCID: PMC7013272 DOI: 10.1016/j.ajpath.2019.10.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 09/28/2019] [Accepted: 10/21/2019] [Indexed: 12/11/2022]
Abstract
Severe hepatic insults can lead to acute liver failure and hepatic encephalopathy (HE). Transforming growth factor β1 (TGFβ1) has been shown to contribute to HE during acute liver failure; however, TGFβ1 must be activated to bind its receptor and generate downstream effects. One protein that can activate TGFβ1 is thrombospondin-1 (TSP-1). Therefore, the aim of this study was to assess TSP-1 during acute liver failure and HE pathogenesis. C57Bl/6 or TSP-1 knockout (TSP-1-/-) mice were injected with azoxymethane (AOM) to induce acute liver failure and HE. Liver damage, neurologic decline, and molecular analyses of TSP-1 and TGFβ1 signaling were performed. AOM-treated mice had increased TSP-1 and TGFβ1 mRNA and protein expression in the liver. TSP-1-/- mice administered AOM had reduced liver injury as assessed by histology and serum transaminase levels compared with C57Bl/6 AOM-treated mice. TSP-1-/- mice treated with AOM had reduced TGFβ1 signaling that was associated with less hepatic cell death as assessed by terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling staining and cleaved caspase 3 expression. TSP-1-/- AOM-treated mice had a reduced rate of neurologic decline, less cerebral edema, and a decrease in microglia activation in comparison with C57Bl/6 mice treated with AOM. Taken together, TSP-1 is an activator of TGFβ1 signaling during AOM-induced acute liver failure and contributes to both liver pathology and HE progression.
Collapse
Affiliation(s)
| | - Malaika Ali
- Central Texas Veterans Health Care System, Austin, Texas
| | - Stephanie Grant
- Department of Medical Physiology, Texas A&M University Health Science Center, Temple, Texas; Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, Texas
| | - Gabriel Frampton
- Department of Medical Physiology, Texas A&M University Health Science Center, Temple, Texas; Department of Internal Medicine, The University of Texas at Austin Dell Medical School, Austin, Texas
| | - Michaela Ploof
- Central Texas Veterans Health Care System, Austin, Texas
| | - Sarah Andry
- Department of Internal Medicine, Baylor Scott & White Health, Temple, Texas
| | - Sharon DeMorrow
- Central Texas Veterans Health Care System, Austin, Texas; Department of Medical Physiology, Texas A&M University Health Science Center, Temple, Texas; Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, Texas; Department of Internal Medicine, The University of Texas at Austin Dell Medical School, Austin, Texas
| | - Matthew McMillin
- Central Texas Veterans Health Care System, Austin, Texas; Department of Internal Medicine, The University of Texas at Austin Dell Medical School, Austin, Texas.
| |
Collapse
|
12
|
Laezza C, Pagano C, Navarra G, Pastorino O, Proto MC, Fiore D, Piscopo C, Gazzerro P, Bifulco M. The Endocannabinoid System: A Target for Cancer Treatment. Int J Mol Sci 2020; 21:ijms21030747. [PMID: 31979368 PMCID: PMC7037210 DOI: 10.3390/ijms21030747] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Revised: 01/17/2020] [Accepted: 01/20/2020] [Indexed: 12/15/2022] Open
Abstract
In recent years, the endocannabinoid system has received great interest as a potential therapeutic target in numerous pathological conditions. Cannabinoids have shown an anticancer potential by modulating several pathways involved in cell growth, differentiation, migration, and angiogenesis. However, the therapeutic efficacy of cannabinoids is limited to the treatment of chemotherapy-induced symptoms or cancer pain, but their use as anticancer drugs in chemotherapeutic protocols requires further investigation. In this paper, we reviewed the role of cannabinoids in the modulation of signaling mechanisms implicated in tumor progression.
Collapse
Affiliation(s)
- Chiara Laezza
- Institute of Endocrinology and Experimental Oncology, IEOS CNR, 80131 Naples, Italy
- Correspondence: (C.L.); (M.B.)
| | - Cristina Pagano
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, 80131 Naples, Italy; (C.P.); (G.N.); (O.P.)
| | - Giovanna Navarra
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, 80131 Naples, Italy; (C.P.); (G.N.); (O.P.)
| | - Olga Pastorino
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, 80131 Naples, Italy; (C.P.); (G.N.); (O.P.)
| | - Maria Chiara Proto
- Department of Pharmacy, University of Salerno, 84084 Fisciano (SA), Italy; (M.C.P.); (D.F.); (C.P.)
| | - Donatella Fiore
- Department of Pharmacy, University of Salerno, 84084 Fisciano (SA), Italy; (M.C.P.); (D.F.); (C.P.)
| | - Chiara Piscopo
- Department of Pharmacy, University of Salerno, 84084 Fisciano (SA), Italy; (M.C.P.); (D.F.); (C.P.)
| | - Patrizia Gazzerro
- Department of Pharmacy, University of Salerno, 84084 Fisciano (SA), Italy; (M.C.P.); (D.F.); (C.P.)
| | - Maurizio Bifulco
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, 80131 Naples, Italy; (C.P.); (G.N.); (O.P.)
- Correspondence: (C.L.); (M.B.)
| |
Collapse
|
13
|
Tomko A, O'Leary L, Trask H, Achenbach JC, Hall SR, Goralski KB, Ellis LD, Dupré DJ. Antitumor Activity of Abnormal Cannabidiol and Its Analog O-1602 in Taxol-Resistant Preclinical Models of Breast Cancer. Front Pharmacol 2019; 10:1124. [PMID: 31611800 PMCID: PMC6777324 DOI: 10.3389/fphar.2019.01124] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 08/30/2019] [Indexed: 12/31/2022] Open
Abstract
Cannabinoids exhibit anti-inflammatory and antitumorigenic properties. Contrary to most cannabinoids present in the Cannabis plant, some, such as O-1602 and abnormal cannabidiol, have no or only little affinity to the CB1 or CB2 cannabinoid receptors and instead exert their effects through other receptors. Here, we investigated whether the synthetic regioisomers of cannabidiol, abnormal cannabidiol, and a closely related compound, O-1602, display antitumorigenic effects in cellular models of breast cancer and whether it could reduce tumorigenesis in vivo. Several studies have shown the effects of cannabinoids on chemotherapy-sensitive breast cancer cell lines, but less is known about the antitumorigenic effects of cannabinoids in chemotherapy-resistant cell lines. Paclitaxel-resistant MDA-MB-231 and MCF-7 breast cancer cell lines were used to study the effect of O-1602 and abnormal cannabidiol on viability, apoptosis, and migration. The effects of O-1602 and abnormal cannabidiol on cell viability were completely blocked by the combination of GPR55 and GPR18-specific siRNAs. Both O-1602 and abnormal cannabidiol decreased viability in paclitaxel-resistant breast cancer cells in a concentration-dependent manner through induction of apoptosis. The effect of these cannabinoids on tumor growth in vivo was studied in a zebrafish xenograft model. In this model, treatment with O-1602 and abnormal cannabidiol (2 µM) significantly reduced tumor growth. Our results suggest that atypical cannabinoids, like O-1602 and abnormal cannabidiol, exert antitumorigenic effects on paclitaxel-resistant breast cancer cells. Due to their lack of central sedation and psychoactive effects, these atypical cannabinoids could represent new leads for the development of additional anticancer treatments when resistance to conventional chemotherapy occurs during the treatment of breast and possibly other cancers.
Collapse
Affiliation(s)
- Andrea Tomko
- Department of Pharmacology, Faculty of Medicine, Dalhousie University, Halifax, Canada
| | - Lauren O'Leary
- Department of Pharmacology, Faculty of Medicine, Dalhousie University, Halifax, Canada
| | - Hilary Trask
- Department of Pharmacology, Faculty of Medicine, Dalhousie University, Halifax, Canada
| | - John C Achenbach
- Aquatic and Crop Resource Development Research Center, National Research Council of Canada, Halifax, Canada
| | - Steven R Hall
- Department of Pharmacology, Faculty of Medicine, Dalhousie University, Halifax, Canada.,College of Pharmacy, Faculty of Health, Dalhousie University, Halifax, Canada
| | - Kerry B Goralski
- Department of Pharmacology, Faculty of Medicine, Dalhousie University, Halifax, Canada.,College of Pharmacy, Faculty of Health, Dalhousie University, Halifax, Canada
| | - Lee D Ellis
- College of Pharmacy, Faculty of Health, Dalhousie University, Halifax, Canada
| | - Denis J Dupré
- Department of Pharmacology, Faculty of Medicine, Dalhousie University, Halifax, Canada
| |
Collapse
|
14
|
An Autocrine Wnt5a Loop Promotes NF-κB Pathway Activation and Cytokine/Chemokine Secretion in Melanoma. Cells 2019; 8:cells8091060. [PMID: 31510045 PMCID: PMC6770184 DOI: 10.3390/cells8091060] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 08/29/2019] [Accepted: 09/05/2019] [Indexed: 12/19/2022] Open
Abstract
Wnt5a signaling has been implicated in the progression of cancer by regulating multiple cellular processes, largely migration and invasion, epithelial-mesenchymal transition (EMT), and metastasis. Since Wnt5a signaling has also been involved in inflammatory processes in infectious and inflammatory diseases, we addressed the role of Wnt5a in regulating NF-κB, a pivotal mediator of inflammatory responses, in the context of cancer. The treatment of melanoma cells with Wnt5a induced phosphorylation of the NF-κB subunit p65 as well as IKK phosphorylation and IκB degradation. By using cDNA overexpression, RNA interference, and dominant negative mutants we determined that ROR1, Dvl2, and Akt (from the Wnt5a pathway) and TRAF2 and RIP (from the NF-κB pathway) are required for the Wnt5a/NF-κB crosstalk. Wnt5a also induced p65 nuclear translocation and increased NF-κB activity as evidenced by reporter assays and a NF-κB-specific upregulation of RelB, Bcl-2, and Cyclin D1. Further, stimulation of melanoma cells with Wnt5a increased the secretion of cytokines and chemokines, including IL-6, IL-8, IL-11, and IL-6 soluble receptor, MCP-1, and TNF soluble receptor I. The inhibition of endogenous Wnt5a demonstrated that an autocrine Wnt5a loop is a major regulator of the NF-κB pathway in melanoma. Taken together, these results indicate that Wnt5a activates the NF-κB pathway and has an immunomodulatory effect on melanoma through the secretion of cytokines and chemokines.
Collapse
|
15
|
Abstract
Cannabinoid receptors, endocannabinoids and the enzymes responsible for their biosynthesis and degradation constitute the endocannabinoid system. In recent decades, the endocannabinoid system has attracted considerable interest as a potential therapeutic target in numerous pathological conditions. Its involvement in several physiological processes is well known, such as in energy balance, appetite stimulation, blood pressure, pain modulation, embryogenesis, nausea and vomiting control, memory, learning and immune response, among others, as well as in pathological conditions where it exerts a protective role in the development of certain disorders. As a result, it has been reported that changes in endocannabinoid levels may be related to neurological diseases such as Parkinson's disease, Huntington's disease, Alzheimer's disease and multiple sclerosis, as well as anorexia and irritable bowel syndrome. Alterations in the endocannabinoid system have also been associated with cancer, affecting the growth, migration and invasion of some tumours. Cannabinoids have been tested in several cancer types, including brain, breast and prostate cancers. Cannabinoids have shown promise as analgesics for the treatment of both inflammatory and neuropathic pain. There is also evidence for a role of the endocannabinoid system in the control of emotional states, and cannabinoids could prove useful in decreasing and palliating post-traumatic stress disorder symptoms and anxiolytic disorders. The role of the endocannabinoid system in addictions has also been examined, and cannabinoids have been postulated as alternative and co-adjuvant treatments in some abuse syndromes, mainly in ethanol and opioid abuses. The expression of the endocannabinoid system in the eye suggests that it could be a potential therapeutic target for eye diseases. Considering the importance of the endocannabinoid system and the therapeutic potential of cannabinoids in this vast number of medical conditions, several clinical studies with cannabinoid-based medications are ongoing. In addition, some cannabinoid-based medications have already been approved in various countries, including nabilone and dronabinol capsules for the treatment of nausea and vomiting associated with chemotherapy, dronabinol capsules for anorexia, an oral solution of dronabinol for both vomiting associated with chemotherapy and anorexia, a Δ9-tetrahydrocannabinol/cannabidiol oromucosal spray for pain related to cancer and for spasticity and pain associated with multiple sclerosis, and an oral solution of cannabidiol for Dravet and Lennox-Gastaut syndromes. Here, we review the available efficacy, safety and tolerability data for cannabinoids in a range of medical conditions.
Collapse
Affiliation(s)
- Ana Isabel Fraguas-Sánchez
- Department of Pharmaceutics and Food Technology, Faculty of Pharmacy, Plaza Ramón y Cajal s/n, 28040 , Madrid, Spain
| | - Ana Isabel Torres-Suárez
- Department of Pharmaceutics and Food Technology, Faculty of Pharmacy, Plaza Ramón y Cajal s/n, 28040 , Madrid, Spain. .,Institute of Industrial Pharmacy, Complutense University of Madrid, 28040 , Madrid, Spain.
| |
Collapse
|
16
|
Xu L, Lin W, Wen L, Li G. Lgr5 in cancer biology: functional identification of Lgr5 in cancer progression and potential opportunities for novel therapy. Stem Cell Res Ther 2019; 10:219. [PMID: 31358061 PMCID: PMC6664754 DOI: 10.1186/s13287-019-1288-8] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Cancer remains one of the leading lethal diseases worldwide. Identifying biomarkers of cancers might provide insights into the strategies for the development of novel targeted anti-cancer therapies. Leucine-rich repeat-containing G protein-coupled receptor 5 (Lgr5) has been recently discovered as a candidate marker of cancer stem cell populations. Aberrant increased expression of Lgr5 may represent one of the most common molecular alterations in some human cancers, leading to long-term potentiation of canonical Wnt/β-catenin signaling. On the other hand, however, Lgr5-mediated suppression in canonical Wnt/β-catenin signaling has also been reported in certain cancers, such as B cell malignancies. Until now, therapeutic approaches targeting Lgr5-associated signaling axis are not yet clinically available. Increasing evidence have indicated that endogenous Lgr5+ cell population is implicated in tumor initiation, progression, and metastasis. This review is to summarize our current knowledge about the importance of Lgr5 in cancer biology and the underlying molecular mechanisms of Lgr5-mediated tumor-promoting/suppressive activities, as well as potentially useful preventive strategies in treating tumor. Therefore, targeted therapeutic modulation of Lgr5+ cancer cell population by targeting Wnt/β-catenin signaling through targeted drug delivery system or targeted genome editing might be promising for potential novel anti-cancer treatments. Simultaneously, combination of therapeutics targeting both Lgr5+ and Lgr5- cancer cells may deserve further consideration considering the plasticity of cancer cells. Also, a more specific targeting of cancer cells using double biomarkers may be much safer and more effective for anti-cancer therapy.
