1
|
Mehdi SH, Xu YZ, Shultz LD, Kim E, Lee YG, Kendrick S, Yoon D. Development of New Diffuse Large B Cell Lymphoma Mouse Models. Cancers (Basel) 2024; 16:3006. [PMID: 39272864 PMCID: PMC11394112 DOI: 10.3390/cancers16173006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 08/09/2024] [Accepted: 08/27/2024] [Indexed: 09/15/2024] Open
Abstract
Diffuse large B cell lymphoma (DLBCL) is the most diagnosed, aggressive non-Hodgkin lymphoma, with ~40% of patients experiencing refractory or relapsed disease. Given the low response rates to current therapy, alternative treatment strategies are necessary to improve patient outcomes. Here, we sought to develop an easily accessible new xenograft mouse model that better recapitulates the human disease for preclinical studies. We generated two Luciferase (Luc)-EGFP-expressing human DLBCL cell lines representing the different DLBCL cell-of-origin subtypes. After intravenous injection of these cells into humanized NSG mice, we monitored the tumor growth and evaluated the organ-specific engraftment/progression period. Our results showed that human IL6-expressing NSG (NSG-IL6) mice were highly permissive for DLBCL cell growth. In NSG-IL6 mice, systemic engraftments of both U2932 activated B cell-like- and VAL germinal B cell-like-DLBCL (engraftment rate; 75% and 82%, respectively) were detected within 2nd-week post-injection. In the organ-specific ex vivo evaluation, both U2932-Luc and VAL-Luc cells were initially engrafted and expanded in the spleen, liver, and lung and subsequently in the skeleton, ovary, and brain. Consistent with the dual BCL2/MYC translocation association with poor patient outcomes, VAL cells showed heightened proliferation in human IL6-conditioned media and caused rapid tumor expansion and early death in the engrafted mice. We concluded that the U2932 and VAL cell-derived human IL6-expressing mouse models reproduced the clinical features of an aggressive DLBCL with a highly consistent pattern of tumor development. Based on these findings, NSG mice expressing human IL6 have the potential to serve as a new tool to develop DLBCL xenograft models to overcome the limitations of standard subcutaneous DLBCL xenografts.
Collapse
Affiliation(s)
- Syed Hassan Mehdi
- Myeloma Center, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Ying-Zhi Xu
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | | | - Eunkyung Kim
- Myeloma Center, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Yong Gu Lee
- College of Pharmacy, Institute of Pharmaceutical Sciences and Technology, Hanyang University, Ansan 15588, Republic of Korea
| | - Samantha Kendrick
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Donghoon Yoon
- Myeloma Center, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| |
Collapse
|
2
|
Hasanali ZS, Garfall AL, Burzenski L, Shultz LD, Tang Y, Kadu S, Sheppard NC, Liu W, Dopkin D, Vogl DT, Cohen AD, Waxman AJ, Susanibar-Adaniya SP, Carroll M, Stadtmauer EA, Allman D. Human IL-6 fosters long-term engraftment of patient-derived disease-driving myeloma cells in immunodeficient mice. JCI Insight 2024; 9:e177300. [PMID: 38713510 PMCID: PMC11141932 DOI: 10.1172/jci.insight.177300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 04/17/2024] [Indexed: 05/09/2024] Open
Abstract
Multiple myeloma is a largely incurable and life-threatening malignancy of antibody-secreting plasma cells. An effective and widely available animal model that recapitulates human myeloma and related plasma cell disorders is lacking. We show that busulfan-conditioned human IL-6-transgenic (hIL-6-transgenic) NSG (NSG+hIL6) mice reliably support the engraftment of malignant and premalignant human plasma cells, including from patients diagnosed with monoclonal gammopathy of undetermined significance, pre- and postrelapse myeloma, plasma cell leukemia, and amyloid light chain amyloidosis. Consistent with human disease, NSG+hIL6 mice engrafted with patient-derived myeloma cells developed serum M spikes, and a majority developed anemia, hypercalcemia, and/or bone lesions. Single-cell RNA sequencing showed nonmalignant and malignant cell engraftment, the latter expressing a wide array of mRNAs associated with myeloma cell survival and proliferation. Myeloma-engrafted mice given CAR T cells targeting plasma cells or bortezomib experienced reduced tumor burden. Our results establish NSG+hIL6 mice as an effective patient-derived xenograft model for study and preclinical drug development of multiple myeloma and related plasma cell disorders.
Collapse
Affiliation(s)
- Zainul S. Hasanali
- Division of Hematology Oncology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Alfred L. Garfall
- Division of Hematology Oncology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | | | - Yan Tang
- Center for Cellular Immunotherapies
| | | | - Neil C. Sheppard
- Center for Cellular Immunotherapies
- Department of Pathology and Laboratory Medicine, and
| | - Wei Liu
- Center for Cellular Immunotherapies
| | - Derek Dopkin
- Stem Cell and Xenograft Core Facility, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Dan T. Vogl
- Division of Hematology Oncology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Adam D. Cohen
- Division of Hematology Oncology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Adam J. Waxman
- Division of Hematology Oncology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | - Martin Carroll
- Division of Hematology Oncology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Stem Cell and Xenograft Core Facility, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Edward A. Stadtmauer
- Division of Hematology Oncology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - David Allman
- Department of Pathology and Laboratory Medicine, and
| |
Collapse
|
3
|
Hasanali ZS, Garfall AL, Burzenski L, Shultz LD, Tang Y, Kadu S, Sheppard NC, Dopkin D, Vogl DT, Cohen AD, Waxman AJ, Susanibar-Adaniya SP, Carroll M, Stadtmauer EA, Allman D. Human IL-6 fosters long-term engraftment of patient derived disease-driving myeloma cells in immunodeficient mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.21.576547. [PMID: 38328086 PMCID: PMC10849475 DOI: 10.1101/2024.01.21.576547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Multiple myeloma is a largely incurable and life-threatening malignancy of antibody-secreting plasma cells. An effective and widely available animal model that recapitulates human myeloma and related plasma cell disorders is lacking. We show that busulfan-conditioned hIL-6 transgenic NSG mice (NSG+hIL6) reliably support the engraftment of malignant and pre-malignant human plasma cells including from patients diagnosed with monoclonal gammopathy of undetermined significance, pre- and post-relapse myeloma, plasma cell leukemia, and AL amyloidosis. Consistent with human disease, NSG+hIL6 mice engrafted with patient-derived myeloma cells, developed serum M spikes, and a majority developed anemia, hypercalcemia, and/or bone lesions. Single cell RNA sequencing showed non-malignant and malignant cell engraftment, the latter expressing a wide array of mRNAs associated with myeloma cell survival and proliferation. Myeloma engrafted mice given CAR T-cells targeting plasma cells or bortezomib experienced reduced tumor burden. Our results establish NSG+hIL6 mice as an effective patient derived xenograft model for study and preclinical drug development of multiple myeloma and related plasma cell disorders.
Collapse
|
4
|
Kolkmann AM, Van Essen A, Post MJ, Moutsatsou P. Development of a Chemically Defined Medium for in vitro Expansion of Primary Bovine Satellite Cells. Front Bioeng Biotechnol 2022; 10:895289. [PMID: 35992337 PMCID: PMC9385969 DOI: 10.3389/fbioe.2022.895289] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Accepted: 04/18/2022] [Indexed: 11/19/2022] Open
Abstract
The use of fetal bovine serum (FBS) in animal cell culture media is widely spread since it provides a broad spectrum of molecules that are known to support cell attachment and growth. However, the harvest and collection procedures of FBS raise ethical concerns and serum is an ill-defined and expensive component. This is especially problematic when it comes to regulatory approval for food applications like cultured meat. The aim of this study is to develop a chemically defined, cost efficient serum-free and animal-free medium that supports the attachment and expansion of bovine myoblasts while maintaining their differentiation capacity. Bovine satellite cells were harvested and isolated from a fresh sample of skeletal muscle tissue and cultured in planar systems. The efficacy of the tested formulations was assessed with metabolic assays and cell counting techniques. Optical microscopy was used to observe cellular morphology and statistical analysis was applied. Based on a comprehensive literature analysis, a defined serum-free medium (SFM) composition was developed consisting of DMEM/F12 as basal medium, supplemented with L-ascorbic acid 2-phosphate, fibronectin, hydrocortisone, GlutaMAX™, albumin, ITS-X, hIL-6, α-linolenic acid, and growth factors such as FGF-2, VEGF, IGF-1, HGF, and PDGF-BB. To our knowledge, this is the first defined serum-free and animal free medium formulation specific for bovine myoblasts to date. We conclude that the SFM formulation supported exponential cell growth up to 97% of the serum-containing golden standard growth medium. All reagents used in this study are chemically defined.
Collapse
Affiliation(s)
- Anna M. Kolkmann
- Mosa Meat BV, Maastricht, Netherlands
- Department of Physiology, Maastricht University, Maastricht, Netherlands
| | | | - Mark J. Post
- Mosa Meat BV, Maastricht, Netherlands
- Department of Physiology, Maastricht University, Maastricht, Netherlands
| | - Panagiota Moutsatsou
- Mosa Meat BV, Maastricht, Netherlands
- Department of Physiology, Maastricht University, Maastricht, Netherlands
| |
Collapse
|
5
|
Antoon R, Wang XH, Saleh AH, Warrington J, Hedley DW, Keating A. Pancreatic cancer growth promoted by bone marrow mesenchymal stromal cell-derived IL-6 is reversed predominantly by IL-6 blockade. Cytotherapy 2022; 24:699-710. [PMID: 35473998 DOI: 10.1016/j.jcyt.2021.12.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 11/17/2021] [Accepted: 12/06/2021] [Indexed: 12/26/2022]
Abstract
Pancreatic cancer is a highly lethal cancer characterized by local invasiveness, early metastasis, recurrence and high resistance to current therapies. Extensive stroma or desmoplasia is a key histological feature of the disease, and interactions between cancer and stromal cells are critical for pancreatic cancer development and progression. Mesenchymal stromal cells [MSCs] exhibit preferential tropism to primary and metastatic tumor sites and may either suppress or support tumor growth. Although MSCs represent a potential source of pancreatic cancer stroma, their contribution to pancreatic tumor growth remains poorly known. Here, we show that bone marrow MSCs significantly contribute to pancreatic cancer growth in vitro and in vivo. Furthermore, MSCs create a pro-carcinogenic microenvironment through the release of key factors mediating growth and angiogenesis, including interleukin (IL)-6, IL-8, vascular endothelial growth factor and activation of STAT3 signaling in tumor cells. IL-6 released by MSCs was largely responsible for the pro-tumorigenic effects of MSCs. Knockdown of IL-6 expression in MSCs by small interfering RNA (siRNA) abolished the MSC growth-promoting effect in vitro, reducing tumor cell proliferation and clonogenic potential. In addition, in a heterotopic nude mouse model of human pancreatic tumor xenografts, blockade of IL-6 with the anti-IL-6 receptor antibody, tocilizumab, or of its downstream effector STAT3 with the small molecule STAT3 inhibitor S3I-201, abrogated MSC-mediated tumor promotion and delayed tumor formation significantly. Our data demonstrate that MSCs promote pancreatic cancer growth, with IL-6 produced by MSCs playing a pivotal role.