Collapse
Affiliation(s)
- Liangliang Xu
- Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, People’s Republic of China
- Laboratory of Orthopaedics and Traumatology, Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, People’s Republic of China
| | - Weiping Lin
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong SAR PRC
- Stem Cells and Regenerative Medicine Laboratory, Lui Che Woo Institute of Innovative Medicine, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR PRC
| | - Longping Wen
- Nanobio Laboratory, Institute of Life Sciences, South China University of Technology, Guangzhou, Guangdong People’s Republic of China
| | - Gang Li
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong SAR PRC
- Stem Cells and Regenerative Medicine Laboratory, Lui Che Woo Institute of Innovative Medicine, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR PRC
- The CUHK-ACC Space Medicine Centre on Health Maintenance of Musculoskeletal System, The Chinese University of Hong Kong Shenzhen Research Institute, Shenzhen, People’s Republic of China
| |
Collapse
|
17
|
McMillin M, Grant S, Frampton G, Petrescu AD, Williams E, Jefferson B, Thomas A, Brahmaroutu A, DeMorrow S. Elevated circulating TGFβ1 during acute liver failure activates TGFβR2 on cortical neurons and exacerbates neuroinflammation and hepatic encephalopathy in mice. J Neuroinflammation 2019; 16:69. [PMID: 30940161 PMCID: PMC6446280 DOI: 10.1186/s12974-019-1455-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 03/18/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Acute liver failure resulting from drug-induced liver injury can lead to the development of neurological complications called hepatic encephalopathy (HE). Hepatic transforming growth factor beta 1 (TGFβ1) is upregulated due to liver failure in mice and inhibiting circulating TGFβ reduced HE progression. However, the specific contributions of TGFβ1 on brain cell populations and neuroinflammation during HE are not known. Therefore, the aim of this study was to characterize hepatic and brain TGFβ1 signaling during acute liver failure and its contribution to HE progression using a combination of pharmacological and genetic approaches. METHODS C57Bl/6 or neuron-specific transforming growth factor beta receptor 2 (TGFβR2) null mice (TGFβR2ΔNeu) were treated with azoxymethane (AOM) to induce acute liver failure and HE. The activity of circulating TGFβ1 was inhibited in C57Bl/6 mice via injection of a neutralizing antibody against TGFβ1 (anti-TGFβ1) prior to AOM injection. In all mouse treatment groups, liver damage, neuroinflammation, and neurological deficits were assessed. Inflammatory signaling between neurons and microglia were investigated in in vitro studies through the use of pharmacological inhibitors of TGFβ1 signaling in HT-22 and EOC-20 cells. RESULTS TGFβ1 was expressed and upregulated in the liver following AOM injection. Pharmacological inhibition of TGFβ1 after AOM injection attenuated neurological decline, microglia activation, and neuroinflammation with no significant changes in liver damage. TGFβR2ΔNeu mice administered AOM showed no effect on liver pathology but significantly reduced neurological decline compared to control mice. Microglia activation and neuroinflammation were attenuated in mice with pharmacological inhibition of TGFβ1 or in TGFβR2ΔNeu mice. TGFβ1 increased chemokine ligand 2 (CCL2) and decreased C-X3-C motif ligand 1 (CX3CL1) expression in HT-22 cells and reduced interleukin-1 beta (IL-1ß) expression, tumor necrosis factor alpha (TNFα) expression, and phagocytosis activity in EOC-20 cells. CONCLUSION Increased circulating TGFβ1 following acute liver failure results in activation of neuronal TGFβR2 signaling, driving neuroinflammation and neurological decline during AOM-induced HE.
Collapse
Affiliation(s)
- Matthew McMillin
- Central Texas Veterans Health Care System, Temple, TX, USA.,Department of Medical Physiology, College of Medicine, Texas A&M University Health Science Center, Temple, TX, USA.,Department of Internal Medicine, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
| | - Stephanie Grant
- Central Texas Veterans Health Care System, Temple, TX, USA.,Department of Medical Physiology, College of Medicine, Texas A&M University Health Science Center, Temple, TX, USA.,Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX, USA
| | - Gabriel Frampton
- Central Texas Veterans Health Care System, Temple, TX, USA.,Department of Medical Physiology, College of Medicine, Texas A&M University Health Science Center, Temple, TX, USA.,Department of Internal Medicine, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
| | - Anca D Petrescu
- Central Texas Veterans Health Care System, Temple, TX, USA.,Department of Medical Physiology, College of Medicine, Texas A&M University Health Science Center, Temple, TX, USA.,Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX, USA
| | - Elaina Williams
- Central Texas Veterans Health Care System, Temple, TX, USA.,Department of Medical Physiology, College of Medicine, Texas A&M University Health Science Center, Temple, TX, USA.,Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX, USA
| | - Brandi Jefferson
- Central Texas Veterans Health Care System, Temple, TX, USA.,Department of Medical Physiology, College of Medicine, Texas A&M University Health Science Center, Temple, TX, USA
| | - Alison Thomas
- Department of Medical Physiology, College of Medicine, Texas A&M University Health Science Center, Temple, TX, USA
| | - Ankita Brahmaroutu
- Department of Medical Physiology, College of Medicine, Texas A&M University Health Science Center, Temple, TX, USA
| | - Sharon DeMorrow
- Central Texas Veterans Health Care System, Temple, TX, USA. .,Department of Medical Physiology, College of Medicine, Texas A&M University Health Science Center, Temple, TX, USA. .,Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX, USA. .,Department of Internal Medicine, Dell Medical School, The University of Texas at Austin, Austin, TX, USA.
| |
Collapse
|
18
|
Fraguas‐Sánchez AI, Martín‐Sabroso C, Torres‐Suárez AI. Insights into the effects of the endocannabinoid system in cancer: a review. Br J Pharmacol 2018; 175:2566-2580. [PMID: 29663308 PMCID: PMC6003657 DOI: 10.1111/bph.14331] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 03/15/2018] [Accepted: 03/26/2018] [Indexed: 01/03/2023] Open
Abstract
In the last few decades, the endocannabinoid system has attracted a great deal of interest in terms of its applications to clinical medicine. In particular, its applications in cancer probably represent one of the therapeutic areas with most promise. On the one hand, expression of the endocannabinoid system is altered in numerous types of tumours, compared to healthy tissue, and this aberrant expression has been related to cancer prognosis and disease outcome, suggesting a role of this system in tumour growth and progression that depends on cancer type. On the other hand, cannabinoids exert an anticancer activity by inhibiting the proliferation, migration and/or invasion of cancer cells and also tumour angiogenesis. However, some cannabinoids, at lower concentrations, may increase tumour proliferation, inducing cancer growth. Enough data has been provided to consider the endocannabinoid system as a new therapeutic target in cancer, although further studies to fully establish the effect of cannabinoids on tumour progression are still needed.
Collapse
Affiliation(s)
- Ana Isabel Fraguas‐Sánchez
- Department of Pharmaceutical Technology, Faculty of PharmacyComplutense University of MadridMadrid28040Spain
| | - Cristina Martín‐Sabroso
- Department of Pharmaceutical Technology, Faculty of PharmacyComplutense University of MadridMadrid28040Spain
| | - Ana Isabel Torres‐Suárez
- Department of Pharmaceutical Technology, Faculty of PharmacyComplutense University of MadridMadrid28040Spain
- Institute of Industrial PharmacyComplutense University of MadridMadrid28040Spain
| |
Collapse
|
19
|
Wang L, Yao M, Fang M, Zheng WJ, Dong ZZ, Pan LH, Zhang HJ, Yao DF. Expression of hepatic Wnt5a and its clinicopathological features in patients with hepatocellular carcinoma. Hepatobiliary Pancreat Dis Int 2018; 17:227-232. [PMID: 29709351 DOI: 10.1016/j.hbpd.2018.03.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Accepted: 12/27/2017] [Indexed: 02/07/2023]
Abstract
BACKGROUD Wingless-type MMTV integration site family member 5a (Wnt5a) is involved in carcinogenesis. However, little data are available in Wnt5a signaling with hepatocellular carcinoma (HCC). In the present study, we investigated the expression of hepatic Wnt5a in HCC and the role of Wnt5a in HCC progression and outcome. METHODS Wnt5a expression and cellular distribution in HCCs and their matched paracancerous tissues from 87 patients were analyzed with tissue microarray and immunohistochemistry and compared with hepatic Wnt3a signaling. Wnt5a expression was categorized into low or high based on immunohistochemistry. Overall survival rate of HCC patients was estimated in correlation with the hepatic Wnt5a level using Kaplan-Meier method; the survival difference between patients with low and those with high Wnt5a was compared with log-rank test; and prognostic analysis was carried out with Cox regression. RESULTS Total incidence of Wnt5a expression in the HCC tissues was 70.1%, which was significantly lower (χ2 = 13.585, P < 0.001) than that in their paracancerous tissues (88.5%). Significant difference of Wnt5a intensity was found between HCC and their paracancerous tissues (Z = 8.463, P < 0.001). Wnt5a intensity was inversely correlated with Wnt3a signaling (r = -0.402, P < 0.001) in HCC tissues. A decrease of Wnt5a expression in relation to the clinical staging from stage I to IV and low or no staining at advanced HCC were observed. Wnt5a level was related to periportal embolus (χ2 = 11.069, P < 0.001), TNM staging (χ2 = 8.852, P < 0.05), 5-year survival (χ2 = 4.961, P < 0.05), and confirmed as an independent prognosis factor of HCC patients (hazard ratio: 1.957; 95% confidence interval: 1.109-3.456; P < 0.05). CONCLUSIONS The decrease of hepatic Wnt5a signaling is associated with HCC progression and poor prognosis.
Collapse
Affiliation(s)
- Li Wang
- Department of Medical Informatics, Medical College of Nantong University, Nantong, China; Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, No. 20 West Temple Rd, Nantong 226001, China
| | - Min Yao
- Department of Immunology, Medical College of Nantong University, Nantong, China
| | - Miao Fang
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, No. 20 West Temple Rd, Nantong 226001, China
| | - Wen-Jie Zheng
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, No. 20 West Temple Rd, Nantong 226001, China
| | - Zhi-Zhen Dong
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, No. 20 West Temple Rd, Nantong 226001, China
| | - Liu-Hong Pan
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, No. 20 West Temple Rd, Nantong 226001, China
| | - Hai-Jian Zhang
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, No. 20 West Temple Rd, Nantong 226001, China
| | - Deng-Fu Yao
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, No. 20 West Temple Rd, Nantong 226001, China.
| |
Collapse
|
20
|
McMillin M, Grant S, Frampton G, Petrescu AD, Kain J, Williams E, Haines R, Canady L, DeMorrow S. FXR-Mediated Cortical Cholesterol Accumulation Contributes to the Pathogenesis of Type A Hepatic Encephalopathy. Cell Mol Gastroenterol Hepatol 2018; 6:47-63. [PMID: 29928671 PMCID: PMC6008252 DOI: 10.1016/j.jcmgh.2018.02.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 02/26/2018] [Indexed: 12/13/2022]
Abstract
BACKGROUND & AIMS Hepatic encephalopathy is a serious neurologic complication of acute and chronic liver diseases. We previously showed that aberrant bile acid signaling contributes to the development of hepatic encephalopathy via farnesoid X receptor (FXR)-mediated mechanisms in neurons. In the brain, a novel alternative bile acid synthesis pathway, catalyzed by cytochrome p450 46A1 (Cyp46A1), is the primary mechanism by which the brain regulates cholesterol homeostasis. The aim of this study was to determine if FXR activation in the brain altered cholesterol homeostasis during hepatic encephalopathy. METHODS Cyp7A1-/- mice or C57Bl/6 mice pretreated with central infusion of FXR vivo morpholino, 2-hydroxypropyl-β-cyclodextrin, or fed a cholestyramine-supplemented diet were injected with azoxymethane (AOM). Cognitive and neuromuscular impairment as well as liver damage and expression of Cyp46A1 were assessed using standard techniques. The subsequent cholesterol content in the frontal cortex was measured using commercially available kits and by Filipin III and Nile Red staining. RESULTS There was an increase in membrane-bound and intracellular cholesterol in the cortex of mice treated with AOM that was associated with decreased Cyp46A1 expression. Strategies to inhibit FXR signaling prevented the down-regulation of Cyp46A1 and the accumulation of cholesterol. Treatment of mice with 2-hydroxypropyl-β-cyclodextrin attenuated the AOM-induced cholesterol accumulation in the brain and the cognitive and neuromuscular deficits without altering the underlying liver pathology. CONCLUSIONS During hepatic encephalopathy, FXR signaling increases brain cholesterol and contributes to neurologic decline. Targeting cholesterol accumulation in the brain may be a possible therapeutic target for the management of hepatic encephalopathy.
Collapse
Key Words
- 2-HβC, 2-hypdroxypropyl-β-cyclodextrin
- AOM, azoxymethane
- Acute Liver Failure
- Azoxymethane
- CYP46A1, cytochrome p450 46A1
- CYP7A1, cytochrome p450 7A1
- Cytochrome p450 46A1
- FXR, farnesoid X receptor
- Farnesoid X Receptor
- GAPDH, glyceraldehyde-3-phosphate dehydrogenase
- PBS, phosphate-buffered saline
- PFA, paraformaldehyde
- RT-PCR, reverse-transcription polymerase chain reaction
- WT, wild-type
Collapse
Affiliation(s)
- Matthew McMillin
- Central Texas Veterans Healthcare System, Temple, Texas,Department of Medical Physiology, Texas A&M College of Medicine, Temple, Texas
| | - Stephanie Grant
- Central Texas Veterans Healthcare System, Temple, Texas,Department of Medical Physiology, Texas A&M College of Medicine, Temple, Texas
| | - Gabriel Frampton
- Central Texas Veterans Healthcare System, Temple, Texas,Department of Medical Physiology, Texas A&M College of Medicine, Temple, Texas
| | - Anca D. Petrescu
- Central Texas Veterans Healthcare System, Temple, Texas,Department of Medical Physiology, Texas A&M College of Medicine, Temple, Texas
| | - Jessica Kain
- Central Texas Veterans Healthcare System, Temple, Texas,Department of Medical Physiology, Texas A&M College of Medicine, Temple, Texas
| | - Elaina Williams
- Central Texas Veterans Healthcare System, Temple, Texas,Department of Medical Physiology, Texas A&M College of Medicine, Temple, Texas
| | - Rebecca Haines
- Department of Medical Physiology, Texas A&M College of Medicine, Temple, Texas
| | - Lauren Canady
- Department of Medical Physiology, Texas A&M College of Medicine, Temple, Texas
| | - Sharon DeMorrow
- Central Texas Veterans Healthcare System, Temple, Texas,Department of Medical Physiology, Texas A&M College of Medicine, Temple, Texas,Correspondence Address correspondence to: Sharon DeMorrow, PhD, Department of Medical Physiology, Texas A&M Health Science Center, Central Texas Veterans Health Care System, Building 205, 1901 South 1st Street, Temple, Texas 76504. fax: (254) 743-0378.
| |
Collapse
|
21
|
Fiore D, Ramesh P, Proto MC, Piscopo C, Franceschelli S, Anzelmo S, Medema JP, Bifulco M, Gazzerro P. Rimonabant Kills Colon Cancer Stem Cells without Inducing Toxicity in Normal Colon Organoids. Front Pharmacol 2018; 8:949. [PMID: 29354056 PMCID: PMC5758598 DOI: 10.3389/fphar.2017.00949] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 12/14/2017] [Indexed: 12/11/2022] Open
Abstract
Colorectal cancer (CRC), like other tumor types, is a highly heterogeneous disease. Within the tumor bulk, intra-tumoral heterogeneity is also ascribable to Cancer Stem Cells (CSCs) subpopulation, characterized by high chemoresistance and the unique ability to retain tumorigenic potential, thus associated to tumor recurrence. High dynamic plasticity of CSCs, makes the development of winning therapeutic strategies even more complex to completely eradicate tumor fuel. Rimonabant, originally synthesized as antagonist/inverse agonist of Cannabinoid Receptor 1, is able to inactivate Wnt signaling, both in vitro and in vivo, in CRC models, through inhibition of p300-histone acetyltransferase activity. Since Wnt/β-Catenin pathway is the main player underlying CSCs dynamic, this finding candidates Rimonabant as potential modulator of cancer stemness, in CRC. In this work, using established 3D cultures of primary colon CSCs, taking into account the tumor heterogeneity through monitoring of Wnt activity, we demonstrated that Rimonabant was able to reduces both tumor differentiated cells and colon CSCs proliferation and to control their survival in long term cultures. Interestingly, in ex vivo model of wild type human organoids, retaining both architecture and heterogeneity of original tissue, Rimonabant showed no toxicity against cells from healthy colon epithelium, suggesting its potential selectivity toward cancer cells. Overall, results from this work provided new insights on anti-tumor efficacy of Rimonabant, strongly suggesting that it could be a novel lead compound for CRC treatment.
Collapse
Affiliation(s)
- Donatella Fiore
- Department of Pharmacy, University of Salerno, Fisciano, Italy
| | - Prashanthi Ramesh
- Laboratory of Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Academisch Medisch Centrum, University of Amsterdam, Amsterdam, Netherlands
| | - Maria C Proto
- Department of Pharmacy, University of Salerno, Fisciano, Italy
| | - Chiara Piscopo
- Department of Pharmacy, University of Salerno, Fisciano, Italy
| | | | - Serena Anzelmo
- Department of Pharmacy, University of Salerno, Fisciano, Italy
| | - Jan P Medema
- Laboratory of Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Academisch Medisch Centrum, University of Amsterdam, Amsterdam, Netherlands
| | - Maurizio Bifulco
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples "Federico II", Naples, Italy
| | | |
Collapse
|
22
|
McMillin M, DeMorrow S, Glaser S, Venter J, Kyritsi K, Zhou T, Grant S, Giang T, Greene JF, Wu N, Jefferson B, Meng F, Alpini G. Melatonin inhibits hypothalamic gonadotropin-releasing hormone release and reduces biliary hyperplasia and fibrosis in cholestatic rats. Am J Physiol Gastrointest Liver Physiol 2017; 313:G410-G418. [PMID: 28751425 PMCID: PMC5792219 DOI: 10.1152/ajpgi.00421.2016] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 06/23/2017] [Accepted: 07/05/2017] [Indexed: 01/31/2023]
Abstract
Melatonin is a hormone produced by the pineal gland with increased circulating levels shown to inhibit biliary hyperplasia and fibrosis during cholestatic liver injury. Melatonin also has the capability to suppress the release of hypothalamic gonadotropin-releasing hormone (GnRH), a hormone that promotes cholangiocyte proliferation when serum levels are elevated. However, the interplay and contribution of neural melatonin and GnRH to cholangiocyte proliferation and fibrosis in bile duct-ligated (BDL) rats have not been investigated. To test this, cranial levels of melatonin were increased by implanting osmotic minipumps that performed an intracerebroventricular (ICV) infusion of melatonin or saline for 7 days starting at the time of BDL. Hypothalamic GnRH mRNA and cholangiocyte secretion of GnRH and melatonin were assessed. Cholangiocyte proliferation and fibrosis were measured. Primary human hepatic stellate cells (HSCs) were treated with cholangiocyte supernatants, GnRH, or the GnRH receptor antagonist cetrorelix acetate, and cell proliferation and fibrosis gene expression were assessed. Melatonin infusion reduced hypothalamic GnRH mRNA expression and led to decreased GnRH and increased melatonin secretion from cholangiocytes. Infusion of melatonin was found to reduce hepatic injury, cholangiocyte proliferation, and fibrosis during BDL-induced liver injury. HSCs supplemented with BDL cholangiocyte supernatant had increased proliferation, and this increase was reversed when HSCs were supplemented with supernatants from melatonin-infused rats. GnRH stimulated fibrosis gene expression in HSCs, and this was reversed by cetrorelix acetate cotreatment. Increasing bioavailability of melatonin in the brain may improve outcomes during cholestatic liver disease.NEW & NOTEWORTHY We have previously demonstrated that GnRH is expressed in cholangiocytes and promotes their proliferation during cholestasis. In addition, dark therapy, which increases melatonin, reduced cholangiocyte proliferation and fibrosis during cholestasis. This study expands these findings by investigating neural GnRH regulation by melatonin during BDL-induced cholestasis by infusing melatonin into the brain. Melatonin infusion reduced cholangiocyte proliferation and fibrosis, and these effects are due to GNRH receptor 1-dependent paracrine signaling between cholangiocytes and hepatic stellate cells.
Collapse
Affiliation(s)
- Matthew McMillin
- Research, Central Texas Veterans Health Care System, Temple, Texas
- Digestive Disease Research Center, Baylor Scott & White Health, Temple, Texas
- Department of Medicine, Division Gastroenterology, Texas A&M University Health Science Center and Baylor Scott & White Health, Temple, Texas
| | - Sharon DeMorrow
- Research, Central Texas Veterans Health Care System, Temple, Texas
- Digestive Disease Research Center, Baylor Scott & White Health, Temple, Texas
- Department of Medicine, Division Gastroenterology, Texas A&M University Health Science Center and Baylor Scott & White Health, Temple, Texas
| | - Shannon Glaser
- Research, Central Texas Veterans Health Care System, Temple, Texas
- Digestive Disease Research Center, Baylor Scott & White Health, Temple, Texas
- Department of Medicine, Division Gastroenterology, Texas A&M University Health Science Center and Baylor Scott & White Health, Temple, Texas
| | - Julie Venter
- Digestive Disease Research Center, Baylor Scott & White Health, Temple, Texas
- Department of Medicine, Division Gastroenterology, Texas A&M University Health Science Center and Baylor Scott & White Health, Temple, Texas
| | - Konstantina Kyritsi
- Digestive Disease Research Center, Baylor Scott & White Health, Temple, Texas
- Department of Medicine, Division Gastroenterology, Texas A&M University Health Science Center and Baylor Scott & White Health, Temple, Texas
| | - Tianhao Zhou
- Research, Central Texas Veterans Health Care System, Temple, Texas
- Department of Medicine, Division Gastroenterology, Texas A&M University Health Science Center and Baylor Scott & White Health, Temple, Texas
| | - Stephanie Grant
- Digestive Disease Research Center, Baylor Scott & White Health, Temple, Texas
- Department of Medicine, Division Gastroenterology, Texas A&M University Health Science Center and Baylor Scott & White Health, Temple, Texas
| | - Thao Giang
- Research, Central Texas Veterans Health Care System, Temple, Texas
- Digestive Disease Research Center, Baylor Scott & White Health, Temple, Texas
- Department of Medicine, Division Gastroenterology, Texas A&M University Health Science Center and Baylor Scott & White Health, Temple, Texas
| | - John F Greene
- Department of Pathology, Baylor Scott & White Health, Temple, Texas; and
| | - Nan Wu
- Digestive Disease Research Center, Baylor Scott & White Health, Temple, Texas
- Department of Medicine, Division Gastroenterology, Texas A&M University Health Science Center and Baylor Scott & White Health, Temple, Texas
| | - Brandi Jefferson
- Digestive Disease Research Center, Baylor Scott & White Health, Temple, Texas
- Department of Medicine, Division Gastroenterology, Texas A&M University Health Science Center and Baylor Scott & White Health, Temple, Texas
| | - Fanyin Meng
- Research, Central Texas Veterans Health Care System, Temple, Texas
- Digestive Disease Research Center, Baylor Scott & White Health, Temple, Texas
- Department of Medicine, Division Gastroenterology, Texas A&M University Health Science Center and Baylor Scott & White Health, Temple, Texas
- Research Foundation, Baylor Scott & White Health, Temple, Texas
| | - Gianfranco Alpini
- Research, Central Texas Veterans Health Care System, Temple, Texas;
- Digestive Disease Research Center, Baylor Scott & White Health, Temple, Texas
- Department of Medicine, Division Gastroenterology, Texas A&M University Health Science Center and Baylor Scott & White Health, Temple, Texas
| |
Collapse
|
23
|
Inhibition of Wnt/β-Catenin pathway and Histone acetyltransferase activity by Rimonabant: a therapeutic target for colon cancer. Sci Rep 2017; 7:11678. [PMID: 28916833 PMCID: PMC5601949 DOI: 10.1038/s41598-017-11688-x] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 08/09/2017] [Indexed: 12/18/2022] Open
Abstract
In a high percentage (≥85%) of both sporadic and familial adenomatous polyposis forms of colorectal cancer (CRC), the inactivation of the APC tumor suppressor gene initiates tumor formation and modulates the Wnt/β-Catenin transduction pathways involved in the control of cell proliferation, adhesion and metastasis. Increasing evidence showed that the endocannabinoids control tumor growth and progression, both in vitro and in vivo. We evaluated the effect of Rimonabant, a Cannabinoid Receptor 1 (CB1) inverse agonist, on the Wnt/β-Catenin pathway in HCT116 and SW48 cell lines carrying the genetic profile of metastatic CRC poorly responsive to chemotherapies. In these models, Rimonabant inhibited the Wnt/β-Catenin canonical pathway and increased β-Catenin phosphorylation; in HCT116 cells, but not in SW48, the compound also triggered the Wnt/β-Catenin non canonical pathway activation through induction of Wnt5A and activation of CaMKII. The Rimonabant-induced downregulation of Wnt/β-Catenin target genes was partially ascribable to a direct inhibition of p300/KAT3B histone acetyltransferase, a coactivator of β-Catenin dependent gene regulation. Finally, in HCT116 xenografts, Rimonabant significantly reduced tumor growth and destabilized the nuclear localization of β-Catenin. Obtained data heavily supported the rationale for the use of cannabinoids in combined therapies for metastatic CRC harbouring activating mutations of β-Catenin.
Collapse
|
24
|
Identifying novel members of the Wntless interactome through genetic and candidate gene approaches. Brain Res Bull 2017; 138:96-105. [PMID: 28734904 DOI: 10.1016/j.brainresbull.2017.07.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 06/26/2017] [Accepted: 07/06/2017] [Indexed: 02/07/2023]
Abstract
Wnt signaling is an important pathway that regulates several aspects of embryogenesis, stem cell maintenance, and neural connectivity. We have recently determined that opioids decrease Wnt secretion, presumably by inhibiting the recycling of the Wnt trafficking protein Wntless (Wls). This effect appears to be mediated by protein-protein interaction between Wls and the mu-opioid receptor (MOR), the primary cellular target of opioid drugs. The goal of this study was to identify novel protein interactors of Wls that are expressed in the brain and may also play a role in reward or addiction. Using genetic and candidate gene approaches, we show that among a variety of protein, Wls interacts with the dopamine transporter (target of cocaine), cannabinoid receptors (target of THC), Adenosine A2A receptor (target of caffeine), and SGIP1 (endocytic regulator of cannabinoid receptors). Our study shows that aside from opioid receptors, Wntless interacts with additional proteins involved in reward and/or addiction. Future studies will determine whether Wntless and WNT signaling play a more universal role in these processes.
Collapse
|
25
|
McMillin M, Frampton G, Grant S, Khan S, Diocares J, Petrescu A, Wyatt A, Kain J, Jefferson B, DeMorrow S. Bile Acid-Mediated Sphingosine-1-Phosphate Receptor 2 Signaling Promotes Neuroinflammation during Hepatic Encephalopathy in Mice. Front Cell Neurosci 2017; 11:191. [PMID: 28725183 PMCID: PMC5496949 DOI: 10.3389/fncel.2017.00191] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 06/20/2017] [Indexed: 12/16/2022] Open
Abstract
Hepatic encephalopathy (HE) is a neuropsychiatric complication that occurs due to deteriorating hepatic function and this syndrome influences patient quality of life, clinical management strategies and survival. During acute liver failure, circulating bile acids increase due to a disruption of the enterohepatic circulation. We previously identified that bile acid-mediated signaling occurs in the brain during HE and contributes to cognitive impairment. However, the influences of bile acids and their downstream signaling pathways on HE-induced neuroinflammation have not been assessed. Conjugated bile acids, such as taurocholic acid (TCA), can activate sphingosine-1-phosphate receptor 2 (S1PR2), which has been shown to promote immune cell infiltration and inflammation in other models. The current study aimed to assess the role of bile-acid mediated S1PR2 signaling in neuroinflammation and disease progression during azoxymethane (AOM)-induced HE in mice. Our findings demonstrate a temporal increase of bile acids in the cortex during AOM-induced HE and identified that cortical bile acids were elevated as an early event in this model. In order to classify the specific bile acids that were elevated during HE, a metabolic screen was performed and this assay identified that TCA was increased in the serum and cortex during AOM-induced HE. To reduce bile acid concentrations in the brain, mice were fed a diet supplemented with cholestyramine, which alleviated neuroinflammation by reducing proinflammatory cytokine expression in the cortex compared to the control diet-fed AOM-treated mice. S1PR2 was expressed primarily in neurons and TCA treatment increased chemokine ligand 2 mRNA expression in these cells. The infusion of JTE-013, a S1PR2 antagonist, into the lateral ventricle prior to AOM injection protected against neurological decline and reduced neuroinflammation compared to DMSO-infused AOM-treated mice. Together, this identifies that reducing bile acid levels or S1PR2 signaling are potential therapeutic strategies for the management of HE.