Collapse
Affiliation(s)
- Roula Antoon
- Krembil Research Institute, Toronto, ON, Canada; Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | | | - Amr H Saleh
- Krembil Research Institute, Toronto, ON, Canada; Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | | | - David W Hedley
- Princess Margaret Cancer Centre, Toronto, ON, Canada; Institute of Medical Science, University of Toronto, Toronto, ON, Canada; Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Armand Keating
- Krembil Research Institute, Toronto, ON, Canada; Princess Margaret Cancer Centre, Toronto, ON, Canada; Institute of Medical Science, University of Toronto, Toronto, ON, Canada; Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
6
|
Rauf A, Badoni H, Abu-Izneid T, Olatunde A, Rahman MM, Painuli S, Semwal P, Wilairatana P, Mubarak MS. Neuroinflammatory Markers: Key Indicators in the Pathology of Neurodegenerative Diseases. Molecules 2022; 27:molecules27103194. [PMID: 35630670 PMCID: PMC9146652 DOI: 10.3390/molecules27103194] [Citation(s) in RCA: 78] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Revised: 05/05/2022] [Accepted: 05/12/2022] [Indexed: 12/12/2022] Open
Abstract
Neuroinflammation, a protective response of the central nervous system (CNS), is associated with the pathogenesis of neurodegenerative diseases. The CNS is composed of neurons and glial cells consisting of microglia, oligodendrocytes, and astrocytes. Entry of any foreign pathogen activates the glial cells (astrocytes and microglia) and overactivation of these cells triggers the release of various neuroinflammatory markers (NMs), such as the tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), interleukin-1β (IL-10), nitric oxide (NO), and cyclooxygenase-2 (COX-2), among others. Various studies have shown the role of neuroinflammatory markers in the occurrence, diagnosis, and treatment of neurodegenerative diseases. These markers also trigger the formation of various other factors responsible for causing several neuronal diseases including Alzheimer’s disease (AD), Parkinson’s disease (PD), Huntington’s disease (HD), multiple sclerosis (MS), ischemia, and several others. This comprehensive review aims to reveal the mechanism of neuroinflammatory markers (NMs), which could cause different neurodegenerative disorders. Important NMs may represent pathophysiologic processes leading to the generation of neurodegenerative diseases. In addition, various molecular alterations related to neurodegenerative diseases are discussed. Identifying these NMs may assist in the early diagnosis and detection of therapeutic targets for treating various neurodegenerative diseases.
Collapse
Affiliation(s)
- Abdur Rauf
- Department of Chemistry, University of Swabi, Anbar 23561, Khyber Pakhtunkhwa, Pakistan
- Correspondence: (A.R.); (P.W.); (M.S.M.)
| | - Himani Badoni
- Department of Biotechnology, School of Applied and Life Sciences, Uttaranchal University, Premnagar, Dehradun 248006, India;
| | - Tareq Abu-Izneid
- Pharmaceutical Sciences Department, College of Pharmacy, Al Ain University for Science and Technology, Al Ain 64141, United Arab Emirates;
| | - Ahmed Olatunde
- Department of Medical Biochemistry, Abubakar Tafawa Balewa University, Bauchi 740272, Nigeria;
| | - Md. Mominur Rahman
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka 1207, Bangladesh;
| | - Sakshi Painuli
- Uttarakhand Council for Biotechnology (UCB), Premnagar, Dehradun 248007, India;
| | - Prabhakar Semwal
- Department of Life Sciences, Graphic Era (Deemed To Be University), Dehradun 248002, India;
| | - Polrat Wilairatana
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
- Correspondence: (A.R.); (P.W.); (M.S.M.)
| | - Mohammad S. Mubarak
- Department of Chemistry, The University of Jordan, Amman 11942, Jordan
- Correspondence: (A.R.); (P.W.); (M.S.M.)
| |
Collapse
|
7
|
Komarova N, Panova O, Titov A, Kuznetsov A. Aptamers Targeting Cardiac Biomarkers as an Analytical Tool for the Diagnostics of Cardiovascular Diseases: A Review. Biomedicines 2022; 10:biomedicines10051085. [PMID: 35625822 PMCID: PMC9138532 DOI: 10.3390/biomedicines10051085] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 05/05/2022] [Accepted: 05/06/2022] [Indexed: 02/04/2023] Open
Abstract
The detection of cardiac biomarkers is used for diagnostics, prognostics, and the risk assessment of cardiovascular diseases. The analysis of cardiac biomarkers is routinely performed with high-sensitivity immunological assays. Aptamers offer an attractive alternative to antibodies for analytical applications but, to date, are not widely practically implemented in diagnostics and medicinal research. This review summarizes the information on the most common cardiac biomarkers and the current state of aptamer research regarding these biomarkers. Aptamers as an analytical tool are well established for troponin I, troponin T, myoglobin, and C-reactive protein. For the rest of the considered cardiac biomarkers, the isolation of novel aptamers or more detailed characterization of the known aptamers are required. More attention should be addressed to the development of dual-aptamer sandwich detection assays and to the studies of aptamer sensing in alternative biological fluids. The universalization of aptamer-based biomarker detection platforms and the integration of aptamer-based sensing to clinical studies are demanded for the practical implementation of aptamers to routine diagnostics. Nevertheless, the wide usage of aptamers for the diagnostics of cardiovascular diseases is promising for the future, with respect to both point-of-care and laboratory testing.
Collapse
|
8
|
Tocilizumab overcomes chemotherapy resistance in mesenchymal stem-like breast cancer by negating autocrine IL-1A induction of IL-6. NPJ Breast Cancer 2022; 8:30. [PMID: 35260569 PMCID: PMC8904846 DOI: 10.1038/s41523-021-00371-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 11/23/2021] [Indexed: 12/19/2022] Open
Abstract
Triple-negative breast cancer (TNBC) patients with mesenchymal stem-like (MSL) subtype have responded poorly to chemotherapy whereas patients with basal-like 1 (BL1) subtype achieved the best clinical response. In order to gain insight into pathways that may contribute to the divergent sensitivity to chemotherapy, we compared the inflammatory profile of the two TNBC subtypes treated with docetaxel. Cellular signaling analysis determined that docetaxel activated MAPK pathway in MSL TNBCs but not BL1 TNBCs. The subsequent MAPK pathway activation in MSL TNBCs led to an IL-1A mediated cascade of autocrine inflammatory mediators including IL-6. Utilizing the humanized IL-6R antibody, tocilizumab, our in vitro and in vivo data show that MSL TNBCs treated with tocilizumab together with chemotherapy results in delayed tumor progression compared to MSL TNBCs treated with docetaxel alone. Our study highlights a molecular subset of TNBC that may be responsive to tocilizumab therapy for potential translational impact.
Collapse
|
9
|
Wang T, Sun X, Guo X, Zhang J, Yang J, Tao S, Guan J, Zhou L, Han J, Wang C, Yao H, Wang G. Ultraefficiently Calming Cytokine Storm Using Ti 3C 2T x MXene. SMALL METHODS 2021; 5:e2001108. [PMID: 33786372 PMCID: PMC7995020 DOI: 10.1002/smtd.202001108] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 12/17/2020] [Indexed: 05/17/2023]
Abstract
During the global outbreak of COVID-19 pandemic, "cytokine storm" conditions are regarded as the fatal step resulting in most mortality. Hemoperfusion is widely used to remove cytokines from the blood of severely ill patients to prevent uncontrolled inflammation induced by a cytokine storm. This article discoveres, for the first time, that 2D Ti3C2T x MXene sheet demonstrates an ultrahigh removal capability for typical cytokine interleukin-6. In particular, MXene shows a 13.4 times higher removal efficiency over traditional activated carbon absorbents. Molecular-level investigations reveal that MXene exhibits a strong chemisorption mechanism for immobilizing cytokine interleukin-6 molecules, which is different from activated carbon absorbents. MXene sheet also demonstrates excellent blood compatibility without any deleterious side influence on the composition of human blood. This work can open a new avenue to use MXene sheets as an ultraefficient hemoperfusion absorbent to eliminate the cytokine storm syndrome in treatment of severe COVID-19 patients.
Collapse
Affiliation(s)
- Tianyi Wang
- Faculty of ScienceUniversity of Technology SydneySydneyNSW2007Australia
| | - Xiaoyu Sun
- School of Chemistry and Chemical EngineeringYangzhou UniversityYangzhouJiangsu Province225002P. R. China
| | - Xin Guo
- Faculty of ScienceUniversity of Technology SydneySydneyNSW2007Australia
| | - Jinqiang Zhang
- Faculty of ScienceUniversity of Technology SydneySydneyNSW2007Australia
| | - Jian Yang
- School of Chemistry and Chemical EngineeringYangzhou UniversityYangzhouJiangsu Province225002P. R. China
| | - Shouxuan Tao
- School of Chemistry and Chemical EngineeringYangzhou UniversityYangzhouJiangsu Province225002P. R. China
| | - Jun Guan
- Clinical Medical CollegeNorthern Jiangsu People's HospitalYangzhou UniversityYangzhouJiangsu Province225009P. R. China
| | - Lin Zhou
- Clinical Medical CollegeNorthern Jiangsu People's HospitalYangzhou UniversityYangzhouJiangsu Province225009P. R. China
| | - Jie Han
- School of Chemistry and Chemical EngineeringYangzhou UniversityYangzhouJiangsu Province225002P. R. China
| | - Chengyin Wang
- School of Chemistry and Chemical EngineeringYangzhou UniversityYangzhouJiangsu Province225002P. R. China
| | - Hang Yao
- School of Chemistry and Chemical EngineeringYangzhou UniversityYangzhouJiangsu Province225002P. R. China
| | - Guoxiu Wang
- Faculty of ScienceUniversity of Technology SydneySydneyNSW2007Australia
| |
Collapse
|
10
|
Ng J, Guo F, Marneth AE, Ghanta S, Kwon MY, Keegan J, Liu X, Wright KT, Kamaz B, Cahill LA, Mullally A, Perrella MA, Lederer JA. Augmenting emergency granulopoiesis with CpG conditioned mesenchymal stromal cells in murine neutropenic sepsis. Blood Adv 2020; 4:4965-4979. [PMID: 33049055 PMCID: PMC7556132 DOI: 10.1182/bloodadvances.2020002556] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 09/02/2020] [Indexed: 12/17/2022] Open
Abstract
Patients with immune deficiencies from cancers and associated treatments represent a growing population within the intensive care unit with increased risk of morbidity and mortality from sepsis. Mesenchymal stromal cells (MSCs) are an integral part of the hematopoietic niche and express toll-like receptors, making them candidate cells to sense and translate pathogenic signals into an innate immune response. In this study, we demonstrate that MSCs administered therapeutically in a murine model of radiation-associated neutropenia have dual actions to confer a survival benefit in Pseudomonas aeruginosa pneumo-sepsis that is not from improved bacterial clearance. First, MSCs augment the neutrophil response to infection, an effect that is enhanced when MSCs are preconditioned with CpG oligodeoxynucleotide, a toll-like receptor 9 agonist. Using cytometry by time of flight, we identified proliferating neutrophils (Ly6GlowKi-67+) as the main expanded cell population within the bone marrow. Further analysis revealed that CpG-MSCs expand a lineage restricted progenitor population (Lin-Sca1+C-kit+CD150-CD48+) in the bone marrow, which corresponded to a doubling in the myeloid proliferation and differentiation potential in response to infection compared with control. Despite increased neutrophils, no reduction in organ bacterial count was observed between experimental groups. However, the second effect exerted by CpG-MSCs is to attenuate organ damage, particularly in the lungs. Neutrophils obtained from irradiated mice and cocultured with CpG-MSCs had decreased neutrophil extracellular trap formation, which was associated with decreased citrullinated H3 staining in the lungs of mice given CpG-MSCs in vivo. Thus, this preclinical study provides evidence for the therapeutic potential of MSCs in neutropenic sepsis.