Collapse
Affiliation(s)
- Matthew McMillin
- Department of Research, Central Texas Veterans Health Care SystemTemple, TX, United States.,Department of Internal Medicine, College of Medicine, Texas A&M University Health Science CenterTemple, TX, United States
| | - Gabriel Frampton
- Department of Research, Central Texas Veterans Health Care SystemTemple, TX, United States.,Department of Internal Medicine, College of Medicine, Texas A&M University Health Science CenterTemple, TX, United States
| | - Stephanie Grant
- Department of Research, Central Texas Veterans Health Care SystemTemple, TX, United States.,Department of Internal Medicine, College of Medicine, Texas A&M University Health Science CenterTemple, TX, United States
| | - Shamyal Khan
- Department of Internal Medicine, Baylor Scott & White HealthTemple, TX, United States
| | - Juan Diocares
- Department of Internal Medicine, Baylor Scott & White HealthTemple, TX, United States
| | - Anca Petrescu
- Department of Research, Central Texas Veterans Health Care SystemTemple, TX, United States.,Department of Internal Medicine, College of Medicine, Texas A&M University Health Science CenterTemple, TX, United States
| | - Amy Wyatt
- Department of Research, Central Texas Veterans Health Care SystemTemple, TX, United States.,Department of Internal Medicine, College of Medicine, Texas A&M University Health Science CenterTemple, TX, United States
| | - Jessica Kain
- Department of Research, Central Texas Veterans Health Care SystemTemple, TX, United States.,Department of Internal Medicine, College of Medicine, Texas A&M University Health Science CenterTemple, TX, United States
| | - Brandi Jefferson
- Department of Research, Central Texas Veterans Health Care SystemTemple, TX, United States.,Department of Internal Medicine, College of Medicine, Texas A&M University Health Science CenterTemple, TX, United States
| | - Sharon DeMorrow
- Department of Research, Central Texas Veterans Health Care SystemTemple, TX, United States.,Department of Internal Medicine, College of Medicine, Texas A&M University Health Science CenterTemple, TX, United States
| |
Collapse
|
26
|
Li G, Chen Z, Bhat OM, Zhang Q, Abais-Battad JM, Conley SM, Ritter JK, Li PL. NLRP3 inflammasome as a novel target for docosahexaenoic acid metabolites to abrogate glomerular injury. J Lipid Res 2017; 58:1080-1090. [PMID: 28404641 DOI: 10.1194/jlr.m072587] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Revised: 03/31/2017] [Indexed: 01/09/2023] Open
Abstract
The nucleotide-binding oligomerization domain-like receptor containing pyrin domain 3 (NLRP3) inflammasome has been implicated in podocyte injury and glomerular sclerosis during hyperhomocysteinemia (hHcys). However, it remains unclear whether the NLRP3 inflammasome can be a therapeutic target for treatment of hHcys-induced kidney injury. Given that DHA metabolites-resolvins have potent anti-inflammatory effects, the present study tested whether the prototype, resolvin D1 (RvD1), and 17S-hydroxy DHA (17S-HDHA), an intermediate product, abrogate hHcys-induced podocyte injury by targeting the NLRP3 inflammasome. In vitro, confocal microscopy demonstrated that 17S-HDHA (100 nM) and RvD1 (60 nM) prevented Hcys-induced formation of NLRP3 inflammasomes, as shown by reduced colocalization of NLRP3 with apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC) or caspase-1. Both DHA metabolites inhibited Hcys-induced caspase-1 activation and interleukin-1β production. However, DHA had no significant effect on these Hcys-induced changes in podocytes. In vivo, DHA lipoxygenase metabolites substantially inhibited podocyte NLRP3 inflammasome formation and activation and consequent glomerular sclerosis in mice with hHcys. Mechanistically, RvD1 and 17S-HDHA were shown to suppress Hcys-induced formation of lipid raft redox signaling platforms and subsequent O2·- production in podocytes. It is concluded that inhibition of NLRP3 inflammasome activation is one of the important mechanisms mediating the beneficial action of RvD1 and 17S-HDHA on Hcys-induced podocyte injury and glomerular sclerosis.
Collapse
Affiliation(s)
- Guangbi Li
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA
| | - Zhida Chen
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA
| | - Owais M Bhat
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA
| | - Qinghua Zhang
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA
| | - Justine M Abais-Battad
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA
| | - Sabena M Conley
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA
| | - Joseph K Ritter
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA
| | - Pin-Lan Li
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA
| |
Collapse
|
27
|
Ladin DA, Soliman E, Escobedo R, Fitzgerald TL, Yang LV, Burns C, Van Dross R. Synthesis and Evaluation of the Novel Prostamide, 15-Deoxy, Δ 12,14-Prostamide J 2, as a Selective Antitumor Therapeutic. Mol Cancer Ther 2017; 16:838-849. [PMID: 28292936 DOI: 10.1158/1535-7163.mct-16-0484] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 08/18/2016] [Accepted: 02/17/2017] [Indexed: 11/16/2022]
Abstract
15-deoxy, Δ12,14-prostaglandin J2-ethanolamide, also known as 15-deoxy, Δ12,14-prostamide J2 (15d-PMJ2) is a novel product of the metabolism of arachidonoyl ethanolamide (AEA) by COX-2. 15d-PMJ2 preferentially induced cell death and apoptosis in tumorigenic A431 keratinocytes and B16F10 melanoma cells compared with nontumorigenic HaCaT keratinocytes and Melan-A melanocytes. Activation of the ER stress execution proteins, PERK and CHOP10, was evaluated to determine whether this process was involved in 15d-PMJ2 cell death. 15d-PMJ2 increased the phosphorylation of PERK and expression of CHOP10 in tumorigenic but not nontumorigenic cells. The known ER stress inhibitors, salubrinal and 4-phenylbutaric acid, significantly inhibited 15d-PMJ2-mediated apoptosis, suggesting ER stress as a primary apoptotic mediator. Furthermore, the reactive double bond present within the cyclopentenone structure of 15d-PMJ2 was identified as a required moiety for the induction of ER stress apoptosis. The effect of 15d-PMJ2 on B16F10 melanoma growth was also evaluated by dosing C57BL/6 mice with 0.5 mg/kg 15d-PMJ2 Tumors of animals treated with 15d-PMJ2 exhibited significantly reduced growth and mean weights compared with vehicle and untreated animals. TUNEL and IHC analysis of tumor tissues showed significant cell death and ER stress in tumors of 15d-PMJ2-treated compared with control group animals. Taken together, these findings suggest that the novel prostamide, 15d-PMJ2, possesses potent antitumor activity in vitro and in vivoMol Cancer Ther; 16(5); 838-49. ©2017 AACR.
Collapse
Affiliation(s)
- Daniel A Ladin
- Department of Pharmacology and Toxicology, East Carolina University, Greenville, North Carolina
| | - Eman Soliman
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Egypt
| | - Rene Escobedo
- Department of Pharmacology and Toxicology, East Carolina University, Greenville, North Carolina
| | | | - Li V Yang
- Department of Internal Medicine, Brody School of Medicine, East Carolina University, Greenville, North Carolina
| | - Colin Burns
- Department of Chemistry, East Carolina University, Greenville, North Carolina
| | - Rukiyah Van Dross
- Department of Pharmacology and Toxicology, East Carolina University, Greenville, North Carolina.
- Center for Health Disparities, East Carolina University, Greenville, North Carolina
| |
Collapse
|
28
|
Ladin DA, Soliman E, Griffin L, Van Dross R. Preclinical and Clinical Assessment of Cannabinoids as Anti-Cancer Agents. Front Pharmacol 2016; 7:361. [PMID: 27774065 PMCID: PMC5054289 DOI: 10.3389/fphar.2016.00361] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 09/20/2016] [Indexed: 12/13/2022] Open
Abstract
Cancer is the second leading cause of death in the United States with 1.7 million new cases estimated to be diagnosed in 2016. This disease remains a formidable clinical challenge and represents a substantial financial burden to the US health care system. Therefore, research and development of novel therapeutics for the treatment of cancer is of high priority. Cannabinoids and their derivatives have been utilized for their medicinal and therapeutic properties throughout history. Cannabinoid activity is regulated by the endocannabinoid system (ECS), which is comprised of cannabinoid receptors, transporters, and enzymes involved in cannabinoid synthesis and breakdown. More recently, cannabinoids have gained special attention for their role in cancer cell proliferation and death. However, many studies investigated these effects using in vitro models which may not adequately mimic tumor growth and metastasis. As such, this article aims to review study results which evaluated effects of cannabinoids from plant, synthetic and endogenous origins on cancer development in preclinical animal models and to examine the current standing of cannabinoids that are being tested in human cancer patients.
Collapse
Affiliation(s)
- Daniel A Ladin
- Department of Pharmacology and Toxicology, Brody School of Medicine, East Carolina University Greenville, NC, USA
| | - Eman Soliman
- Department of Pharmacology and Toxicology, Brody School of Medicine, East Carolina UniversityGreenville, NC, USA; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig UniversityZagazig, Egypt
| | - LaToya Griffin
- Department of Pharmacology and Toxicology, Brody School of Medicine, East Carolina University Greenville, NC, USA
| | - Rukiyah Van Dross
- Department of Pharmacology and Toxicology, Brody School of Medicine, East Carolina UniversityGreenville, NC, USA; Center for Health Disparities, East Carolina UniversityGreenville, NC, USA
| |
Collapse
|
29
|
McMillin M, Grant S, Frampton G, Andry S, Brown A, DeMorrow S. Fractalkine suppression during hepatic encephalopathy promotes neuroinflammation in mice. J Neuroinflammation 2016; 13:198. [PMID: 27561705 PMCID: PMC5000400 DOI: 10.1186/s12974-016-0674-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2015] [Accepted: 08/17/2016] [Indexed: 12/31/2022] Open
Abstract
Background Acute liver failure is associated with numerous systemic consequences including neurological dysfunction, termed hepatic encephalopathy, which contributes to mortality and is a challenge to manage in the clinic. During hepatic encephalopathy, microglia activation and neuroinflammation occur due to dysregulated cell signaling and an increase of toxic metabolites in the brain. Fractalkine is a chemokine that is expressed primarily in neurons and through signaling with its receptor CX3CR1 on microglia, leads to microglia remaining in a quiescent state. Fractalkine is often suppressed during neuropathies that are characterized by neuroinflammation. However, the expression and subsequent role of fractalkine on microglia activation and the pathogenesis of hepatic encephalopathy due to acute liver failure is unknown. Methods Hepatic encephalopathy was induced in mice via injection of azoxymethane (AOM) or saline for controls. Subsets of these mice were implanted with osmotic minipumps that infused soluble fractalkine or saline into the lateral ventricle of the brain. Neurological decline and the latency to coma were recorded in these mice, and brain, serum, and liver samples were collected. Neurons or microglia were isolated from whole brain samples using immunoprecipitation. Liver damage was assessed using hematoxylin and eosin staining and by measuring serum liver enzyme concentrations. Fractalkine and CX3CR1 expression were assessed by real-time PCR, and proinflammatory cytokine expression was assessed using ELISA assays. Results Following AOM administration, fractalkine expression is suppressed in the cortex and in isolated neurons compared to vehicle-treated mice. CX3CR1 is suppressed in isolated microglia from AOM-treated mice. Soluble fractalkine infusion into the brain significantly reduced neurological decline in AOM-treated mice compared to saline-infused AOM-treated mice. Infusion of soluble fractalkine into AOM-treated mice reduced liver damage, lessened microglia activation, and suppressed expression of chemokine ligand 2, interleukin-6, and tumor necrosis factor alpha compared to saline-infused mice. Conclusions These findings suggest that fractalkine-mediated signaling is suppressed in the brain following the development of hepatic encephalopathy. Supplementation of AOM-treated mice with soluble fractalkine led to improved outcomes, which identifies this pathway as a possible therapeutic target for the management of hepatic encephalopathy following acute liver injury.
Collapse
Affiliation(s)
- Matthew McMillin
- Department of Internal Medicine, Texas A&M Health Science Center, College of Medicine, Temple, TX, USA.,Central Texas Veterans Healthcare System, 1901 S. 1st Street, Building 205, Temple, TX, 76504, USA
| | - Stephanie Grant
- Department of Internal Medicine, Texas A&M Health Science Center, College of Medicine, Temple, TX, USA.,Central Texas Veterans Healthcare System, 1901 S. 1st Street, Building 205, Temple, TX, 76504, USA
| | - Gabriel Frampton
- Department of Internal Medicine, Texas A&M Health Science Center, College of Medicine, Temple, TX, USA.,Central Texas Veterans Healthcare System, 1901 S. 1st Street, Building 205, Temple, TX, 76504, USA
| | - Sarah Andry
- Department of Internal Medicine, Baylor Scott & White Health, 2401 S. 31st Street, Temple, TX, 76508, USA
| | - Adam Brown
- Department of Internal Medicine, Baylor Scott & White Health, 2401 S. 31st Street, Temple, TX, 76508, USA
| | - Sharon DeMorrow
- Department of Internal Medicine, Texas A&M Health Science Center, College of Medicine, Temple, TX, USA. .,Central Texas Veterans Healthcare System, 1901 S. 1st Street, Building 205, Temple, TX, 76504, USA.
| |
Collapse
|
30
|
Li G, Xia M, Abais JM, Boini K, Li PL, Ritter JK. Protective Action of Anandamide and Its COX-2 Metabolite against l-Homocysteine-Induced NLRP3 Inflammasome Activation and Injury in Podocytes. J Pharmacol Exp Ther 2016; 358:61-70. [PMID: 27189966 DOI: 10.1124/jpet.116.233239] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 04/28/2016] [Indexed: 12/17/2022] Open
Abstract
Recent studies have demonstrated that l-homocysteine (Hcys)-induced podocyte injury leading to glomerular damage or sclerosis is attributable to the activation of the nucleotide-binding oligomerization domain-like receptor containing pyrin domain 3 (NLRP3) inflammasome. Given the demonstrated anti-inflammatory effects of endocannabinoids, the present study was designed to test whether anandamide (AEA) or its metabolites diminish NLRP3 inflammasome activation and prevent podocyte injury and associated glomerular damage during hyperhomocysteinemia (hHcys). AEA (100 μM) inhibited Hcys-induced NLRP3 inflammasome activation in cultured podocytes, as indicated by elevated caspase-1 activity and interleukin-1β levels, and attenuated podocyte dysfunction, as shown by reduced vascular endothelial growth factor production. These effects of AEA were inhibited by the cyclooxygenase-2 (COX-2) inhibitor celecoxib (CEL). In mice in vivo, AEA treatment attenuated glomerular NLRP3 inflammasome activation induced by hHcys accompanying a folate-free diet, on the basis of inhibition of hHcys-induced colocalization of NLRP3 molecules and increased interleukin-1β levels in glomeruli. Correspondingly, AEA prevented hHcys-induced proteinuria, albuminuria, and glomerular damage observed microscopically. Hcys- and AEA-induced effects were absent in NLRP3-knockout mice. These beneficial effects of AEA against hHcys-induced NLRP3 inflammasome activation and glomerular injury were not observed in mice cotreated with CEL. We further demonstrated that prostaglandin E2-ethanolamide (PGE2-EA), a COX-2 product of AEA, at 10 μM had a similar inhibitory effect to that of 100 μM AEA on Hcys-induced NLRP3 inflammasome formation and activation in cultured podocytes. From these results, we conclude that AEA has anti-inflammatory properties, protecting podocytes from Hcys-induced injury by inhibition of NLRP3 inflammasome activation through its COX-2 metabolite, PGE2-EA.
Collapse
Affiliation(s)
- Guangbi Li
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| | - Min Xia
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| | - Justine M Abais
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| | - Krishna Boini
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| | - Pin-Lan Li
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| | - Joseph K Ritter
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| |
Collapse
|
31
|
Banales JM, Cardinale V, Carpino G, Marzioni M, Andersen JB, Invernizzi P, Lind GE, Folseraas T, Forbes SJ, Fouassier L, Geier A, Calvisi DF, Mertens JC, Trauner M, Benedetti A, Maroni L, Vaquero J, Macias RIR, Raggi C, Perugorria MJ, Gaudio E, Boberg KM, Marin JJG, Alvaro D. Expert consensus document: Cholangiocarcinoma: current knowledge and future perspectives consensus statement from the European Network for the Study of Cholangiocarcinoma (ENS-CCA). Nat Rev Gastroenterol Hepatol 2016; 13:261-80. [PMID: 27095655 DOI: 10.1038/nrgastro.2016.51] [Citation(s) in RCA: 892] [Impact Index Per Article: 111.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cholangiocarcinoma (CCA) is a heterogeneous group of malignancies with features of biliary tract differentiation. CCA is the second most common primary liver tumour and the incidence is increasing worldwide. CCA has high mortality owing to its aggressiveness, late diagnosis and refractory nature. In May 2015, the "European Network for the Study of Cholangiocarcinoma" (ENS-CCA: www.enscca.org or www.cholangiocarcinoma.eu) was created to promote and boost international research collaboration on the study of CCA at basic, translational and clinical level. In this Consensus Statement, we aim to provide valuable information on classifications, pathological features, risk factors, cells of origin, genetic and epigenetic modifications and current therapies available for this cancer. Moreover, future directions on basic and clinical investigations and plans for the ENS-CCA are highlighted.
Collapse
Affiliation(s)
- Jesus M Banales
- Department of Liver and Gastrointestinal Diseases, Biodonostia Health Research Institute - Donostia University Hospital, Ikerbasque, CIBERehd, Paseo del Dr. Begiristain s/n, E-20014, San Sebastian, Spain
| | - Vincenzo Cardinale
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Viale dell'Università 37, 00185, Rome, Italy
| | - Guido Carpino
- Department of Movement, Human and Health Sciences, University of Rome "Foro Italico", Piazza Lauro De Bosis 6, 00135, Rome, Italy
| | - Marco Marzioni
- Department of Clinic and Molecular Sciences, Polytechnic University of Marche, Via Tronto 10, 60020, Ancona, Italy
| | - Jesper B Andersen
- Biotech Research and Innovation Centre, University of Copenhagen, Ole Maaløes Vej 5, DK-2200, Copenhagen N, Denmark
| | - Pietro Invernizzi
- Humanitas Clinical and Research Center, Via Manzoni 56, Rozzano, 20089, Milan, Italy
- Program for Autoimmune Liver Diseases, International Center for Digestive Health, Department of Medicine and Surgery, University of Milan-Bicocca, Via Cadore 48, 20900, Monza, Italy
| | - Guro E Lind
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Montebello, 0310, Oslo, Norway
| | - Trine Folseraas
- Department of Transplantation Medicine, Division of Cancer Medicine, Surgery and Transplantation, Oslo University Hospital, Rikshospitalet, Pb. 4950 Nydalen, N-0424, Oslo, Norway
| | - Stuart J Forbes
- MRC Centre for Regenerative Medicine, University of Edinburgh, 49 Little France Crescent, EH16 4SB, Edinburgh, United Kingdom
| | - Laura Fouassier
- INSERM UMR S938, Centre de Recherche Saint-Antoine, 184 rue du Faubourg Saint-Antoine, 75571, Paris cedex 12, Fondation ARC, 9 rue Guy Môquet 94803 Villejuif, France
| | - Andreas Geier
- Department of Internal Medicine II, University Hospital Würzburg, Oberdürrbacherstrasse 6, D-97080, Würzburg, Germany
| | - Diego F Calvisi
- Institute of Pathology, Universitätsmedizin Greifswald, Friedrich-Löffler-Strasse 23e, 17489, Greifswald, Germany
| | - Joachim C Mertens
- Division of Gastroenterology and Hepatology, University Hospital Zurich, Rämistrasse 100, 8091, Zürich, Switzerland
| | - Michael Trauner
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Waehringer Guertel 18-20, A-1090, Vienna, Austria
| | - Antonio Benedetti
- Department of Clinic and Molecular Sciences, Polytechnic University of Marche, Via Tronto 10, 60020, Ancona, Italy
| | - Luca Maroni
- Department of Clinic and Molecular Sciences, Polytechnic University of Marche, Via Tronto 10, 60020, Ancona, Italy
| | - Javier Vaquero
- INSERM UMR S938, Centre de Recherche Saint-Antoine, 184 rue du Faubourg Saint-Antoine, 75571, Paris cedex 12, Fondation ARC, 9 rue Guy Môquet 94803 Villejuif, France
| | - Rocio I R Macias
- Department of Physiology and Pharmacology, Experimental Hepatology and Drug Targeting (HEVEFARM), Campus Miguel de Unamuno, E.I.D. S-09, University of Salamanca, IBSAL, CIBERehd, 37007, Salamanca, Spain
| | - Chiara Raggi
- Humanitas Clinical and Research Center, Via Manzoni 56, Rozzano, 20089, Milan, Italy
| | - Maria J Perugorria
- Department of Liver and Gastrointestinal Diseases, Biodonostia Health Research Institute - Donostia University Hospital, Ikerbasque, CIBERehd, Paseo del Dr. Begiristain s/n, E-20014, San Sebastian, Spain
| | - Eugenio Gaudio
- Department of Anatomical, Histological, Forensic Medicine and Orthopedics Sciences, Sapienza University of Rome, Via Alfonso Borelli 50, 00161, Rome, Italy
| | - Kirsten M Boberg
- Department of Transplantation Medicine, Division of Cancer Medicine, Surgery and Transplantation, Oslo University Hospital, Rikshospitalet, Pb. 4950 Nydalen, N-0424, Oslo, Norway
| | - Jose J G Marin
- Department of Physiology and Pharmacology, Experimental Hepatology and Drug Targeting (HEVEFARM), Campus Miguel de Unamuno, E.I.D. S-09, University of Salamanca, IBSAL, CIBERehd, 37007, Salamanca, Spain
| | - Domenico Alvaro
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Viale dell'Università 37, 00185, Rome, Italy
| |
Collapse
|
32
|
Yang G, Zong H. Overexpression of PDZK1IP1, EEF1A2 and RPL41 genes in intrahepatic cholangiocarcinoma. Mol Med Rep 2016; 13:4786-90. [PMID: 27082702 DOI: 10.3892/mmr.2016.5110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Accepted: 02/24/2016] [Indexed: 11/05/2022] Open
Abstract
Intrahepatic cholangiocarcinoma (iCCA) is an aggressive malignancy in the liver, which is associated with a poor prognosis. However, the molecular pathogenesis of iCCA remains unclear. RNA-Seq for tumor and para-tumor sample pairs enables the characterization of changes in the gene expression profiles of patients with iCCA. The present study analyzed RNA‑Seq data of seven iCCA para‑tumor and tumor sample pairs. Differential gene expression analysis demonstrated significant upregulation of PDZK1IP1, EEF1A2 and RPL41 (ENSG00000279483) genes in the iCCA samples when compared with the matched para‑tumor samples. Furthermore, genes associated with the immune system, metabolism and metabolic energy were significantly downregulated in the iCCA tumor tissues, indicating that this is involved in the pathogenesis of iCCA. The present study aimed to elucidate the gene expression patterns associated with the tumorigenesis of iCCA by comparing tumor and normal tissues, in order to isolate novel diagnostic factors for iCCA.
Collapse
Affiliation(s)
- Guanghua Yang
- Department of General Surgery, Huashan Hospital Affiliated to Fudan University, Shanghai 200040, P.R. China
| | - Huajie Zong
- Department of General Surgery, Huashan Hospital Affiliated to Fudan University, Shanghai 200040, P.R. China
| |
Collapse
|
33
|
Bile Acid Signaling Is Involved in the Neurological Decline in a Murine Model of Acute Liver Failure. THE AMERICAN JOURNAL OF PATHOLOGY 2015; 186:312-23. [PMID: 26683664 DOI: 10.1016/j.ajpath.2015.10.005] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Revised: 10/06/2015] [Accepted: 10/09/2015] [Indexed: 12/13/2022]
Abstract
Hepatic encephalopathy is a serious neurological complication of liver failure. Serum bile acids are elevated after liver damage and may disrupt the blood-brain barrier and enter the brain. Our aim was to assess the role of serum bile acids in the neurological complications after acute liver failure. C57Bl/6 or cytochrome p450 7A1 knockout (Cyp7A1(-/-)) mice were fed a control, cholestyramine-containing, or bile acid-containing diet before azoxymethane (AOM)-induced acute liver failure. In parallel, mice were given an intracerebroventricular infusion of farnesoid X receptor (FXR) Vivo-morpholino before AOM injection. Liver damage, neurological decline, and molecular analyses of bile acid signaling were performed. Total bile acid levels were increased in the cortex of AOM-treated mice. Reducing serum bile acids via cholestyramine feeding or using Cyp7A1(-/-) mice reduced bile acid levels and delayed AOM-induced neurological decline, whereas cholic acid or deoxycholic acid feeding worsened AOM-induced neurological decline. The expression of bile acid signaling machinery apical sodium-dependent bile acid transporter, FXR, and small heterodimer partner increased in the frontal cortex, and blocking FXR signaling delayed AOM-induced neurological decline. In conclusion, circulating bile acids may play a pathological role during hepatic encephalopathy, although precisely how they dysregulate normal brain function is unknown. Strategies to minimize serum bile acid concentrations may reduce the severity of neurological complications associated with liver failure.
Collapse
|
34
|
McMillin M, Frampton G, Quinn M, Divan A, Grant S, Patel N, Newell-Rogers K, DeMorrow S. Suppression of the HPA Axis During Cholestasis Can Be Attributed to Hypothalamic Bile Acid Signaling. Mol Endocrinol 2015; 29:1720-30. [PMID: 26431088 DOI: 10.1210/me.2015-1087] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Suppression of the hypothalamic-pituitary-adrenal (HPA) axis has been shown to occur during cholestatic liver injury. Furthermore, we have demonstrated that in a model of cholestasis, serum bile acids gain entry into the brain via a leaky blood brain barrier and that hypothalamic bile acid content is increased. Therefore, the aim of the current study was to determine the effects of bile acid signaling on the HPA axis. The data presented show that HPA axis suppression during cholestatic liver injury, specifically circulating corticosterone levels and hypothalamic corticotropin releasing hormone (CRH) expression, can be attenuated by administration of the bile acid sequestrant cholestyramine. Secondly, treatment of hypothalamic neurons with various bile acids suppressed CRH expression and secretion in vitro. However, in vivo HPA axis suppression was only evident after the central injection of the bile acids taurocholic acid or glycochenodeoxycholic acid but not the other bile acids studied. Furthermore, we demonstrate that taurocholic acid and glycochenodeoxycholic acid are exerting their effects on hypothalamic CRH expression after their uptake through the apical sodium-dependent bile acid transporter and subsequent activation of the glucocorticoid receptor. Taken together with previous studies, our data support the hypothesis that during cholestatic liver injury, bile acids gain entry into the brain, are transported into neurons through the apical sodium-dependent bile acid transporter and can activate the glucocorticoid receptor to suppress the HPA axis. These data also lend themselves to the broader hypothesis that bile acids may act as central modulators of hypothalamic peptides that may be altered during liver disease.