Collapse
Affiliation(s)
- Julie Ng
- Division of Pulmonary and Critical Care, Department of Medicine
| | | | | | | | - Min-Young Kwon
- Division of Pulmonary and Critical Care, Department of Medicine
| | | | - Xiaoli Liu
- Division of Pulmonary and Critical Care, Department of Medicine
- Department of Pediatric Newborn Medicine, and
| | - Kyle T Wright
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | | | | | | | - Mark A Perrella
- Division of Pulmonary and Critical Care, Department of Medicine
- Department of Pediatric Newborn Medicine, and
| | | |
Collapse
|
11
|
Wajima D, Hourani S, Dodd W, Patel D, Jones C, Motwani K, Fazal HZ, Hosaka K, Hoh BL. Interleukin-6 Promotes Murine Estrogen Deficiency-Associated Cerebral Aneurysm Rupture. Neurosurgery 2020; 86:583-592. [PMID: 31264696 PMCID: PMC7317988 DOI: 10.1093/neuros/nyz220] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 03/08/2019] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Estrogen deficiency is associated with cerebral aneurysm rupture, but the precise mechanism is unknown. OBJECTIVE To test the hypothesis that IL-6 is required for the increase in aneurysm rupture rate observed in estrogen-deficient mice. METHODS We analyzed IL-6 expression in human cerebral aneurysms. We induced cerebral aneurysms in estrogen-deficient female C57BL/6 mice that had undergone 4-vinylcyclohexene diepoxide (VCD) treatment or bilateral ovariectomy (OVE). Mice were blindly randomized to selective IL-6 inhibition (IL-6 receptor [IL-6R] neutralizing antibody, n = 25) or control (isotype-matched IgG, n = 28). Murine cerebral arteries at the circle of Willis were assessed for aneurysm rupture and macrophage infiltration. RESULTS IL-6 is expressed in human cerebral aneurysms, but not in control arteries. Serum IL-6 is elevated in ovariectomized female mice compared to sham control (14.3 ± 1.7 pg/mL vs 7.4 ± 1.5 pg/mL, P = .008). Selective IL-6R inhibition suppressed cerebral aneurysm rupture in estrogen-deficient mice compared with control (VCD: 31.6% vs 70.0%, P = .026; OVE: 28.6% vs 65.2%, P = .019). IL-6R inhibition had no effect on formation or rupture rate in wild-type mice. IL-6R neutralizing antibody significantly reduced macrophage infiltration at the circle of Willis (1.9 ± 0.2 vs 5.7 ± 0.6 cells/2500 μm2; n = 8 vs n = 15; P < .001). CONCLUSION IL-6 is increased in the serum of estrogen-deficient mice and appears to play a role in promoting murine estrogen deficiency-associated cerebral aneurysm rupture via enhanced macrophage infiltration at the circle of Willis. Inhibition of IL-6 signaling via IL-6 receptor neutralizing antibody inhibits aneurysm rupture in estrogen-deficient mice. IL-6 receptor inhibition had no effect on aneurysm formation or rupture in wild-type animals.
Collapse
Affiliation(s)
- Daisuke Wajima
- Department of Neurosurgery, University of Florida, Gainesville, Florida.,Department of Neurosurgery, School of Medicine, Nara Medical University, Kashihara, Japan
| | - Siham Hourani
- Department of Neurosurgery, University of Florida, Gainesville, Florida
| | - William Dodd
- Department of Neurosurgery, University of Florida, Gainesville, Florida
| | - Devan Patel
- College of Medicine, Florida State University, Tallahassee, Florida
| | - Chad Jones
- Department of Neurosurgery, University of Florida, Gainesville, Florida
| | - Kartik Motwani
- Department of Neurosurgery, University of Florida, Gainesville, Florida
| | - Hanain Z Fazal
- Department of Neurosurgery, University of Florida, Gainesville, Florida
| | - Koji Hosaka
- Department of Neurosurgery, University of Florida, Gainesville, Florida
| | - Brian L Hoh
- Department of Neurosurgery, University of Florida, Gainesville, Florida
| |
Collapse
|
12
|
Gupta M, Ha K, Agarwal R, Quarles LD, Smith JC. Molecular dynamics analysis of the binding of human interleukin-6 with interleukin-6 α-receptor. Proteins 2020; 89:163-173. [PMID: 32881084 DOI: 10.1002/prot.26002] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 07/26/2020] [Accepted: 08/25/2020] [Indexed: 11/07/2022]
Abstract
Human interleukin-6 (hIL-6) is a multifunctional cytokine that regulates immune and inflammatory responses in addition to metabolic and regenerative processes and cancer. hIL-6 binding to the IL-6 receptor (IL-6Rα) induces homodimerization and recruitment of the glycoprotein (gp130) to form a hexameric signaling complex. Anti-IL-6 and IL-6R antibodies are clinically approved inhibitors of IL-6 signaling pathway for treating rheumatoid arthritis and Castleman's disease, respectively. There is a potential to develop novel small molecule IL-6 antagonists derived from understanding the structural basis for IL-6/IL-6Rα interactions. Here, we combine homology modeling with extensive molecular dynamics (MD) simulations to examine the association of hIL-6 with IL-6Rα. A comparison with MD of apo hIL-6 reveals that the binding of hIL-6 to IL-6Rα induces structural and dynamic rearrangements in the AB loop region of hIL-6, disrupting intraprotein contacts and increasing the flexibility of residues 48 to 58 of the AB loop. In contrast, due to the involvement of residues 59 to 78 in forming contacts with the receptor, these residues of the AB loop are observed to rigidify in the presence of the receptor. The binary complex is primarily stabilized by two pairs of salt bridges, Arg181 (hIL-6)- Glu182 (IL-6Rα) and Arg184 (hIL-6)- Glu183 (IL-6Rα) as well as hydrophobic and aromatic stacking interactions mediated essentially by Phe residues in both proteins. An interplay of electrostatic, hydrophobic, hydrogen bonding, and aromatic stacking interactions facilitates the formation of the hIL-6/IL-6Rα complex.
Collapse
Affiliation(s)
- Madhulika Gupta
- UT/ORNL Center for Molecular Biophysics, Oak Ridge National Lab, Oak Ridge, Tennessee, USA
| | - Khanh Ha
- Tickle College of Engineering, University of Tennessee, Knoxville, Tennessee, USA
| | - Rupesh Agarwal
- UT/ORNL Center for Molecular Biophysics, Oak Ridge National Lab, Oak Ridge, Tennessee, USA
| | - Leigh Darryl Quarles
- Division of Nephrology, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Jeremy C Smith
- UT/ORNL Center for Molecular Biophysics, Oak Ridge National Lab, Oak Ridge, Tennessee, USA
| |
Collapse
|
13
|
Siersbæk R, Scabia V, Nagarajan S, Chernukhin I, Papachristou EK, Broome R, Johnston SJ, Joosten SEP, Green AR, Kumar S, Jones J, Omarjee S, Alvarez-Fernandez R, Glont S, Aitken SJ, Kishore K, Cheeseman D, Rakha EA, D'Santos C, Zwart W, Russell A, Brisken C, Carroll JS. IL6/STAT3 Signaling Hijacks Estrogen Receptor α Enhancers to Drive Breast Cancer Metastasis. Cancer Cell 2020; 38:412-423.e9. [PMID: 32679107 PMCID: PMC7116707 DOI: 10.1016/j.ccell.2020.06.007] [Citation(s) in RCA: 140] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 03/20/2020] [Accepted: 06/12/2020] [Indexed: 02/06/2023]
Abstract
The cytokine interleukin-6 (IL6) and its downstream effector STAT3 constitute a key oncogenic pathway, which has been thought to be functionally connected to estrogen receptor α (ER) in breast cancer. We demonstrate that IL6/STAT3 signaling drives metastasis in ER+ breast cancer independent of ER. STAT3 hijacks a subset of ER enhancers to drive a distinct transcriptional program. Although these enhancers are shared by both STAT3 and ER, IL6/STAT3 activity is refractory to standard ER-targeted therapies. Instead, inhibition of STAT3 activity using the JAK inhibitor ruxolitinib decreases breast cancer invasion in vivo. Therefore, IL6/STAT3 and ER oncogenic pathways are functionally decoupled, highlighting the potential of IL6/STAT3-targeted therapies in ER+ breast cancer.
Collapse
Affiliation(s)
- Rasmus Siersbæk
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge CB2 0RE, UK.
| | - Valentina Scabia
- ISREC - Swiss Institute for Experimental Cancer Research, School of Life Sciences, Ecole Polytechnique fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | - Sankari Nagarajan
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge CB2 0RE, UK
| | - Igor Chernukhin
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge CB2 0RE, UK
| | | | - Rebecca Broome
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge CB2 0RE, UK
| | - Simon J Johnston
- Nottingham Breast Cancer Research Centre, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham Biodiscovery Institute, University Park, Nottingham NG7 2RD, UK
| | - Stacey E P Joosten
- Division of Oncogenomics, Oncode Institute, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Andrew R Green
- Nottingham Breast Cancer Research Centre, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham Biodiscovery Institute, University Park, Nottingham NG7 2RD, UK
| | - Sanjeev Kumar
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge CB2 0RE, UK; Addenbrookes Hospital, Cambridge CB2 0QQ, UK
| | - Julia Jones
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge CB2 0RE, UK
| | - Soleilmane Omarjee
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge CB2 0RE, UK
| | | | - Silvia Glont
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge CB2 0RE, UK
| | - Sarah J Aitken
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge CB2 0RE, UK; Department of Histopathology, Cambridge University Hospitals NHS Foundation Trust, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK; Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK
| | - Kamal Kishore
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge CB2 0RE, UK
| | - Danya Cheeseman
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge CB2 0RE, UK
| | - Emad A Rakha
- Nottingham Breast Cancer Research Centre, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham Biodiscovery Institute, University Park, Nottingham NG7 2RD, UK
| | - Clive D'Santos
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge CB2 0RE, UK
| | - Wilbert Zwart
- Division of Oncogenomics, Oncode Institute, Netherlands Cancer Institute, Amsterdam, the Netherlands; Laboratory of Chemical Biology and Institute for Complex Molecular Systems, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Alasdair Russell
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge CB2 0RE, UK
| | - Cathrin Brisken
- ISREC - Swiss Institute for Experimental Cancer Research, School of Life Sciences, Ecole Polytechnique fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | - Jason S Carroll
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge CB2 0RE, UK.
| |
Collapse
|
14
|
Alahdal M, Duan L, Ouyang H, Wang D. The role of indoleamine 2,3 dioxygenase 1 in the osteoarthritis. Am J Transl Res 2020; 12:2322-2343. [PMID: 32655775 PMCID: PMC7344072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 05/05/2020] [Indexed: 06/11/2023]
Abstract
Osteoarthritis (OA) is a chronic degenerative joint disease and a leading cause of disability. It involves articular cartilage destruction and a whole joint inflammation. In spite of OA pathogenesis is still unclear, new studies on the OA pathophysiological aetiology and immunomodulation therapy continuously achieve significant advances with new concepts. Here, we focus on the indoleamine-2,3-dioxygenase1 (IDO1) activity in the osteoarthritis (OA), which is one of the noticeable enzymes in the synovial fluid of arthritis patients. It was recognized as an essential mediator of autoreactive B and T cell responses in rheumatoid arthritis (RA) and an interesting therapeutic target against RA. However, the role IDO1 plays in the OA pathogenesis hasn't been discussed. The new OA experimental analysis evidenced IDO1 overexpression in the synovial fluid of OA patients, and recent studies reported that IDO1 metabolites were found higher in the OA synovial fluid than RA and spondyloarthropathies (SpA) patients. Moreover, the positive relation of IDO1 metabolites with OA pain and joint stiffness has been confirmed. Thus, the IDO1 plays a pivotal role in the pathogenesis of OA. In this review, the role IDO1 plays in the OA pathogenesis has been deeply discussed. It could be a promising target in the immunotherapy of OA disease.
Collapse
Affiliation(s)
- Murad Alahdal
- Shenzhen Key Laboratory of Tissue Engineering, Shenzhen Laboratory of Digital Orthopedic Engineering, Shenzhen Second People’s Hospital (The First Hospital Affiliated to Shenzhen University, Health Science Center)Shenzhen 518035, P. R. China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of MedicineHangzhou, P. R. China
- Guangdong Provincial Research Center for Artificial Intelligence and Digital Orthopedic TechnologyShenzhen 518035, P. R. China
| | - Li Duan
- Shenzhen Key Laboratory of Tissue Engineering, Shenzhen Laboratory of Digital Orthopedic Engineering, Shenzhen Second People’s Hospital (The First Hospital Affiliated to Shenzhen University, Health Science Center)Shenzhen 518035, P. R. China
- Guangdong Provincial Research Center for Artificial Intelligence and Digital Orthopedic TechnologyShenzhen 518035, P. R. China
| | - Hongwei Ouyang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of MedicineHangzhou, P. R. China
| | - Daping Wang
- Shenzhen Key Laboratory of Tissue Engineering, Shenzhen Laboratory of Digital Orthopedic Engineering, Shenzhen Second People’s Hospital (The First Hospital Affiliated to Shenzhen University, Health Science Center)Shenzhen 518035, P. R. China
- Guangdong Provincial Research Center for Artificial Intelligence and Digital Orthopedic TechnologyShenzhen 518035, P. R. China
| |
Collapse
|
15
|
Wei Q, Chen X, Chen H. Association of Single Nucleotide Polymorphisms of the IL-6, IL-10, and TNF-α Genes with Susceptibility to Gestational Diabetes Mellitus. Genet Test Mol Biomarkers 2020; 24:390-398. [PMID: 32513030 DOI: 10.1089/gtmb.2020.0069] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Objective: To investigate any associations between the single nucleotide polymorphisms (SNPs) at the interleukin-6 (IL-6) rs1800796, interleukin-10 (IL-10) rs1800896, and the tumor necrosis factor-alpha (TNF-α) rs1800629 loci with gestational diabetes mellitus (GDM) susceptibility. Materials and Methods: A total of 242 GDM patients and 242 healthy controls were enrolled in this study. The genotypes of the IL-6 rs1800796, IL-10 rs1800896, and TNF-α rs1800629 loci were analyzed by Sanger sequencing. Interactions among these SNPs were analyzed through multifactor dimensionality reduction. Results: Women with the IL-6 rs1800796 G allele had a higher GDM susceptibility risk than those with the C allele (odds ratio [OR] = 1.44, 95% confidence interval [CI]: 1.10-1.90, p = 0.010). Women with the IL-10 rs1800896 C allele had a higher risk of GDM susceptibility than those with the T allele (OR = 2.94, 95% CI: 1.87-4.63, p < 0.001). Women with the TNF-α rs1800629 A allele had a higher risk of susceptibility to GDM than those with the G allele (OR = 3.73, 95% CI: 2.25-6.18, p < 0.001). The plasma levels of IL-6 and TNF-α in GDM patients were significantly higher than those in the control group, and the levels of IL-10 were significantly lower than those in the control group (p < 0.001). Women with the IL-6 rs1800796 CG/GG genotypes had higher plasma IL-6 levels than those with the CC genotype (p < 0.05). Women with the IL-10 rs1800896 TT genotype had higher IL-10 levels than those with the TC/CC genotypes (p < 0.05), and those with the TNF-α rs1800629 GA/AA genotypes had higher TNF-α levels than those with the GG genotype (p < 0.05). Conclusion: The results of this study show that the IL-6 rs1800796 G allele, the IL-10 rs1800896 C allele, and the TNF-α rs1800629 A allele are significantly associated with an increased risk of susceptibility to GDM.
Collapse
Affiliation(s)
- Qing Wei
- Department of Women's Health, Hangzhou Fuyang Women's and Children's Hospital, Hangzhou, China
| | - Xufeng Chen
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Heng Chen
- Department of Clinical Laboratory, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
16
|
Kulkarni P, Martson A, Vidya R, Chitnavis S, Harsulkar A. Pathophysiological landscape of osteoarthritis. Adv Clin Chem 2020; 100:37-90. [PMID: 33453867 DOI: 10.1016/bs.acc.2020.04.002] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
A sharp rise in osteoarthritis (OA) incidence is expected as over 25% of world population ages in the coming decade. Although OA is considered a degenerative disease, mounting evidence suggests a strong connection with chronic metabolic conditions and low-grade inflammation. OA pathology is increasingly understood as a complex interplay of multiple pathological events including oxidative stress, synovitis and immune responses revealing its intricate nature. Cellular, biochemical and molecular aspects of these pathological events along with major outcomes of the relevant research studies in this area are discussed in the present review. With reference to their published and unpublished work, the authors strongly propose synovitis as a central OA pathology and the key OA pathological events are described in connection with it. Recent research outcomes also have succeeded to establish a linkage between metabolic syndrome and OA, which has been precisely included in the present review. Impact of aging process cannot be neglected in OA. Cell senescence is an important mechanism of aging through which it facilitates development of OA like other degenerative disorders, also discussed within a frame of OA. Conclusively, the reviewers urge low-grade inflammation linked to aging and derailed immune function as a pathological platform for OA development and progression. Thus, interventions targeted to prevent inflammaging hold a promising potential in effective OA management and efforts should be invested in this direction.
Collapse
Affiliation(s)
- Priya Kulkarni
- Department of Pathophysiology, Biomedicine and Translational medicine, University of Tartu, Tartu, Estonia; Department of Traumatology and Orthopaedics, Tartu University Hospital, Tartu, Estonia
| | - Aare Martson
- Department of Traumatology and Orthopaedics, Tartu University Hospital, Tartu, Estonia; Clinic of Traumatology and Orthopaedics, Tartu University Hospital, Tartu, Estonia
| | - Ragini Vidya
- Department of Pharmaceutical Biotechnology, Poona College of Pharmacy, Pune, India
| | - Shreya Chitnavis
- Department of Pharmaceutical Biotechnology, Poona College of Pharmacy, Pune, India
| | - Abhay Harsulkar
- Department of Pathophysiology, Biomedicine and Translational medicine, University of Tartu, Tartu, Estonia; Department of Pharmaceutical Biotechnology, Poona College of Pharmacy, Pune, India.
| |
Collapse
|
17
|
Nieto-Nicolau N, Martínez-Conesa EM, Velasco-García AM, Aloy-Reverté C, Vilarrodona A, Casaroli-Marano RP. Xenofree generation of limbal stem cells for ocular surface advanced cell therapy. Stem Cell Res Ther 2019; 10:374. [PMID: 31801638 PMCID: PMC6894225 DOI: 10.1186/s13287-019-1501-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 11/09/2019] [Accepted: 11/20/2019] [Indexed: 12/13/2022] Open
Abstract
Background Limbal stem cells (LSC) sustain the corneal integrity and homeostasis. LSC deficiency (LSCD) leads to loss of corneal transparency and blindness. A clinical approach to treat unilateral LSCD comprises autologous cultured limbal epithelial stem cell transplantation (CLET). CLET uses xenobiotic culture systems with potential zoonotic transmission risks, and regulatory guidelines make necessary to find xenofree alternatives. Methods We compared two xenofree clinical grade media and two feeder layers. We used CnT07, a defined commercial medium for keratinocytes, and a modified xenofree supplemented hormonal epithelial medium with human serum (XSHEM). Optimal formulation was used to compare two feeder layers: the gold standard 3T3 murine fibroblasts and human processed lipoaspirate cells (PLA). We tested the expressions of ΔNp63α and cytokeratin 3 and 12 by qPCR and immunofluorescence. Morphology, viability, clonogenicity, proliferation, and cell growth assays were carried out. We also evaluated interleukin 6 (IL-6) and stromal-derived factor 1 (SDF-1) by qPCR and ELISA. Results XSHEM maintained better LSC culture viability and morphology than CnT07. Irradiated PLA feeder cells improved the undifferentiated state of LSC and enhanced their growth and clonogenicity stimulating IL-6 secretion and SDF-1 expression, as well as increased proliferation and cell growth when compared with irradiated 3T3 feeder cells. Conclusions The combination of XSHEM and PLA feeder cells efficiently sustained LSC xenofree cultures for clinical application. Moreover, PLA feeder layers were able to improve the LSC potential characteristics. Our results would have direct clinical application in CLET for advanced therapy. Graphical abstract ![]()
Collapse
Affiliation(s)
- Nuria Nieto-Nicolau
- Barcelona Tissue Bank, Banc de Sang i Teixits (BST), Barcelona, Spain.,Institute of Biomedical Research (IIB-Sant Pau; SGR1113), Barcelona, Spain
| | - Eva M Martínez-Conesa
- Barcelona Tissue Bank, Banc de Sang i Teixits (BST), Barcelona, Spain.,Institute of Biomedical Research (IIB-Sant Pau; SGR1113), Barcelona, Spain
| | - Alba M Velasco-García
- Department of Surgery, School of Medicine & Hospital Clinic de Barcelona, University of Barcelona, Barcelona, Spain
| | - Caterina Aloy-Reverté
- Barcelona Tissue Bank, Banc de Sang i Teixits (BST), Barcelona, Spain.,Institute of Biomedical Research (IIB-Sant Pau; SGR1113), Barcelona, Spain
| | - Anna Vilarrodona
- Barcelona Tissue Bank, Banc de Sang i Teixits (BST), Barcelona, Spain.,Institute of Biomedical Research (IIB-Sant Pau; SGR1113), Barcelona, Spain
| | - Ricardo P Casaroli-Marano
- Barcelona Tissue Bank, Banc de Sang i Teixits (BST), Barcelona, Spain. .,Institute of Biomedical Research (IIB-Sant Pau; SGR1113), Barcelona, Spain. .,Department of Surgery, School of Medicine & Hospital Clinic de Barcelona, University of Barcelona, Barcelona, Spain.
| |
Collapse
|
18
|
Verboogen DRJ, Revelo NH, ter Beest M, van den Bogaart G. Interleukin-6 secretion is limited by self-signaling in endosomes. J Mol Cell Biol 2019; 11:144-157. [PMID: 30016456 PMCID: PMC6392102 DOI: 10.1093/jmcb/mjy038] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 05/04/2018] [Accepted: 07/12/2018] [Indexed: 12/13/2022] Open
Abstract
Cells producing cytokines often express the receptor for the same cytokine, which makes them prone to autocrine signaling. How cytokine release and signaling are regulated in the same cell is not understood. In this study, we demonstrate that signaling by exogenous and self-synthesized inflammatory cytokine interleukin-6 (IL-6) within endosomal compartments acts as a cellular brake that limits the synthesis of IL-6. Our data show that IL-6 is internalized by dendritic cells and signals from endosomal compartments containing the IL-6 receptor. Newly synthesized IL-6 also traffics via these endosomal compartments and signals in transit to the plasma membrane. This allows activation of STAT3 which in turn limits toll-like receptor 4 stimulant lipopolysaccharide (LPS) triggered transcription of IL-6. Long-term exposure to LPS removes this brake via inhibition of STAT3 by increased expression of suppressor of cytokine signaling 3 and results in fully fledged IL-6 production. This transient regulation could prevent excessive IL-6 production during early infections.