Collapse
Affiliation(s)
- Matthew McMillin
- Central Texas Veterans Health Care System 76504 (M.M., G.F., S.G., S.D.); Department of Internal Medicine (M.M., G.F., S.G., N.P., S.D.), Texas A&M Health Science Center College of Medicine 76508; Digestive Disease Research Center (M.M., G.F., S.G., S.D.) 76508; and Department of Surgery (A.D., M.K.N.-R.), Baylor Scott & White Health, Temple 76508, Texas; and Signal Transduction Laboratory (M.Q.), National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina 27709
| | - Gabriel Frampton
- Central Texas Veterans Health Care System 76504 (M.M., G.F., S.G., S.D.); Department of Internal Medicine (M.M., G.F., S.G., N.P., S.D.), Texas A&M Health Science Center College of Medicine 76508; Digestive Disease Research Center (M.M., G.F., S.G., S.D.) 76508; and Department of Surgery (A.D., M.K.N.-R.), Baylor Scott & White Health, Temple 76508, Texas; and Signal Transduction Laboratory (M.Q.), National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina 27709
| | - Matthew Quinn
- Central Texas Veterans Health Care System 76504 (M.M., G.F., S.G., S.D.); Department of Internal Medicine (M.M., G.F., S.G., N.P., S.D.), Texas A&M Health Science Center College of Medicine 76508; Digestive Disease Research Center (M.M., G.F., S.G., S.D.) 76508; and Department of Surgery (A.D., M.K.N.-R.), Baylor Scott & White Health, Temple 76508, Texas; and Signal Transduction Laboratory (M.Q.), National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina 27709
| | - Ali Divan
- Central Texas Veterans Health Care System 76504 (M.M., G.F., S.G., S.D.); Department of Internal Medicine (M.M., G.F., S.G., N.P., S.D.), Texas A&M Health Science Center College of Medicine 76508; Digestive Disease Research Center (M.M., G.F., S.G., S.D.) 76508; and Department of Surgery (A.D., M.K.N.-R.), Baylor Scott & White Health, Temple 76508, Texas; and Signal Transduction Laboratory (M.Q.), National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina 27709
| | - Stephanie Grant
- Central Texas Veterans Health Care System 76504 (M.M., G.F., S.G., S.D.); Department of Internal Medicine (M.M., G.F., S.G., N.P., S.D.), Texas A&M Health Science Center College of Medicine 76508; Digestive Disease Research Center (M.M., G.F., S.G., S.D.) 76508; and Department of Surgery (A.D., M.K.N.-R.), Baylor Scott & White Health, Temple 76508, Texas; and Signal Transduction Laboratory (M.Q.), National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina 27709
| | - Nisha Patel
- Central Texas Veterans Health Care System 76504 (M.M., G.F., S.G., S.D.); Department of Internal Medicine (M.M., G.F., S.G., N.P., S.D.), Texas A&M Health Science Center College of Medicine 76508; Digestive Disease Research Center (M.M., G.F., S.G., S.D.) 76508; and Department of Surgery (A.D., M.K.N.-R.), Baylor Scott & White Health, Temple 76508, Texas; and Signal Transduction Laboratory (M.Q.), National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina 27709
| | - Karen Newell-Rogers
- Central Texas Veterans Health Care System 76504 (M.M., G.F., S.G., S.D.); Department of Internal Medicine (M.M., G.F., S.G., N.P., S.D.), Texas A&M Health Science Center College of Medicine 76508; Digestive Disease Research Center (M.M., G.F., S.G., S.D.) 76508; and Department of Surgery (A.D., M.K.N.-R.), Baylor Scott & White Health, Temple 76508, Texas; and Signal Transduction Laboratory (M.Q.), National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina 27709
| | - Sharon DeMorrow
- Central Texas Veterans Health Care System 76504 (M.M., G.F., S.G., S.D.); Department of Internal Medicine (M.M., G.F., S.G., N.P., S.D.), Texas A&M Health Science Center College of Medicine 76508; Digestive Disease Research Center (M.M., G.F., S.G., S.D.) 76508; and Department of Surgery (A.D., M.K.N.-R.), Baylor Scott & White Health, Temple 76508, Texas; and Signal Transduction Laboratory (M.Q.), National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina 27709
| |
Collapse
|
35
|
McMillin M, Frampton G, Tobin R, Dusio G, Smith J, Shin H, Newell-Rogers K, Grant S, DeMorrow S. TGR5 signaling reduces neuroinflammation during hepatic encephalopathy. J Neurochem 2015; 135:565-76. [PMID: 26179031 DOI: 10.1111/jnc.13243] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Revised: 06/29/2015] [Accepted: 06/30/2015] [Indexed: 12/29/2022]
Abstract
Hepatic encephalopathy (HE) is a serious neurological complication of acute and chronic liver failure. Expression of the neurosteroid/bile acid receptor Takeda G protein-coupled receptor 5 (TGR5) has been demonstrated in the brain and is thought to be neuroprotective. However, it is unknown how TGR5 signaling can influence the progression and associated neuroinflammation of HE. HE was induced in C57Bl/6 mice via intraperitoneal injection of azoxymethane (AOM) and tissue was collected throughout disease progression. TGR5 expression was elevated in the frontal cortex following AOM injection in mice. The cellular localization of TGR5 was found in both neurons and microglia in the cortex of C57Bl/6 mice. Central infusion of the TGR5 agonist, betulinic acid, prior to AOM injection delayed neurological decline, increased cortical cyclic adenosine monophosphate concentrations, reduced microglia activation and proliferation, and reduced proinflammatory cytokine production. Betulinic acid treatment in vitro reduced the neuronal expression of chemokine ligand 2, a chemokine previously demonstrated to contribute to HE pathogenesis. Lastly, treatment of the microglia cell line EOC-20 with conditioned media from betulinic acid-treated primary neurons decreased phagocytic activity and cytokine production. Together, these data identify that activation of TGR5, which is up-regulated during HE, alleviates neuroinflammation and improves outcomes of AOM-treated mice through neuron and microglia paracrine signaling.
Collapse
Affiliation(s)
- Matthew McMillin
- Central Texas Veterans Healthcare System, Temple, Texas, USA.,Department of Internal Medicine, Texas A&M Health Science Center, College of Medicine, Temple, Texas, USA
| | - Gabriel Frampton
- Central Texas Veterans Healthcare System, Temple, Texas, USA.,Department of Internal Medicine, Texas A&M Health Science Center, College of Medicine, Temple, Texas, USA
| | - Richard Tobin
- Department of Surgery, Texas A&M Health Science Center and Baylor Scott & White Health, College of Medicine, Temple, Texas, USA
| | - Giuseppina Dusio
- Department of Surgery, Texas A&M Health Science Center and Baylor Scott & White Health, College of Medicine, Temple, Texas, USA
| | - Jenny Smith
- Department of Internal Medicine, Texas A&M Health Science Center, College of Medicine, Temple, Texas, USA
| | - Hope Shin
- Department of Internal Medicine, Texas A&M Health Science Center, College of Medicine, Temple, Texas, USA
| | - Karen Newell-Rogers
- Department of Surgery, Texas A&M Health Science Center and Baylor Scott & White Health, College of Medicine, Temple, Texas, USA
| | - Stephanie Grant
- Central Texas Veterans Healthcare System, Temple, Texas, USA.,Department of Internal Medicine, Texas A&M Health Science Center, College of Medicine, Temple, Texas, USA
| | - Sharon DeMorrow
- Central Texas Veterans Healthcare System, Temple, Texas, USA.,Department of Internal Medicine, Texas A&M Health Science Center, College of Medicine, Temple, Texas, USA.,Digestive Disease Research Center, Baylor Scott & White Health, Temple, Texas, USA
| |
Collapse
|
36
|
TGFβ1 exacerbates blood-brain barrier permeability in a mouse model of hepatic encephalopathy via upregulation of MMP9 and downregulation of claudin-5. J Transl Med 2015; 95:903-13. [PMID: 26006017 PMCID: PMC5040071 DOI: 10.1038/labinvest.2015.70] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Revised: 04/09/2015] [Accepted: 04/26/2015] [Indexed: 02/07/2023] Open
Abstract
Recent studies have found that vasogenic brain edema is present during hepatic encephalopathy following acute liver failure and is dependent on increased matrix metalloproteinase 9 (MMP9) activity and downregulation of tight junction proteins. Furthermore, circulating transforming growth factor β1 (TGFβ1) is increased following liver damage and may promote endothelial cell permeability. This study aimed to assess whether increased circulating TGFβ1 drives changes in tight junction protein expression and MMP9 activity following acute liver failure. Blood-brain barrier permeability was assessed in azoxymethane (AOM)-treated mice at 6, 12, and 18 h post-injection via Evan's blue extravasation. Monolayers of immortalized mouse brain endothelial cells (bEnd.3) were treated with recombinant TGFβ1 (rTGFβ1) and permeability to fluorescein isothiocyanate-dextran (FITC-dextran), MMP9 and claudin-5 expression was assessed. Antagonism of TGFβ1 signaling was performed in vivo to determine its role in blood-brain barrier permeability. Blood-brain barrier permeability was increased in mice at 18 h following AOM injection. Treatment of bEnd.3 cells with rTGFβ1 led to a dose-dependent increase of MMP9 expression as well as a suppression of claudin-5 expression. These effects of rTGFβ1 on MMP9 and claudin-5 expression could be reversed following treatment with a SMAD3 inhibitor. AOM-treated mice injected with neutralizing antibodies against TGFβ demonstrated significantly reduced blood-brain barrier permeability. Blood-brain barrier permeability is induced in AOM mice via a mechanism involving the TGFβ1-driven SMAD3-dependent upregulation of MMP9 expression and decrease of claudin-5 expression. Therefore, treatment modalities aimed at reducing TGFβ1 levels or SMAD3 activity may be beneficial in promoting blood-brain barrier integrity following liver failure.
Collapse
|
37
|
Franchitto A, Onori P, Renzi A, Carpino G, Mancinelli R, Alvaro D, Gaudio E. Recent advances on the mechanisms regulating cholangiocyte proliferation and the significance of the neuroendocrine regulation of cholangiocyte pathophysiology. ANNALS OF TRANSLATIONAL MEDICINE 2014; 1:27. [PMID: 25332971 DOI: 10.3978/j.issn.2305-5839.2012.10.03] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 09/10/2012] [Accepted: 10/15/2012] [Indexed: 12/14/2022]
Abstract
Cholangiocytes are epithelial cells lining the biliary epithelium. Cholangiocytes play several key roles in the modification of ductal bile and are also the target cells in chronic cholestatic liver diseases (i.e., cholangiopathies) such as PSC, PBC, polycystic liver disease (PCLD) and cholangiocarcinoma (CCA). During these pathologies, cholangiocytes (which in normal condition are in a quiescent state) begin to proliferate acquiring phenotypes of neuroendocrine cells, and start secreting different cytokines, growth factors, neuropeptides, and hormones to modulate cholangiocytes proliferation and interaction with the surrounding environment, trying to reestablish the balance between proliferation/loss of cholangiocytes for the maintenance of biliary homeostasis. The purpose of this review is to summarize the recent findings on the mechanisms regulating cholangiocyte proliferation and the significance of the neuroendocrine regulation of cholangiocyte pathophysiology. To clarify the mechanisms of action of these factors we will provide new potential strategies for the management of chronic liver diseases.
Collapse
Affiliation(s)
- Antonio Franchitto
- 1 Department of Anatomical, Histological, Forensic Medicine and Orthopedic Sciences, University of Rome "Sapienza", Rome, Italy ; 2 Eleonora Lorillard Spencer-Cenci Foundation, Rome, Italy ; 3 Department of Experimental Medicine, University of L'Aquila, L'Aquila, Italy ; 4 Department of Health Science, University of Rome "Foro Italico", Italy ; 5 Department of Medico-Surgical Sciences and Biotechnologies, Polo Pontino, Sapienza, University of Rome "Sapienza", Rome, Italy
| | - Paolo Onori
- 1 Department of Anatomical, Histological, Forensic Medicine and Orthopedic Sciences, University of Rome "Sapienza", Rome, Italy ; 2 Eleonora Lorillard Spencer-Cenci Foundation, Rome, Italy ; 3 Department of Experimental Medicine, University of L'Aquila, L'Aquila, Italy ; 4 Department of Health Science, University of Rome "Foro Italico", Italy ; 5 Department of Medico-Surgical Sciences and Biotechnologies, Polo Pontino, Sapienza, University of Rome "Sapienza", Rome, Italy
| | - Anastasia Renzi
- 1 Department of Anatomical, Histological, Forensic Medicine and Orthopedic Sciences, University of Rome "Sapienza", Rome, Italy ; 2 Eleonora Lorillard Spencer-Cenci Foundation, Rome, Italy ; 3 Department of Experimental Medicine, University of L'Aquila, L'Aquila, Italy ; 4 Department of Health Science, University of Rome "Foro Italico", Italy ; 5 Department of Medico-Surgical Sciences and Biotechnologies, Polo Pontino, Sapienza, University of Rome "Sapienza", Rome, Italy
| | - Guido Carpino
- 1 Department of Anatomical, Histological, Forensic Medicine and Orthopedic Sciences, University of Rome "Sapienza", Rome, Italy ; 2 Eleonora Lorillard Spencer-Cenci Foundation, Rome, Italy ; 3 Department of Experimental Medicine, University of L'Aquila, L'Aquila, Italy ; 4 Department of Health Science, University of Rome "Foro Italico", Italy ; 5 Department of Medico-Surgical Sciences and Biotechnologies, Polo Pontino, Sapienza, University of Rome "Sapienza", Rome, Italy
| | - Romina Mancinelli
- 1 Department of Anatomical, Histological, Forensic Medicine and Orthopedic Sciences, University of Rome "Sapienza", Rome, Italy ; 2 Eleonora Lorillard Spencer-Cenci Foundation, Rome, Italy ; 3 Department of Experimental Medicine, University of L'Aquila, L'Aquila, Italy ; 4 Department of Health Science, University of Rome "Foro Italico", Italy ; 5 Department of Medico-Surgical Sciences and Biotechnologies, Polo Pontino, Sapienza, University of Rome "Sapienza", Rome, Italy
| | - Domenico Alvaro
- 1 Department of Anatomical, Histological, Forensic Medicine and Orthopedic Sciences, University of Rome "Sapienza", Rome, Italy ; 2 Eleonora Lorillard Spencer-Cenci Foundation, Rome, Italy ; 3 Department of Experimental Medicine, University of L'Aquila, L'Aquila, Italy ; 4 Department of Health Science, University of Rome "Foro Italico", Italy ; 5 Department of Medico-Surgical Sciences and Biotechnologies, Polo Pontino, Sapienza, University of Rome "Sapienza", Rome, Italy
| | - Eugenio Gaudio
- 1 Department of Anatomical, Histological, Forensic Medicine and Orthopedic Sciences, University of Rome "Sapienza", Rome, Italy ; 2 Eleonora Lorillard Spencer-Cenci Foundation, Rome, Italy ; 3 Department of Experimental Medicine, University of L'Aquila, L'Aquila, Italy ; 4 Department of Health Science, University of Rome "Foro Italico", Italy ; 5 Department of Medico-Surgical Sciences and Biotechnologies, Polo Pontino, Sapienza, University of Rome "Sapienza", Rome, Italy
| |
Collapse
|
38
|
Herr F, Horndasch M, Howe D, Baal N, Goyal P, Fischer S, Zygmunt M, Preissner KT. Human placenta-derived Wnt-5a induces the expression of ICAM-1 and VCAM-1 in CD133(+)CD34(+)-hematopoietic progenitor cells. Reprod Biol 2014; 14:262-75. [PMID: 25454492 DOI: 10.1016/j.repbio.2014.08.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2013] [Revised: 08/15/2014] [Accepted: 08/21/2014] [Indexed: 01/20/2023]
Abstract
Angiogenesis and vascular development are essential for fetal development and growth, whereby early pregnancy loss and other pregnancy-related pathologies have been linked to aberrant vascular development. As Wnt signalling has been suggested to play a role in the vascularization of chorionic villi, we investigated the expression of Wnt family members in trophoblasts and stromal cells isolated from chorionic villi of early placenta and the influence of Wnt signalling on CD133(+)CD34(+)-hematopoietic progenitor (CD133(+)CD34(+)) cells. Wnt-5a was expressed in human placental stromal cells and to a lesser extent in human trophoblast cells. rWnt-5a impeded migration and induced adhesion of CD133(+)CD34(+) cells, in accordance with the expression of adhesion proteins, intercellular adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1). rWnt-5a-related regulation of the ICAM-1 and VCAM-1 expression were dependent on the release of Ca2+ and the activation of transcription factor - nuclear factor of activated T-cells (NF-AT). We propose that Wnt-5a is required during early placenta development to mediate adhesion and homing of CD133(+)CD34(+) cells.