Collapse
Affiliation(s)
- Daniëlle R J Verboogen
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Natalia H Revelo
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Martin ter Beest
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Geert van den Bogaart
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Molecular Immunology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Nijenborgh 7, Groningen, The Netherlands
| |
Collapse
|
19
|
Stromal-derived interleukin 6 drives epithelial-to-mesenchymal transition and therapy resistance in esophageal adenocarcinoma. Proc Natl Acad Sci U S A 2019; 116:2237-2242. [PMID: 30670657 PMCID: PMC6369811 DOI: 10.1073/pnas.1820459116] [Citation(s) in RCA: 116] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Esophageal adenocarcinoma (EAC) has a dismal prognosis, and survival benefits of recent multimodality treatments remain small. Cancer-associated fibroblasts (CAFs) are known to contribute to poor outcome by conferring therapy resistance to various cancer types, but this has not been explored in EAC. Importantly, a targeted strategy to circumvent CAF-induced resistance has yet to be identified. By using EAC patient-derived CAFs, organoid cultures, and xenograft models we identified IL-6 as the stromal driver of therapy resistance in EAC. IL-6 activated epithelial-to-mesenchymal transition in cancer cells, which was accompanied by enhanced treatment resistance, migratory capacity, and clonogenicity. Inhibition of IL-6 restored drug sensitivity in patient-derived organoid cultures and cell lines. Analysis of patient gene expression profiles identified ADAM12 as a noninflammation-related serum-borne marker for IL-6-producing CAFs, and serum levels of this marker predicted unfavorable responses to neoadjuvant chemoradiation in EAC patients. These results demonstrate a stromal contribution to therapy resistance in EAC. This signaling can be targeted to resensitize EAC to therapy, and its activity can be measured using serum-borne markers.
Collapse
|
20
|
In Vitro Th17-Polarized Human CD4 + T Cells Exacerbate Xenogeneic Graft-versus-Host Disease. Biol Blood Marrow Transplant 2018; 25:204-215. [PMID: 30326279 DOI: 10.1016/j.bbmt.2018.10.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2018] [Accepted: 10/08/2018] [Indexed: 12/14/2022]
Abstract
Acute graft-versus-host disease (aGVHD) is a severe complication of allogeneic hematopoietic stem cell transplantation. The role of Th17 cells in its pathophysiology remains a matter of debate. In this study, we assessed whether enrichment of human peripheral blood mononuclear cells (PBMCs) with in vitro Th17-polarized CD4+ T cells would exacerbate xenogeneic GVHD (xGVHD) into NOD-scid IL-2Rγ null (NSG) mice. Naive human CD4+ T cells were stimulated under Th17-skewing conditions for 8 to 10 days and then coinjected in NSG mice with fresh PBMCs from the same donor. We observed that Th17-polarized cells engrafted and migrated toward xGVHD target organs. They also acquired a double-expressing IL-17A+IFNγ+ profile in vivo. Importantly, cotransfer of Th17-polarized cells (1 × 106) with PBMCs (1 × 106) exacerbated xGVHD compared with transplantation of PBMCs alone (2 × 106). Furthermore, PBMC cotransfer with Th17-polarized cells was more potent for xGVHD induction than cotransfer with naive CD4+ T cells stimulated in nonpolarizing conditions (Th0 cells, 1 × 106 + 1 × 106 PBMCs) or with Th1-polarized cells (1 × 106 + 1 × 106 PBMCs). In summary, our results suggest that human Th17-polarized cells can cooperate with PBMCs and be pathogenic in the NSG xGVHD model.
Collapse
|
21
|
Pich C, Meylan P, Mastelic-Gavillet B, Nguyen TN, Loyon R, Trang BK, Moser H, Moret C, Goepfert C, Hafner J, Levesque MP, Romero P, Jandus C, Michalik L. Induction of Paracrine Signaling in Metastatic Melanoma Cells by PPARγ Agonist Rosiglitazone Activates Stromal Cells and Enhances Tumor Growth. Cancer Res 2018; 78:6447-6461. [PMID: 30185551 DOI: 10.1158/0008-5472.can-18-0912] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 07/27/2018] [Accepted: 08/28/2018] [Indexed: 01/10/2023]
Abstract
In addition to improving insulin sensitivity in type 2 diabetes, the thiazolidinedione family of compounds and the pharmacologic activation of their best-characterized target PPARγ have been proposed as a therapeutic option for cancer treatment. In this study, we reveal a new mode of action for the thiazolidinedione rosiglitazone that can contribute to tumorigenesis. Rosiglitazone activated a tumorigenic paracrine communication program in a subset of human melanoma cells that involves the secretion of cytokines, chemokines, and angiogenic factors. This complex blend of paracrine signals activated nonmalignant fibroblasts, endothelial cells, and macrophages in a tumor-friendly way. In agreement with these data, rosiglitazone promoted human melanoma development in xenografts, and tumors exposed to rosiglitazone exhibited enhanced angiogenesis and inflammation. Together, these findings establish an important tumorigenic action of rosiglitazone in a subset of melanoma cells. Although studies conducted on cohorts of diabetic patients report overall benefits of thiazolidinediones in cancer prevention, our data suggest that exposure of established tumors to rosiglitazone may be deleterious.Significance: These findings uncover a novel mechanism by which the thiazolidinedione compound rosiglitazone contributes to tumorigenesis, thus highlighting a potential risk associated with its use in patients with established tumors. Cancer Res; 78(22); 6447-61. ©2018 AACR.
Collapse
Affiliation(s)
- Christine Pich
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Patrick Meylan
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Beatris Mastelic-Gavillet
- Department of Oncology, University of Lausanne, Ludwig Cancer Research Center, Lausanne, Switzerland
| | - Thanh Nhan Nguyen
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Romain Loyon
- Department of Oncology, University of Lausanne, Ludwig Cancer Research Center, Lausanne, Switzerland
| | - Bao Khanh Trang
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Hélène Moser
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Catherine Moret
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Christine Goepfert
- COMPATH, Institute of Animal Pathology, University of Bern, Bern, Switzerland
| | - Jürg Hafner
- Department of Dermatology, University Hospital Zürich, University of Zürich, Zürich, Switzerland
| | - Mitchell P Levesque
- Department of Dermatology, University Hospital Zürich, University of Zürich, Zürich, Switzerland
| | - Pedro Romero
- Department of Oncology, University of Lausanne, Ludwig Cancer Research Center, Lausanne, Switzerland
| | - Camilla Jandus
- Department of Oncology, University of Lausanne, Ludwig Cancer Research Center, Lausanne, Switzerland
| | - Liliane Michalik
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
22
|
Wojdasiewicz P, Poniatowski ŁA, Nauman P, Mandat T, Paradowska-Gorycka A, Romanowska-Próchnicka K, Szukiewicz D, Kotela A, Kubaszewski Ł, Kotela I, Kurkowska-Jastrzębska I, Gasik R. Cytokines in the pathogenesis of hemophilic arthropathy. Cytokine Growth Factor Rev 2018; 39:71-91. [DOI: 10.1016/j.cytogfr.2017.11.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2017] [Accepted: 11/09/2017] [Indexed: 01/26/2023]
|
23
|
Paula FMM, Leite NC, Borck PC, Freitas-Dias R, Cnop M, Chacon-Mikahil MPT, Cavaglieri CR, Marchetti P, Boschero AC, Zoppi CC, Eizirik DL. Exercise training protects human and rodent β cells against endoplasmic reticulum stress and apoptosis. FASEB J 2018; 32:1524-1536. [PMID: 29133342 DOI: 10.1096/fj.201700710r] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Prolonged exercise has positive metabolic effects in obese or diabetic individuals. These effects are usually ascribed to improvements in insulin sensitivity. We evaluated whether exercise also generates circulating signals that protect human and rodent β cells against endoplasmic reticulum (ER) stress and apoptosis. For this purpose, we obtained serum from humans or mice before and after an 8 wk training period. Exposure of human islets or mouse or rat β cells to human or rodent sera, respectively, obtained from trained individuals reduced cytokine (IL-1β+IFN-γ)- or chemical ER stressor-induced β-cell ER stress and apoptosis, at least in part via activation of the transcription factor STAT3. These findings indicate that exercise training improves human and rodent β-cell survival under diabetogenic conditions and support lifestyle interventions as a protective approach for both type 1 and 2 diabetes.-Paula, F. M. M., Leite, N. C., Borck, P. C., Freitas-Dias, R., Cnop, M., Chacon-Mikahil, M. P. T., Cavaglieri, C. R., Marchetti, P., Boschero, A. C., Zoppi, C. C., Eizirik, D. L. Exercise training protects human and rodent β cells against endoplasmic reticulum stress and apoptosis.
Collapse
Affiliation(s)
- Flavia M M Paula
- Center for Diabetes Research, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Nayara C Leite
- Department of Structural and Functional Biology, Institute of Biology, Obesity and Comorbidities Research Center, University of Campinas (UNICAMP), Campinas, Brazil
| | - Patricia C Borck
- Department of Structural and Functional Biology, Institute of Biology, Obesity and Comorbidities Research Center, University of Campinas (UNICAMP), Campinas, Brazil
| | - Ricardo Freitas-Dias
- Department of Structural and Functional Biology, Institute of Biology, Obesity and Comorbidities Research Center, University of Campinas (UNICAMP), Campinas, Brazil.,Department of Physical Therapy, University of Pernambuco, Petrolina, Brazil
| | - Miriam Cnop
- Center for Diabetes Research, Université Libre de Bruxelles (ULB), Brussels, Belgium.,Division of Endocrinology, Erasmus Hospital, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Mara P T Chacon-Mikahil
- Exercise Physiology Laboratory (FISEX), Faculty of Physical Education, University of Campinas (UNICAMP), Campinas, Brazil; and
| | - Claudia R Cavaglieri
- Exercise Physiology Laboratory (FISEX), Faculty of Physical Education, University of Campinas (UNICAMP), Campinas, Brazil; and
| | - Piero Marchetti
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Antonio C Boschero
- Department of Structural and Functional Biology, Institute of Biology, Obesity and Comorbidities Research Center, University of Campinas (UNICAMP), Campinas, Brazil
| | - Claudio C Zoppi
- Department of Structural and Functional Biology, Institute of Biology, Obesity and Comorbidities Research Center, University of Campinas (UNICAMP), Campinas, Brazil
| | - Decio L Eizirik
- Center for Diabetes Research, Université Libre de Bruxelles (ULB), Brussels, Belgium
| |
Collapse
|
24
|
Kim YS, Lee KJ, Kim H. Serum tumour necrosis factor-α and interleukin-6 levels in Alzheimer's disease and mild cognitive impairment. Psychogeriatrics 2017; 17:224-230. [PMID: 28130814 DOI: 10.1111/psyg.12218] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2016] [Revised: 06/29/2016] [Accepted: 07/22/2016] [Indexed: 12/27/2022]
Abstract
BACKGROUND Neuroinflammation has been recognized as a feature of Alzheimer's disease (AD), and mild cognitive impairment (MCI) is believed to share several pathological features with AD. The aim of the present study was to compare serum cytokine levels between patients with AD, subjects with MCI, and healthy controls, and to assess the correlation between cytokine levels and cognitive performance in these subjects. METHODS Participants included 35 patients with AD, 29 subjects with MCI, and 28 healthy controls from the Department of Psychiatry of IIlsan Paik Hospital in South Korea. Demographic and neuropsychological information were obtained, and peripheral cytokine levels, specifically tumour necrosis factor (TNF)-α and interleukin (IL)-6 levels, were measured for all subjects. RESULTS After adjustment for age, a significant difference in IL-6 levels (P = 0.045), but not in TNF-α (P = 0.082) levels, was observed among the three groups. IL-6 levels were higher in patients with AD than in subjects with MCI and healthy controls. TNF-α and IL-6 levels negatively and positively correlated with Mini-Mental State Examination and Global Deterioration Scale scores, respectively. TNF-α and IL-6 levels were also positively correlated with each other. CONCLUSIONS The present study suggests that serum IL-6 levels of patients with AD might be higher than those of subjects with MCI and healthy controls. Serum TNF-α and IL-6 levels might be negatively correlated with cognitive function, and we suspect that serum IL-6 levels could be biomarkers for AD.