Collapse
Affiliation(s)
- Friederike Herr
- Department of Obstetrics and Gynecology, Ernst-Moritz-Arndt-University, D-17475 Greifswald, Germany
| | - Manuela Horndasch
- Department of Obstetrics & Gynecology, Justus-Liebig-University, D-35385 Giessen, Germany
| | - Désirée Howe
- Department of Obstetrics & Gynecology, Justus-Liebig-University, D-35385 Giessen, Germany; Department of Anesthesiology, Justus-Liebig-University, D-35385 Giessen, Germany
| | - Nelli Baal
- Department of Obstetrics & Gynecology, Justus-Liebig-University, D-35385 Giessen, Germany; Institute for Clinical Immunology & Transfusion Medicine, Justus-Liebig-University, D-35385 Giessen, Germany
| | - Pankaj Goyal
- Department of Obstetrics and Gynecology, Ernst-Moritz-Arndt-University, D-17475 Greifswald, Germany; Department of Biotechnology, School of Life Sciences, Central University of Rajasthan NH-8, Bandar Sindri, Distt. Ajmer, Rajasthan 305801, India
| | - Silvia Fischer
- Institute of Biochemistry, Medical School, Justus-Liebig-University, D-35392 Giessen, Germany
| | - Marek Zygmunt
- Department of Obstetrics and Gynecology, Ernst-Moritz-Arndt-University, D-17475 Greifswald, Germany
| | - Klaus T Preissner
- Institute of Biochemistry, Medical School, Justus-Liebig-University, D-35392 Giessen, Germany.
| |
Collapse
|
39
|
McMillin M, Frampton G, Thompson M, Galindo C, Standeford H, Whittington E, Alpini G, DeMorrow S. Neuronal CCL2 is upregulated during hepatic encephalopathy and contributes to microglia activation and neurological decline. J Neuroinflammation 2014; 11:121. [PMID: 25012628 PMCID: PMC4128607 DOI: 10.1186/1742-2094-11-121] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Accepted: 06/30/2014] [Indexed: 12/31/2022] Open
Abstract
Background Acute liver failure leads to systemic complications with one of the most dangerous being a decline in neurological function, termed hepatic encephalopathy. Neurological dysfunction is exacerbated by an increase of toxic metabolites in the brain that lead to neuroinflammation. Following various liver diseases, hepatic and circulating chemokines, such as chemokine ligand 2 (CCL2), are elevated, though their effects on the brain following acute liver injury and subsequent hepatic encephalopathy are unknown. CCL2 is known to activate microglia in other neuropathies, leading to a proinflammatory response. However, the effects of CCL2 on microglia activation and the pathogenesis of hepatic encephalopathy following acute liver injury remain to be determined. Methods Hepatic encephalopathy was induced in mice via injection of azoxymethane (AOM) in the presence or absence of INCB 3284 dimesylate (INCB), a chemokine receptor 2 inhibitor, or C 021 dihydrochloride (C021), a chemokine receptor 4 inhibitor. Mice were monitored for neurological decline and time to coma (loss of all reflexes) was recorded. Tissue was collected at coma and used for real-time PCR, immunoblots, ELISA, or immunostaining analyses to assess the activation of microglia and consequences on pro-inflammatory cytokine expression. Results Following AOM administration, microglia activation was significantly increased in AOM-treated mice compared to controls. Concentrations of CCL2 in the liver, serum, and cortex were significantly elevated in AOM-treated mice compared to controls. Systemic administration of INCB or C021 reduced liver damage as assessed by serum liver enzyme biochemistry. Administration of INCB or C021 significantly improved the neurological outcomes of AOM-treated mice, reduced microglia activation, reduced phosphorylation of ERK1/2, and alleviated AOM-induced cytokine upregulation. Conclusions These findings suggest that CCL2 is elevated systemically following acute liver injury and that CCL2 is involved in both the microglia activation and neurological decline associated with hepatic encephalopathy. Methods used to modulate CCL2 levels and/or reduce CCR2/CCR4 activity may be potential therapeutic targets for the management of hepatic encephalopathy due to acute liver injury.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Sharon DeMorrow
- Department of Internal Medicine, Texas A&M Health Science Center, College of Medicine, 1901 South 1st Street, Building 205, Temple, Texas, USA.
| |
Collapse
|
40
|
Quinn M, McMillin M, Galindo C, Frampton G, Pae HY, DeMorrow S. Bile acids permeabilize the blood brain barrier after bile duct ligation in rats via Rac1-dependent mechanisms. Dig Liver Dis 2014; 46:527-34. [PMID: 24629820 PMCID: PMC4065628 DOI: 10.1016/j.dld.2014.01.159] [Citation(s) in RCA: 157] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Revised: 01/10/2014] [Accepted: 01/28/2014] [Indexed: 02/08/2023]
Abstract
BACKGROUND The blood brain barrier tightly regulates the passage of molecules into the brain and becomes leaky following obstructive cholestasis. The aim of this study was to determine if increased serum bile acids observed during cholestasis permeabilize the blood brain barrier. METHODS Rats underwent bile duct ligation or deoxycholic or chenodeoxycholic acid injections and blood brain barrier permeability assessed. In vitro, the permeability of rat brain microvessel endothelial cell monolayers, the expression and phosphorylation of occludin, ZO-1 and ZO-2 as well as the activity of Rac1 was assessed after treatment with plasma from cholestatic rats, or bile acid treatment, in the presence of a Rac1 inhibitor. RESULTS Blood brain barrier permeability was increased in vivo and in vitro following bile duct ligation or treatment with bile acids. Associated with the bile acid-stimulated increase in endothelial cell monolayer permeability was elevated Rac1 activity and increased phosphorylation of occludin. Pretreatment of endothelial cell monolayers with a Rac1 inhibitor prevented the effects of bile acid treatment on occludin phosphorylation and monolayer permeability. CONCLUSIONS These data suggest that increased circulating serum bile acids may contribute to the increased permeability of the blood brain barrier seen during obstructive cholestasis via disruption of tight junctions.
Collapse
Affiliation(s)
- Matthew Quinn
- Department of Internal Medicine, Texas A&M Health Science Center College of Medicine, Temple, Texas, USA
| | - Matthew McMillin
- Department of Internal Medicine, Texas A&M Health Science Center College of Medicine, Temple, Texas, USA
| | - Cheryl Galindo
- Department of Internal Medicine, Texas A&M Health Science Center College of Medicine, Temple, Texas, USA
| | - Gabriel Frampton
- Department of Internal Medicine, Texas A&M Health Science Center College of Medicine, Temple, Texas, USA
| | - Hae Yong Pae
- Department of Internal Medicine, Texas A&M Health Science Center College of Medicine, Temple, Texas, USA
| | - Sharon DeMorrow
- Department of Internal Medicine, Texas A&M Health Science Center College of Medicine, Temple, Texas, USA,Digestive Disease Research Center, Scott & White Hospital, Temple, Texas, USA,Central Texas Veterans Health Care System, Temple, Texas, USA
| |
Collapse
|
41
|
Loilome W, Bungkanjana P, Techasen A, Namwat N, Yongvanit P, Puapairoj A, Khuntikeo N, Riggins GJ. Activated macrophages promote Wnt/β-catenin signaling in cholangiocarcinoma cells. Tumour Biol 2014; 35:5357-67. [PMID: 24549785 DOI: 10.1007/s13277-014-1698-2] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Accepted: 01/26/2014] [Indexed: 12/13/2022] Open
Abstract
The Wnt/β-catenin signaling pathway is pathologically activated in cholangiocarcinoma (CCA). Here, we determined the expression profile as well as biological role of activated Wnt/β-catenin signaling in CCA. The quantitative reverse transcription polymerase chain reaction demonstrated that Wnt3a, Wnt5a, and Wnt7b mRNA were significantly higher in CCA tissues than adjacent non-tumor tissues and normal liver tissues. Immunohistochemical staining revealed that Wnt3a, Wnt5a, and Wnt7b were positive in 92.1, 76.3, and 100 % of 38 CCA tissues studied. It was noted that Wnt3 had a low expression in tumor cells, whereas a high expression was mainly found in inflammatory cells. Interestingly, a high expression level of Wnt5a was significantly correlated to poor survival of CCA patients (P=0.009). Membrane localization of β-catenin was reduced in the tumors compared to normal bile duct epithelia, and we also found that 73.7 % of CCA cases showed the cytoplasmic localization. Inflammation is known to be a risk factor for CCA development, and we tested whether this might induce Wnt/β-catenin signaling. We found that lipopolysaccharides (LPS) elevated the expression of Wnt3 both mRNA and protein levels in the macrophage cell line. Additionally, the conditioned media taken from LPS-induced activated macrophage culture promoted β-catenin accumulation in CCA cells. Furthermore, transient suppression of β-catenin by siRNA significantly induced growth inhibition of CCA cells, concurrently with decreasing cyclin D1 protein level. In conclusion, the present study reports the abundant expression of Wnt protein family and β-catenin in CCA as well as the effect of inflammatory condition on Wnt/β-catenin activation in CCA cells. Importantly, abrogation of β-catenin expression caused significant CCA cell growth inhibition. Thus, the Wnt/β-catenin signaling pathway may contribute to CCA cell proliferation and hence may serve as a prognostic marker for CCA progression and provide a potential target for CCA therapy.
Collapse
Affiliation(s)
- Watcharin Loilome
- Department of Biochemistry and Liver Fluke and Cholangiocarcinoma Research Center, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand,
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Bogaerts E, Heindryckx F, Vandewynckel YP, Van Grunsven LA, Van Vlierberghe H. The roles of transforming growth factor-β, Wnt, Notch and hypoxia on liver progenitor cells in primary liver tumours (Review). Int J Oncol 2014; 44:1015-22. [PMID: 24504124 PMCID: PMC3977811 DOI: 10.3892/ijo.2014.2286] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Accepted: 11/28/2013] [Indexed: 12/11/2022] Open
Abstract
Primary liver tumours have a high incidence and mortality. The most important forms are hepatocellular carcinoma and intrahepatic cholangiocarcinoma, both can occur together in the mixed phenotype hepatocellular-cholangiocarcinoma. Liver progenitor cells (LPCs) are bipotential stem cells activated in case of severe liver damage and are capable of forming both cholangiocytes and hepatocytes. Possibly, alterations in Wnt, transforming growth factor-β, Notch and hypoxia pathways in these LPCs can cause them to give rise to cancer stem cells, capable of driving tumourigenesis. In this review, we summarize and discuss current knowledge on the role of these pathways in LPC activation and differentiation during hepatocarcinogenesis.
Collapse
Affiliation(s)
- Eliene Bogaerts
- Department of Gastroenterology and Hepatology, 1K12, Ghent University Hospital, 9000 Gent, Belgium
| | - Femke Heindryckx
- Department of Medical Biochemistry and Microbiology, Uppsala University, 751 23 Uppsala, Sweden
| | - Yves-Paul Vandewynckel
- Department of Gastroenterology and Hepatology, 1K12, Ghent University Hospital, 9000 Gent, Belgium
| | - Leo A Van Grunsven
- Department of Cell Biology, Liver Cell Biology Lab, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Hans Van Vlierberghe
- Department of Gastroenterology and Hepatology, 1K12, Ghent University Hospital, 9000 Gent, Belgium
| |
Collapse
|
43
|
Leyva-Illades D, Demorrow S. Orphan G protein receptor GPR55 as an emerging target in cancer therapy and management. Cancer Manag Res 2013; 5:147-55. [PMID: 23869178 PMCID: PMC3706254 DOI: 10.2147/cmar.s35175] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
G protein-coupled receptors (GPCRs) modulate a vast array of cellular processes. The current review gives an overview of the general characteristics of GPCRs and their role in physiological conditions. In addition, it describes the current knowledge of the physiological and pathophysiological functions of GPR55, an orphan GPCR, and how it can be exploited as a therapeutic target to combat various cancers.
Collapse
Affiliation(s)
- Dinorah Leyva-Illades
- Digestive Disease Research Center, Scott and White Hospital, Temple, TX, USA ; Department of Internal MedicineTexas A&M Health Science Center, Temple, TX, USA ; Research Service, Central Texas Veterans Health Care System, Temple, TX, USA
| | | |
Collapse
|
44
|
Minicis SD, Kisseleva T, Francis H, Baroni GS, Benedetti A, Brenner D, Alvaro D, Alpini G, Marzioni M. Liver carcinogenesis: rodent models of hepatocarcinoma and cholangiocarcinoma. Dig Liver Dis 2013; 45. [PMID: 23177172 PMCID: PMC3716909 DOI: 10.1016/j.dld.2012.10.008] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Hepatocellular carcinoma and cholangiocarcinoma are primary liver cancers, both represent a growing challenge for clinicians due to their increasing morbidity and mortality. In the last few years a number of in vivo models of hepatocellular carcinoma and cholangiocarcinoma have been developed. The study of these models is providing a significant contribution in unveiling the pathophysiology of primary liver malignancies. They are also fundamental tools to evaluate newly designed molecules to be tested as new potential therapeutic agents in a pre-clinical set. Technical aspects of each model are critical steps, and they should always be considered in order to appropriately interpret the findings of a study or its planning. The purpose of this review is to describe the technical and experimental features of the most significant rodent models, highlighting similarities or differences between the corresponding human diseases. The first part is dedicated to the discussion of models of hepatocellular carcinoma, developed using toxic agents, or through dietary or genetic manipulations. In the second we will address models of cholangiocarcinoma developed in rats or mice by toxin administration, genetic manipulation and/or bile duct incannulation or surgery. Xenograft or syngenic models are also proposed.