Collapse
Affiliation(s)
- Yo Sup Kim
- Department of Psychiatry, Ilsan Paik Hospital, Inje University College of Medicine, Goyang, Korea
| | - Kang Joon Lee
- Department of Psychiatry, Ilsan Paik Hospital, Inje University College of Medicine, Goyang, Korea
| | - Hyun Kim
- Department of Psychiatry, Ilsan Paik Hospital, Inje University College of Medicine, Goyang, Korea
| |
Collapse
|
25
|
Liu G, Zhang J, Frey L, Gang X, Wu K, Liu Q, Lilly M, Wu J. Prostate-specific IL-6 transgene autonomously induce prostate neoplasm through amplifying inflammation in the prostate and peri-prostatic adipose tissue. J Hematol Oncol 2017; 10:14. [PMID: 28077171 PMCID: PMC5225646 DOI: 10.1186/s13045-016-0386-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2016] [Accepted: 12/30/2016] [Indexed: 12/11/2022] Open
Abstract
Background The causative role of the pro-inflammatory cytokine IL-6 in prostate cancer progression has been well established at molecular level. However, whether and how IL-6 may play a role in prostate cancer risk and development is not well defined. One limitation factor to acquiring this knowledge is the lack of appropriate animal models. Methods We generated a novel line of prostate-specific IL-6 transgenic mouse model. We compared the prostate pathology, tumorigenic signaling components, and prostate tumor microenvironment of the IL-6 transgenic mice with wild type littermates. Results With this model, we demonstrate that IL-6 induces prostate neoplasm autonomously. We further demonstrate that transgenic expression of IL-6 in the prostate activates oncogenic pathways, induces autocrine IL-6 secretion and steadily-state of STAT3 activation in the prostate tissue, upregulates paracrine insulin-like growth factor (IGF) signaling axis, reprograms prostate oncogenic gene expression, and more intriguingly, amplifies inflammation in the prostate and peri-prostatic adipose tissue. Conclusions The pro-inflammatory IL-6 is autonomous oncogene for the prostate. IL-6 induces prostate oncogenesis through amplifying local inflammation. We also presented a valuable animal model to study inflammation and prostate cancer development. Electronic supplementary material The online version of this article (doi:10.1186/s13045-016-0386-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Gang Liu
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Jinyu Zhang
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Lewis Frey
- Public Health Science, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Xiao Gang
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, 29425, USA.,Present address: Department of Laboratory Medicine, The Third Hospital of South Medical University, Guangzhou, China
| | - Kongming Wu
- Department of Oncology, Tongji Medical College, Huazhong University of Science and Technology and Tongji Hospital, Wuhan, China
| | - Qian Liu
- Department of Oncology, Tongji Medical College, Huazhong University of Science and Technology and Tongji Hospital, Wuhan, China
| | - Michael Lilly
- Department of Hematology and Oncology, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Jennifer Wu
- Department of Medicine, University of Washington, Seattle, WA, USA. .,Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, 29425, USA. .,Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, 29425, USA.
| |
Collapse
|
26
|
Lee LL, Aung HH, Wilson DW, Anderson SE, Rutledge JC, Rutkowsky JM. Triglyceride-rich lipoprotein lipolysis products increase blood-brain barrier transfer coefficient and induce astrocyte lipid droplets and cell stress. Am J Physiol Cell Physiol 2017; 312:C500-C516. [PMID: 28077357 DOI: 10.1152/ajpcell.00120.2016] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Revised: 01/06/2017] [Accepted: 01/09/2017] [Indexed: 12/19/2022]
Abstract
Elevation of blood triglycerides, primarily as triglyceride-rich lipoproteins (TGRL), has been linked to cerebrovascular inflammation, vascular dementia, and Alzheimer's disease (AD). Brain microvascular endothelial cells and astrocytes, two cell components of the neurovascular unit, participate in controlling blood-brain barrier (BBB) permeability and regulating neurovascular unit homeostasis. Our studies showed that infusion of high physiological concentrations of TGRL lipolysis products (TGRL + lipoprotein lipase) activate and injure brain endothelial cells and transiently increase the BBB transfer coefficient (Ki = permeability × surface area/volume) in vivo. However, little is known about how blood lipids affect astrocyte lipid accumulation and inflammation. To address this, we first demonstrated TGRL lipolysis products increased lipid droplet formation in cultured normal human astrocytes. We then evaluated the transcriptional pathways activated in astrocytes by TGRL lipolysis products and found upregulated stress and inflammatory-related genes including activating transcription factor 3 (ATF3), macrophage inflammatory protein-3α (MIP-3α), growth differentiation factor-15 (GDF15), and prostaglandin-endoperoxide synthase 2 (COX2). TGRL lipolysis products also activated the JNK/cJUN/ATF3 pathway, induced endoplasmic reticulum stress protein C/EBP homologous protein (CHOP), and the NF-κB pathway, while increasing secretion of MIP-3α, GDF15, and IL-8. Thus our results demonstrate TGRL lipolysis products increase the BBB transfer coefficient (Ki), induce astrocyte lipid droplet formation, activate cell stress pathways, and induce secretion of inflammatory cytokines. Our observations are consistent with evidence for lipid-induced neurovascular injury and inflammation, and we, therefore, speculate that lipid-induced astrocyte injury could play a role in cognitive decline.
Collapse
Affiliation(s)
- Linda L Lee
- Department of Internal Medicine, University of California, Davis, California
| | - Hnin H Aung
- Department of Internal Medicine, University of California, Davis, California
| | - Dennis W Wilson
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California, Davis, California; and
| | - Steven E Anderson
- Department of Physiology and Membrane Biology, University of California, Davis, California
| | - John C Rutledge
- Department of Internal Medicine, University of California, Davis, California
| | | |
Collapse
|
27
|
Liao L, Chen Y, Wang W. The current progress in understanding the molecular functions and mechanisms of visfatin in osteoarthritis. J Bone Miner Metab 2016; 34:485-90. [PMID: 26969394 DOI: 10.1007/s00774-016-0743-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2015] [Accepted: 01/25/2016] [Indexed: 12/19/2022]
Abstract
Osteoarthritis, (OA), also known as degenerative arthritis or degenerative joint disease, is the most common form of arthritis, affecting millions of people worldwide. It is a group of mechanical abnormalities involving degradation of the joints and occurs when the protective cartilage (articular cartilage) on the ends of bones such as the knees, hips and fingers abrades over time. It mainly affects the whole joint structure, including the articular cartilage, subchondral bone and synovial tissue. Extensive work has been done in the past decades to investigate the cellular mechanism of this disease. However, to date, it is still poorly understood, and there is no effective treatment. Recently, both in vitro and in vivo studies have confirmed adipokines play critical roles during OA development. Among these, leptin and adiponectin have been well investigated, whereas the effect of the novel adipokine, visfatin, on OA still needs to be revealed. Therefore, in this short review, we will focus on visfatin and summarize the current progress in the research on its role in OA development.
Collapse
Affiliation(s)
- Lele Liao
- Department of Orthopedics, The Second Xiangya Hospital of Central South University, 86 Renmin Middle Rd, Yuhua, Changsha, Hunan, China
| | - Yiyue Chen
- Department of Orthopedics, The Second Xiangya Hospital of Central South University, 86 Renmin Middle Rd, Yuhua, Changsha, Hunan, China
| | - Wanchun Wang
- Department of Orthopedics, The Second Xiangya Hospital of Central South University, 86 Renmin Middle Rd, Yuhua, Changsha, Hunan, China.
| |
Collapse
|
28
|
Abstract
The human body combats infection and promotes wound healing through the remarkable process of inflammation. Inflammation is characterized by the recruitment of stromal cell activity including recruitment of immune cells and induction of angiogenesis. These cellular processes are regulated by a class of soluble molecules called cytokines. Based on function, cell target, and structure, cytokines are subdivided into several classes including: interleukins, chemokines, and lymphokines. While cytokines regulate normal physiological processes, chronic deregulation of cytokine expression and activity contributes to cancer in many ways. Gene polymorphisms of all types of cytokines are associated with risk of disease development. Deregulation RNA and protein expression of interleukins, chemokines, and lymphokines have been detected in many solid tumors and hematopoetic malignancies, correlating with poor patient prognosis. The current body of literature suggests that in some tumor types, interleukins and chemokines work against the human body by signaling to cancer cells and remodeling the local microenvironment to support the growth, survival, and invasion of primary tumors and enhance metastatic colonization. Some lymphokines are downregulated to suppress tumor progression by enhancing cytotoxic T cell activity and inhibiting tumor cell survival. In this review, we will describe the structure/function of several cytokine families and review our current understanding on the roles and mechanisms of cytokines in tumor progression. In addition, we will also discuss strategies for exploiting the expression and activity of cytokines in therapeutic intervention.
Collapse
Affiliation(s)
- M Yao
- University of Kansas Medical Center, Kansas City, KS, United States
| | - G Brummer
- University of Kansas Medical Center, Kansas City, KS, United States
| | - D Acevedo
- University of Kansas Medical Center, Kansas City, KS, United States
| | - N Cheng
- University of Kansas Medical Center, Kansas City, KS, United States.
| |
Collapse
|
29
|
Gong X, Jiang J, Zhang M. Exercise preconditioning reduces neonatal incision surgery-induced enhanced hyperalgesia via inhibition of P38 mitogen-activated protein kinase and IL-1β, TNF-α release. Int J Dev Neurosci 2016; 52:46-54. [PMID: 27235543 DOI: 10.1016/j.ijdevneu.2016.05.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2016] [Revised: 05/03/2016] [Accepted: 05/20/2016] [Indexed: 01/05/2023] Open
Abstract
Neonatal surgery leads to enhanced hyperalgesia to noxious stimulation in adulthood via a mechanism caused by enhanced phosphorylated (p)-p38 expression in microglia. We tested the effect of exercise on reducing enhanced hypersensitivity primed by neonatal incision surgery. Adult female Wistar rats, with or without neonatal incision surgery at postnatal day (P) 3, received right hind paw plantar incision surgery under anesthesia at P44. The rats performed wheel-running exercise from P22 to P41. Paw withdrawal threshold (PWT) and paw withdrawal latency (PWL) were measured and ipsilateral spinal cords were collected for protein quantification. For PWT and PWL, exercise reduced the pain index after incision surgery at P44 in rats with neonatal surgery (P<0.01). Western blots showed that exercise suppressed P-p38 expression relative to adult rats without neonatal surgery (P<0.05). Results of ELISA showed that exercise reduced IL-1β and TNF-α (P<0.05) concentration in the ipsilateral spinal cord. Exercise preconditioning is an effective approach to reducing enhanced adult hyperalgesia primed by neonatal surgery. The mechanism may be explained by exercise-induced inhibition of P-p38 activation and IL-1β, TNF-α release.
Collapse
Affiliation(s)
- Xingrui Gong
- Department of Anesthesiology & Pediatric Clinical Pharmacology Laboratory, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing Jiang
- Department of Anesthesiology & Pediatric Clinical Pharmacology Laboratory, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mazhong Zhang
- Department of Anesthesiology & Pediatric Clinical Pharmacology Laboratory, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
30
|
Crosstalk between bone marrow-derived myofibroblasts and gastric cancer cells regulates cancer stemness and promotes tumorigenesis. Oncogene 2016; 35:5388-5399. [PMID: 27109105 PMCID: PMC5063653 DOI: 10.1038/onc.2016.76] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 02/08/2016] [Accepted: 02/23/2016] [Indexed: 12/14/2022]
Abstract
Bone marrow-derived cells have important roles in cancer development and progression. Our previous studies demonstrated that murine bone marrow-derived myofibroblasts (BMFs) enhanced tumor growth. In this study, we investigated the mechanisms of BMF actions. We found that co-injection of BMFs with gastric cancer cells markedly promoted tumorigenesis. Co-cultured BMFs or BMF-conditioned medium (BMF-CM) induced the formation of spheres, which expressed stem cell signatures and exhibited features of self-renewal, epithelial-to-mesenchymal transition and tumor initiation. Furthermore, CD44+ fractions in spheres were able to initiate tumorigenesis and re-establish tumors in serially passaged xenografts. In co-culture systems, BMFs secreted high levels of murine interleukin-6 (IL-6) and hepatocyte growth factor (HGF), whereas cancer cells produced high level of transformation growth factor-β1 (TGF-β1). BMF-CM and IL-6 activated BMFs to produce mHGF, which activated signal transducer and activator of transcription 3 (STAT3) and upregulated TGF-β1 in human cancer cells. In return, cancer cell-CM stimulated BMFs to produce IL-6, which was inhibited by anti-TGF-β1 neutralizing antibody. Blockade of HGF/Met, Janus kinase 2 (JAK2)/STAT3 and TGF-β1 signaling by specific inhibitors inhibited BMF-induced sphere formation. STAT3 knockdown in cancer cells also inhibited BMF-induced sphere formation and tumorigenesis. Moreover, TGF-β1 overexpression in cancer cells was co-related with IL-6 and HGF overexpression in stromal cells in human gastric cancer tissues. Our results show that BMF-derived IL-6/HGF and cancer cell-derived TGF-β1 mediate the interactions between BMFs and gastric cancer cells, which regulate cancer stemness and promote tumorigenesis. Targeting inhibition of the interactions between BMFs and cancer cells may be a new strategy for cancer therapy.
Collapse
|
31
|
Noguchi-Sasaki M, Sasaki Y, Shimonaka Y, Mori K, Fujimoto-Ouchi K. Treatment with anti-IL-6 receptor antibody prevented increase in serum hepcidin levels and improved anemia in mice inoculated with IL-6-producing lung carcinoma cells. BMC Cancer 2016; 16:270. [PMID: 27068103 PMCID: PMC4828826 DOI: 10.1186/s12885-016-2305-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 04/07/2016] [Indexed: 12/21/2022] Open
Abstract
Background Hepcidin, a key regulator of iron metabolism, is produced mainly by interleukin-6 (IL-6) during inflammation. A mechanism linking cancer-related anemia and IL-6 through hepcidin production is suggested. To clarify the hypothesis that overproduction of IL-6 elevates hepcidin levels and contributes to the development of cancer-related anemia, we evaluated anti-IL-6 receptor antibody treatment of cancer-related anemia in an IL-6–producing human lung cancer xenograft model. Methods Nude mice were subcutaneously inoculated with cells of the IL-6–producing human lung cancer cell line LC-06-JCK and assessed as a model of cancer-related anemia. Mice bearing LC-06-JCK were administered rat anti-mouse IL-6 receptor antibody MR16-1 and their serum hepcidin levels and hematological parameters were determined. Results LC-06-JCK–bearing mice developed anemia according to the production of human IL-6 from xenografts, with decreased values of hemoglobin, hematocrit, and mean corpuscular volume (MCV) compared to non–tumor-bearing (NTB) mice. LC-06-JCK–bearing mice showed decreased body weight and serum albumin with increased serum amyloid A. MR16-1 treatment showed significant inhibition of decreased body weight and serum albumin levels, and suppressed serum amyloid A level. There was no difference in tumor volume between MR16-1-treated mice and immunoglobulin G (IgG)-treated control mice. Decreased hemoglobin, hematocrit, and MCV in LC-06-JCK–bearing mice was significantly relieved by MR16-1 treatment. LC-06-JCK–bearing mice showed high red blood cell counts and erythropoietin levels as compared to NTB mice, whereas MR16-1 treatment did not affect their levels. Serum hepcidin and ferritin levels were statistically elevated in mice bearing LC-06-JCK. LC-06-JCK–bearing mice showed lower values of MCV, mean corpuscular hemoglobin (MCH), and serum iron as compared to NTB mice. Administration of MR16-1 to mice bearing LC-06-JCK significantly suppressed levels of both serum hepcidin and ferritin, with increased values of MCV and MCH. Conclusions Our results suggest that overproduction of hepcidin by IL-6 signaling might be a major factor that leads to functionally iron-deficient cancer-related anemia in the LC-06-JCK model. We demonstrated that inhibition of the IL-6 signaling pathway by MR16-1 treatment resulted in significant recovery of iron-deficiency anemia and alleviation of cancer-related symptoms. These results indicate that IL-6 signaling might be one possible target pathway to treat cancer-related anemia disorders.
Collapse
Affiliation(s)
- Mariko Noguchi-Sasaki
- Product Research Department, Chugai Pharmaceutical Co., Ltd., 200 Kajiwara, Kamakura, Kanagawa, 247-8530, Japan.
| | - Yusuke Sasaki
- Product Research Department, Chugai Pharmaceutical Co., Ltd., 200 Kajiwara, Kamakura, Kanagawa, 247-8530, Japan
| | - Yasushi Shimonaka
- Product Research Department, Chugai Pharmaceutical Co., Ltd., 200 Kajiwara, Kamakura, Kanagawa, 247-8530, Japan
| | - Kazushige Mori
- Product Research Department, Chugai Pharmaceutical Co., Ltd., 200 Kajiwara, Kamakura, Kanagawa, 247-8530, Japan
| | - Kaori Fujimoto-Ouchi
- Product Research Department, Chugai Pharmaceutical Co., Ltd., 200 Kajiwara, Kamakura, Kanagawa, 247-8530, Japan
| |
Collapse
|
32
|
Herroon MK, Diedrich JD, Podgorski I. New 3D-Culture Approaches to Study Interactions of Bone Marrow Adipocytes with Metastatic Prostate Cancer Cells. Front Endocrinol (Lausanne) 2016; 7:84. [PMID: 27458427 PMCID: PMC4933721 DOI: 10.3389/fendo.2016.00084] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2016] [Accepted: 06/20/2016] [Indexed: 12/11/2022] Open
Abstract
Adipocytes are a major component of the bone marrow that can critically affect metastatic progression in bone. Understanding how the marrow fat cells influence growth, behavior, and survival of tumor cells requires utilization of in vitro cell systems that can closely mimic the physiological microenvironment. Herein, we present two new three-dimensional (3D) culture approaches to study adipocyte-tumor cell interactions in vitro. The first is a transwell-based system composed of the marrow-derived adipocytes in 3D collagen I gels and reconstituted basement membrane-overlayed prostate tumor cell spheroids. Tumor cells cultured under these 3D conditions are continuously exposed to adipocyte-derived factors, and their response can be evaluated by morphological and immunohistochemical analyses. We show via immunofluorescence analysis of metabolism-associated proteins that under 3D conditions tumor cells have significantly different metabolic response to adipocytes than tumor cells grown in 2D culture. We also demonstrate that this model allows for incorporation of other cell types, such as bone marrow macrophages, and utilization of dye-quenched collagen substrates for examination of proteolysis-driven responses to adipocyte- and macrophage-derived factors. Our second 3D culture system is designed to study tumor cell invasion toward the adipocytes and the consequent interaction between the two cell types. In this model, marrow adipocytes are separated from the fluorescently labeled tumor cells by a layer of collagen I. At designated time points, adipocytes are stained with BODIPY and confocal z-stacks are taken through the depth of the entire culture to determine the distance traveled between the two cell types over time. We demonstrate that this system can be utilized to study effects of candidate factors on tumor invasion toward the adipocytes. We also show that immunohistochemical analyses can be performed to evaluate the impact of direct interaction of prostate tumor cells with adipocytes. Our models underline the importance of using the appropriate culture conditions to mimic physiological interactions between marrow adipocytes and metastatic tumor cells. These systems have a potential to be utilized for analyses of various factors that may be regulated by the adipocytes in bone. Their application likely extends beyond metastatic prostate cancer to other tumors that colonize the bone marrow microenvironment.
Collapse
Affiliation(s)
| | - Jonathan Driscoll Diedrich
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI, USA
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA
| | - Izabela Podgorski
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI, USA
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA
- *Correspondence: Izabela Podgorski,
| |
Collapse
|
33
|
Kamperos G, Nikitakis N, Sfakianou A, Avgoustidis D, Sklavounou-Andrikopoulou A. Expression of NF-κB and IL-6 in oral precancerous and cancerous lesions: An immunohistochemical study. Med Oral Patol Oral Cir Bucal 2016; 21:e6-13. [PMID: 26595830 PMCID: PMC4765752 DOI: 10.4317/medoral.20570] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2014] [Accepted: 10/27/2015] [Indexed: 12/18/2022] Open
Abstract
Background The purpose of this study was to evaluate the immunohistochemical expression of NF-κB and IL-6 in oral premalignant and malignant lesions and to investigate their possible correlation with the presence of subepithelial inflammation. Material and Methods Thirty two oral premalignant lesions, clinically compatible with leukoplakia or erythroplakia, were investigated. Microscopically, 11 of them showed hyperkeratosis and acanthosis (epithelial hyperplasia) and 21 showed dysplasia of varying degrees. Nine cases of OSCC and four control cases of normal oral mucosa were also included in the study. Immunohistochemical staining with NF-κB (p65) and IL-6 was performed. IL-6 and nuclear NF-κB staining were assessed as positive or negative. For cytoplasmic localization of NF-κB, a total score combining intensity and percentage of positive epithelial cells was additionally calculated. The presence of inflammation was also recorded. Results Intensity and total scores for NF-κΒ cytoplasmic immunostaining showed a statistically significant gradual increase from normal mucosa to OSCC (p=0.012 and p=0.026 respectively). Non-statistically significant increased NF-κΒ nuclear localization was detected in dysplasias and OSCCs. Positive statistical correlation was detected between the presence of inflammation and IL-6 expression (p=0.015). No correlation between NF-κΒ and IL-6 was detected. Conclusions NF-κΒ is activated in the early stages of oral carcinogenesis. IL-6 may have an NF-κΒ-independent role, possibly through regulation of the inflammatory response. Key words:NF-κB, IL-6, immunohistochemistry, oral squamous cell carcinoma, oral precancerous lesion.
Collapse
|
34
|
Thao NP, Luyen BTT, Koo JE, Kim S, Koh YS, Thanh NV, Cuong NX, Kiem PV, Minh CV, Kim YH. In vitro anti-inflammatory components isolated from the carnivorous plant Nepenthes mirabilis (Lour.) Rafarin. PHARMACEUTICAL BIOLOGY 2015; 54:588-94. [PMID: 26186458 DOI: 10.3109/13880209.2015.1067234] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
CONTEXT Nepenthes mirabilis (Lour.) Rafarin (Nepenthaceae) is a carnivorous plant used as a folk medicine in the treatment of jaundice, hepatitis, gastric ulcers, ureteral stones, diarrhea, diabetes, and high blood pressure. Neither the phytochemical content nor biological activities of N. mirabilis have been reported. OBJECTIVE The anti-inflammatory activity from the N. mirabilis methanolic extract led to the isolation of compounds (1-26). MATERIALS AND METHODS Chromatographic methods were used to isolate compounds from the methanol extract of N. mirabilis branches and leaves. The anti-inflammatory activity of these isolated compounds was investigated in lipopolysaccharide (LPS)-stimulated bone marrow-derived dendritic cells (BMDCs) using ELISA. Primary BMDCs were used to examine the production of pro-inflammatory cytokines (IL-12 p40, IL-6, and TNF-α, at concentrations of 0.1, 0.2, and 1.0 μM) as compared with a positive control, SB203580 (1.0 μM). MTT assays showed that isolated compounds (1-26) did not exhibit significant cytotoxicity at concentrations up to 20.0 μM. RESULTS Compound 9 showed potent inhibition of IL-12 p40, IL-6, and TNF-α production (IC50 = 0.17 ± 0.02, 0.46 ± 0.01, and 8.28 ± 0.21 μM, respectively). Compound 4 showed potent inhibition of IL-12 p40 and IL-6 production (IC50 = 1.17 ± 0.01 and 2.15 ± 0.04 μM). In addition, IL-12 p40 inhibition by naphthalene derivatives (1-7, 9, and 10), phenolic compounds (11-15), lupeone (18), and flavonoids (22, 25, and 26) was more potent than with the positive control. The isolated compounds exhibited little and/or no inhibitory effects on TNF-α production in LPS-stimulated BMDCs. DISCUSSION AND CONCLUSION Taken together, these data suggest that the isolated components have significant inhibitory effects on pro-inflammatory cytokine production and warrant further study concerning their potential medicinal use.