Collapse
Affiliation(s)
- Samuele De Minicis
- Department of Gastroenterology, Università Politecnica delle Marche, Ancona, Italy
| | - Tatiana Kisseleva
- Division of Gastroenterology, Department of Medicine, University of California San Diego, School of Medicine, CA, United States
| | - Heather Francis
- Division Research, Central Texas Veterans Health Care System, Scott & White Digestive Disease Research Center, Department of Medicine, Division Gastroenterology, Scott & White Hospital and Texas A&M Health Science Center, College of Medicine, Temple, TX, United States
| | | | - Antonio Benedetti
- Department of Gastroenterology, Università Politecnica delle Marche, Ancona, Italy
| | - David Brenner
- Division of Gastroenterology, Department of Medicine, University of California San Diego, School of Medicine, CA, United States
| | - Domenico Alvaro
- Division of Gastroenterology, Polo Pontino, Università degli Studi “La Sapienza”, Rome, Italy
| | - Gianfranco Alpini
- Division of Gastroenterology, Department of Medicine, University of California San Diego, School of Medicine, CA, United States,Co-corresponding author. Tel.: +1 254 743 1041/1044; fax: +1 254 743 0378/0555. (M. Marzioni)
| | - Marco Marzioni
- Department of Gastroenterology, Università Politecnica delle Marche, Ancona, Italy,Corresponding author at: Department of Gastroenterology, Università Politecnica delle Marche, Nuovo Polo Didattico, III Piano, Via Tronto 10, 60020 Ancona, Italy. Tel.: +39 0712206043; fax: +39 0712206044
| |
Collapse
|
45
|
Oh JH, Lee JY, Baeg MK, Han KH, Choi MG, Park JM. Antineoplastic Effect of WIN 55,212-2, a Cannabinoid Agonist, in a Murine Xenograft Model of Gastric Cancer. Chemotherapy 2013; 59:200-6. [DOI: 10.1159/000355666] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2013] [Accepted: 09/09/2013] [Indexed: 11/19/2022]
|
46
|
Frampton G, Ueno Y, Quinn M, McMillin M, Pae HY, Galindo C, Leyva-Illades D, DeMorrow S. The novel growth factor, progranulin, stimulates mouse cholangiocyte proliferation via sirtuin-1-mediated inactivation of FOXO1. Am J Physiol Gastrointest Liver Physiol 2012; 303:G1202-11. [PMID: 23086914 PMCID: PMC3532458 DOI: 10.1152/ajpgi.00104.2012] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Progranulin (PGRN), a secreted growth factor, regulates the proliferation of various epithelial cells. Its mechanism of action is largely unknown. Sirtuin 1 (Sirt1) is a protein deacetylase that is known to regulate the transcriptional activity of the forkhead receptor FOXO1, thereby modulating the balance between proapoptotic and cell cycle-arresting genes. We have shown that PGRN is overexpressed in cholangiocarcinoma and stimulates proliferation. However, its effects on hyperplastic cholangiocyte proliferation are unknown. In the present study, the expression of PGRN and its downstream targets was determined after bile duct ligation (BDL) in mice and in a mouse cholangiocyte cell line after stimulation with PGRN. The effects of PGRN on cholangiocyte proliferation were assessed in sham-operated (sham) and BDL mice treated with PGRN or by specifically knocking down endogenous PGRN expression using Vivo-Morpholinos or short hairpin RNA. PGRN expression and secretion were upregulated in proliferating cholangiocytes isolated after BDL. Treatment of mice with PGRN increased biliary mass and cholangiocyte proliferation in vivo and in vitro and enhanced cholangiocyte proliferation observed after BDL. PGRN treatment decreased Sirt1 expression and increased the acetylation of FOXO1, resulting in the cytoplasmic accumulation of FOXO1 in cholangiocytes. Overexpression of Sirt1 in vitro prevented the proliferative effects of PGRN. Conversely, knocking down PGRN expression in vitro or in vivo inhibited cholangiocyte proliferation. In conclusion, these data suggest that the upregulation of PGRN may be a key feature stimulating cholangiocyte proliferation. Modulating PGRN levels may be a viable technique for regulating the balance between ductal proliferation and ductopenia observed in a variety of cholangiopathies.
Collapse
Affiliation(s)
- Gabriel Frampton
- 1Department of Internal Medicine, Texas A&M Health Science Center College of Medicine, Temple, Texas; ,2Digestive Disease Research Center, Scott & White Hospital, Temple, Texas; ,3Central Texas Veterans Health Care System, Temple, Texas; and
| | - Yoshiyuki Ueno
- 4Department of Gastroenterology, Yamagata University Faculty of Medicine, Yamagata, Japan
| | - Matthew Quinn
- 1Department of Internal Medicine, Texas A&M Health Science Center College of Medicine, Temple, Texas; ,2Digestive Disease Research Center, Scott & White Hospital, Temple, Texas; ,3Central Texas Veterans Health Care System, Temple, Texas; and
| | - Matthew McMillin
- 1Department of Internal Medicine, Texas A&M Health Science Center College of Medicine, Temple, Texas; ,2Digestive Disease Research Center, Scott & White Hospital, Temple, Texas; ,3Central Texas Veterans Health Care System, Temple, Texas; and
| | - Hae Yong Pae
- 1Department of Internal Medicine, Texas A&M Health Science Center College of Medicine, Temple, Texas; ,2Digestive Disease Research Center, Scott & White Hospital, Temple, Texas; ,3Central Texas Veterans Health Care System, Temple, Texas; and
| | - Cheryl Galindo
- 1Department of Internal Medicine, Texas A&M Health Science Center College of Medicine, Temple, Texas; ,2Digestive Disease Research Center, Scott & White Hospital, Temple, Texas; ,3Central Texas Veterans Health Care System, Temple, Texas; and
| | - Dinorah Leyva-Illades
- 1Department of Internal Medicine, Texas A&M Health Science Center College of Medicine, Temple, Texas; ,2Digestive Disease Research Center, Scott & White Hospital, Temple, Texas; ,3Central Texas Veterans Health Care System, Temple, Texas; and
| | - Sharon DeMorrow
- 1Department of Internal Medicine, Texas A&M Health Science Center College of Medicine, Temple, Texas; ,2Digestive Disease Research Center, Scott & White Hospital, Temple, Texas; ,3Central Texas Veterans Health Care System, Temple, Texas; and
| |
Collapse
|
47
|
Zhu SS, Li WG. Advances in research of signaling pathways in cholangiocarcinoma. Shijie Huaren Xiaohua Zazhi 2012; 20:2913-2919. [DOI: 10.11569/wcjd.v20.i30.2913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Epidemiological data indicate that the incidence and mortality of cholangiocarcinoma (CC) show an increasing trend worldwide over the past several years. Many pathophysiologic aspects of this neoplasia are still unknown and need to be fully discovered. However, progress has been recently made in understanding molecular mechanisms involved in the transformation and growth of malignant cholangiocytes. It is found that cholangiocarcinogenesis is a multistep cellular process evolving from a normal condition of the epithelial biliary cells and ending with malignant transformation through a chronic inflammation status. The bad prognosis related to CC justifies why a better identification of the molecular mechanisms involved in the growth and progression of this cancer is required for the development of effective preventive measures and valid treatment regimens. Signaling pathways can regulate substance and energy metabolism in organisms and are closely related to biological growth and development. This paper mainly introduces signaling pathways which occur in cholangiocarcinoma and their roles in cholangiocarcinoma cells.
Collapse
|
48
|
Monoamine oxidase A expression is suppressed in human cholangiocarcinoma via coordinated epigenetic and IL-6-driven events. J Transl Med 2012; 92:1451-60. [PMID: 22906985 PMCID: PMC3959781 DOI: 10.1038/labinvest.2012.110] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The secretion of dopamine and serotonin is increased in cholangiocarcinoma, which has growth-promoting effects. Monoamine oxidase A (MAOA), the degradation enzyme of serotonin and dopamine, is suppressed in cholangiocarcinoma via an unknown mechanism. The aims of this study were to (i) correlate MAOA immunoreactivity with pathophysiological parameters of cholangiocarcinoma, (ii) determine the mechanism by which MAOA expression is suppressed and (iii) evaluate the consequences of restored MAOA expression in cholangiocarcinoma. MAOA expression was assessed in cholangiocarcinoma and nonmalignant controls. The control of MAOA expression by promoter hypermethylation was evaluated and the contribution of interleukin-6 (IL-6) signaling to the suppression of MAOA expression was determined. The effects of MAOA overexpression on cholangiocarcinoma growth and invasion were also assessed. MAOA expression is correlated with differentiation, invasion and survival in cholangiocarcinoma. The MAOA promoter was hypermethylated immediately upstream of the start codon in cholangiocarcinoma samples and cell lines but not in nonmalignant counterparts. IL-6 signaling also decreased MAOA expression via a mechanism independent of hypermethylation, involving the regulation of the balance between SP-1 transcriptional activity and its inhibitor, R1 repressor. Inhibition of both IL-6 signaling and DNA methylation restored MAOA levels to those observed in cholangiocytes. Forced MAOA overexpression inhibited cholangiocarcinoma growth and invasion. MAOA expression is suppressed by the coordinated control of promoter hypermethylation and IL-6 signaling. MAOA may be a useful prognostic marker in the management of cholangiocarcinoma, and therapies designed to increase MAOA expression might prove beneficial in the treatment of cholangiocarcinoma.
Collapse
|
49
|
Francis H, DeMorrow S, Venter J, Onori P, White M, Gaudio E, Francis T, Greene JF, Tran S, Meininger CJ, Alpini G. Inhibition of histidine decarboxylase ablates the autocrine tumorigenic effects of histamine in human cholangiocarcinoma. Gut 2012; 61:753-64. [PMID: 21873469 PMCID: PMC3244572 DOI: 10.1136/gutjnl-2011-300007] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
BACKGROUND In several tumours the endogenous activity of histidine decarboxylase (HDC), the enzyme stimulating histamine synthesis, sustains the autocrine trophic effect of histamine on cancer progression. Cholangiocarcinoma is a biliary cancer with limited treatment options. Histamine interacts with four G-protein coupled receptors, H1-H4 histamine receptors (HRs). OBJECTIVE To determine the effects of histamine stimulation and inhibition of histamine synthesis (by modulation of HDC) on cholangiocarcinoma growth. METHODS In vitro studies were performed using multiple human cholangiocarcinoma lines. The expression levels of the histamine synthetic machinery and HRs were evaluated along with the effects of histamine stimulation and inhibition on cholangiocarcinoma proliferation. A xenograft tumour model was used to measure tumour volume after treatment with histamine or inhibition of histamine synthesis by manipulation of HDC. Vascular endothelial growth factor (VEGF) expression was measured in cholangiocarcinoma cells concomitant with the evaluation of the expression of CD31 in endothelial cells in the tumour microenvironment. RESULTS Cholangiocarcinoma cells display (1) enhanced HDC and decreased monoamine oxidase B expression resulting in increased histamine secretion; and (2) increased expression of H1-H4 HRs. Inhibition of HDC and antagonising H1HR decreased histamine secretion in Mz-ChA-1 cells. Long-term treatment with histamine increased proliferation and VEGF expression in cholangiocarcinoma that was blocked by HDC inhibitor and the H1HR antagonist. In nude mice, histamine increased tumour growth (up to 25%) and VEGF expression whereas inhibition of histamine synthesis (by reduction of HDC) ablated the autocrine stimulation of histamine on tumour growth (~80%) and VEGF expression. No changes in angiogenesis (evaluated by changes in CD31 immunoreactivity) were detected in the in vivo treatment groups. CONCLUSION The novel concept that an autocrine loop (consisting of enhanced histamine synthesis by HDC) sustains cholangiocarcinoma growth is proposed. Drug targeting of HDC may be important for treatment of patients with cholangiocarcinoma.
Collapse
Affiliation(s)
- Heather Francis
- Division of Research, Central Texas Veterans Health Care System, Scott & White and Texas A&M Health Science Center, College of Medicine, Temple, Texas, USA,Scott & White Digestive Disease Research Center, Texas A&M Health Science Center, College of Medicine, Temple, Texas, USA,Department of Medicine, Division of Gastroenterology, Scott & White and Texas A&M Health Science Center, College of Medicine, Temple, Texas, USA,Division of Research and Education, Scott & White and Texas A&M Health Science Center, College of Medicine, Temple, Texas, USA
| | - Sharon DeMorrow
- Division of Research, Central Texas Veterans Health Care System, Scott & White and Texas A&M Health Science Center, College of Medicine, Temple, Texas, USA,Scott & White Digestive Disease Research Center, Texas A&M Health Science Center, College of Medicine, Temple, Texas, USA,Department of Medicine, Division of Gastroenterology, Scott & White and Texas A&M Health Science Center, College of Medicine, Temple, Texas, USA
| | - Julie Venter
- Department of Medicine, Division of Gastroenterology, Scott & White and Texas A&M Health Science Center, College of Medicine, Temple, Texas, USA
| | - Paolo Onori
- Department of Experimental Medicine, State University of L’Aquila, Italy
| | - Mellanie White
- Department of Medicine, Division of Gastroenterology, Scott & White and Texas A&M Health Science Center, College of Medicine, Temple, Texas, USA
| | - Eugenio Gaudio
- Department of Anatomical, Histological, Forensic Medicine and Orthopedic Sciences, University of the Studies of La Sapienza, Rome, Italy
| | - Taylor Francis
- Division of Research and Education, Scott & White and Texas A&M Health Science Center, College of Medicine, Temple, Texas, USA
| | - John F Greene
- Department of Pathology, Scott & White and Texas A&M Health Science Center, College of Medicine, Temple, Texas, USA
| | - Steve Tran
- Division of Research and Education, Scott & White and Texas A&M Health Science Center, College of Medicine, Temple, Texas, USA
| | - Cynthia J Meininger
- Department of Systems Biology and Translational Medicine, Texas A&M Health Science Center, College of Medicine, Temple, Texas, USA
| | - Gianfranco Alpini
- Division of Research, Central Texas Veterans Health Care System, Scott & White and Texas A&M Health Science Center, College of Medicine, Temple, Texas, USA,Scott & White Digestive Disease Research Center, Texas A&M Health Science Center, College of Medicine, Temple, Texas, USA,Department of Medicine, Division of Gastroenterology, Scott & White and Texas A&M Health Science Center, College of Medicine, Temple, Texas, USA
| |
Collapse
|
50
|
Abstract
BACKGROUND AND OBJECTIVES Cholangiocarcinoma is a devastating cancer of biliary origin with limited treatment options. The growth factor, progranulin, is overexpressed in a number of tumours. The study aims were to assess the expression of progranulin in cholangiocarcinoma and to determine its effects on tumour growth. METHODS The expression and secretion of progranulin were evaluated in multiple cholangiocarcinoma cell lines and in clinical samples from patients with cholangiocarcinoma. The role of interleukin 6 (IL-6)-mediated signalling in the expression of progranulin was assessed using a combination of specific inhibitors and shRNA knockdown techniques. The effect of progranulin on proliferation and Akt activation and subsequent effects of FOXO1 phosphorylation were assessed in vitro. Progranulin knockdown cell lines were established, and the effects on cholangiocarcinoma growth were determined. RESULTS Progranulin expression and secretion were upregulated in cholangiocarcinoma cell lines and tissue, which were in part via IL-6-mediated activation of the ERK1/2/RSK1/C/EBPβ pathway. Blocking any of these signalling molecules, by either pharmacological inhibitors or shRNA, prevented the IL-6-dependent activation of progranulin expression. Treatment of cholangiocarcinoma cells with recombinant progranulin increased cell proliferation in vitro by a mechanism involving Akt phosphorylation leading to phosphorylation and nuclear extrusion of FOXO1. Knockdown of progranulin expression in cholangiocarcinoma cells decreased the expression of proliferating cellular nuclear antigen, a marker of proliferative capacity, and slowed tumour growth in vivo. CONCLUSIONS Evidence is presented for a role for progranulin as a novel growth factor regulating cholangiocarcinoma growth. Specific targeting of progranulin may represent an alternative for the development of therapeutic strategies.
Collapse
|