Collapse
Affiliation(s)
- Nguyen Phuong Thao
- a College of Pharmacy, Chungnam National University , Daejeon , Republic of Korea
- b Institute of Marine Biochemistry (IMBC), Vietnam Academy of Science and Technology (VAST), Cau Giay , Hanoi , Vietnam , and
| | - Bui Thi Thuy Luyen
- a College of Pharmacy, Chungnam National University , Daejeon , Republic of Korea
| | - Jung Eun Koo
- c School of Medicine, Brain Korea 21 PLUS Program, Institute of Medical Science, Jeju National University , Jeju , Republic of Korea
| | - Sohyun Kim
- c School of Medicine, Brain Korea 21 PLUS Program, Institute of Medical Science, Jeju National University , Jeju , Republic of Korea
| | - Young Sang Koh
- c School of Medicine, Brain Korea 21 PLUS Program, Institute of Medical Science, Jeju National University , Jeju , Republic of Korea
| | - Nguyen Van Thanh
- b Institute of Marine Biochemistry (IMBC), Vietnam Academy of Science and Technology (VAST), Cau Giay , Hanoi , Vietnam , and
| | - Nguyen Xuan Cuong
- b Institute of Marine Biochemistry (IMBC), Vietnam Academy of Science and Technology (VAST), Cau Giay , Hanoi , Vietnam , and
| | - Phan Van Kiem
- b Institute of Marine Biochemistry (IMBC), Vietnam Academy of Science and Technology (VAST), Cau Giay , Hanoi , Vietnam , and
| | - Chau Van Minh
- b Institute of Marine Biochemistry (IMBC), Vietnam Academy of Science and Technology (VAST), Cau Giay , Hanoi , Vietnam , and
| | - Young Ho Kim
- a College of Pharmacy, Chungnam National University , Daejeon , Republic of Korea
| |
Collapse
|
35
|
Rao YQ, Li J, Wang WJ. Effects of Gengnianchun on learning and memory ability, neurotransmitter, cytokines, and leptin in ovariectomized rats. Int J Clin Exp Med 2015; 8:8648-8660. [PMID: 26309517 PMCID: PMC4538054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Accepted: 06/03/2015] [Indexed: 06/04/2023]
Abstract
This study aimed to evaluate the beneficial effects of a traditional Chinese medicine named Gengnianchun (GNC) in ovariectomized Sprague-Dawley rats. The rats were randomly categorized into sham-operated group (Sham), saline-treated ovariectomized group (OVX), GNC-treated ovariectomized group (OVX+GNC), estradiol valerate-treated ovariectomized group (OVX+E). GNC and estradiol was administered for 1 month at dosages of 125 and 0.1 mg/day, respectively. Ovariectomy caused deterioration of learning and memory ability (P < 0.05), which was restored by treatment with GNC and estradiol (P < 0.05). Estrogen level and endometrial thickness significantly decreased in the OVX group (P < 0.05). These parameters significantly increased in the OVX+E group (P < 0.05) but did not change in the OVX+GNC group (P > 0.05). GNC and estradiol significantly increased the levels of norepinephrine (NE) and dopamine (DA) and decreased the levels of 5-hydroxytryptamine (5-HT) and 5-hydroxyindoleacetic acid (5-HIAA) in the hypothalamus (P < 0.05). The levels of interleukin-1 beta (IL-1β), interleukin-6 (IL-6), and tumor necrosis factor-alpha (TNF-α) significantly decreased and the levels of interleukin-2 (IL-2) and interferon-gamma (IFN-γ) increased in the OVX+GNC and OVX+E groups compared with those in the OVX group (P < 0.05). OVX rats treated with GNC and estradiol further exhibited reversed ovariectomy-induced weight gain and leptin resistance (P < 0.05). These results indicated that GNC demonstrated phytoestrogen-like properties without the side effects of estradiol valerate. Thus, GNC may confer protective and beneficial effects for management of menopausal syndrome.
Collapse
Affiliation(s)
- Yan-Qiu Rao
- Department of Integrated Traditional Chinese Medicine and Western Medicine, Obstetrical and Gynecological Hospital, Fudan UniversityShanghai 200011, China
- Department of Obstetrics and Gynecology of Shanghai Medical College, Fudan UniversityShanghai 200032, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related DiseasesShanghai 200011, China
| | - Jun Li
- Department of Integrated Traditional Chinese Medicine and Western Medicine, Obstetrical and Gynecological Hospital, Fudan UniversityShanghai 200011, China
- Department of Obstetrics and Gynecology of Shanghai Medical College, Fudan UniversityShanghai 200032, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related DiseasesShanghai 200011, China
| | - Wen-Jun Wang
- Department of Integrated Traditional Chinese Medicine and Western Medicine, Obstetrical and Gynecological Hospital, Fudan UniversityShanghai 200011, China
- Department of Obstetrics and Gynecology of Shanghai Medical College, Fudan UniversityShanghai 200032, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related DiseasesShanghai 200011, China
| |
Collapse
|
36
|
Mabey T, Honsawek S. Cytokines as biochemical markers for knee osteoarthritis. World J Orthop 2015; 6:95-105. [PMID: 25621214 PMCID: PMC4303794 DOI: 10.5312/wjo.v6.i1.95] [Citation(s) in RCA: 143] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Revised: 03/24/2014] [Accepted: 07/17/2014] [Indexed: 02/06/2023] Open
Abstract
Osteoarthritis (OA) is a debilitating degenerative joint disease particularly affecting weightbearing joints within the body, principally the hips and knees. Current radiographic techniques are insufficient to show biochemical changes within joint tissue which can occur many years before symptoms become apparent. The need for better diagnostic and prognostic tools is heightened with the prevalence of OA set to increase in aging and obese populations. As inflammation is increasingly being considered an important part of OAs pathophysiology, cytokines are being assessed as possible candidates for biochemical markers. Cytokines, both pro- and anti-inflammatory, as well as angiogenic and chemotactic, have in recent years been studied for relevant characteristics. Biochemical markers show promise in determination of the severity of disease in addition to monitoring of the efficacy and safety of disease-modifying OA drugs, with the potential to act as diagnostic and prognostic tools. Currently, the diagnostic power of interleukin (IL)-6 and the relationship to disease burden of IL-1β, IL-15, tumor necrosis factor-α, and vascular endothelial growth factor make these the best candidates for assessment. Grouping appropriate cytokine markers together and assessing them collectively alongside other bone and cartilage degradation products will yield a more statistically powerful tool in research and clinical applications, and additionally aid in distinguishing between OA and a number of other diseases in which cytokines are known to have an involvement. Further large scale studies are needed to assess the validity and efficacy of current biomarkers, and to discover other potential biomarker candidates.
Collapse
|
37
|
The role of inflammatory and anti-inflammatory cytokines in the pathogenesis of osteoarthritis. Mediators Inflamm 2014; 2014:561459. [PMID: 24876674 PMCID: PMC4021678 DOI: 10.1155/2014/561459] [Citation(s) in RCA: 1046] [Impact Index Per Article: 104.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Accepted: 04/12/2014] [Indexed: 12/11/2022] Open
Abstract
Osteoarthritis (OA) is the most common chronic disease of human joints. The basis of pathologic changes involves all the tissues forming the joint; already, at an early stage, it has the nature of inflammation with varying degrees of severity. An analysis of the complex relationships indicates that the processes taking place inside the joint are not merely a set that (seemingly) only includes catabolic effects. Apart from them, anti-inflammatory anabolic processes also occur continually. These phenomena are driven by various mediators, of which the key role is attributed to the interactions within the cytokine network. The most important group controlling the disease seems to be inflammatory cytokines, including IL-1β, TNFα, IL-6, IL-15, IL-17, and IL-18. The second group with antagonistic effect is formed by cytokines known as anti-inflammatory cytokines such as IL-4, IL-10, and IL-13. The role of inflammatory and anti-inflammatory cytokines in the pathogenesis of OA with respect to inter- and intracellular signaling pathways is still under investigation. This paper summarizes the current state of knowledge. The cytokine network in OA is put in the context of cells involved in this degenerative joint disease. The possibilities for further implementation of new therapeutic strategies in OA are also pointed.
Collapse
|
38
|
Kulkarni SS, Cary LH, Gambles K, Hauer-Jensen M, Kumar KS, Ghosh SP. Gamma-tocotrienol, a radiation prophylaxis agent, induces high levels of granulocyte colony-stimulating factor. Int Immunopharmacol 2012; 14:495-503. [PMID: 23000517 DOI: 10.1016/j.intimp.2012.09.001] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2012] [Revised: 08/07/2012] [Accepted: 09/07/2012] [Indexed: 01/22/2023]
Abstract
Gamma-tocotrienol (GT3), a promising radioprotectant, is shown to protect CD2F1 mice from radiation-induced neutropenia and thrombocytopenia when given 24h prior to total-body irradiation. GT3 also is shown to increase white blood cells (WBC) and absolute neutrophil counts (ANC) transiently in peripheral blood. We hypothesized that increases in WBC and ANC may involve stimulation of hematopoiesis possibly by cytokines and growth factors. To evaluate the effects of GT3 on hematopoietic system, we measured various cytokines, chemokines and growth factors by cytokine array and Bio-Plex assays. Both showed strong induction of various cytokines and chemokines. GT3 treatment resulted in significant increases in G-CSF, IL-1α, IL-1β, IL-6, IL-12p70, IL-17, MIP-1α, and KC levels. G-CSF levels increased markedly within 12-24h after administration (5441 pg/ml in GT3-treated groups compared to 17 pg/ml in vehicle control). Most of these cytokine levels were elevated in the presence or absence of radiation. Time-course analysis of G-CSF and IL-6 induction showed that both cytokines were induced transiently after GT3 administration, and returned to normal levels by 48 h post-administration. For G-CSF, the peak was observed between 12 and 24h post-administration of GT3; however, the highest levels of IL-6 were obtained between 6 and 12h. These results demonstrate that GT3 induced high levels of G-CSF and other inflammatory cytokines and chemokines within 24h after administration. Survival studies reported showed that the most efficacious time for administering GT3 was 24h prior to irradiation, possibly because it induced key hematopoietic cytokines in that time window. These results also suggest a possible role of GT3-induced G-CSF stimulation in protecting mice from radiation-induced neutropenia and thrombocytopenia.
Collapse
Affiliation(s)
- Shilpa S Kulkarni
- Armed Forces Radiobiology Research Institute, USUHS, Bethesda, MD 20889-5603, United States
| | | | | | | | | | | |
Collapse
|
39
|
Guo DEJ, Han JS, Li YS, Liu ZS, Lu SY, Ren HL. In vitro and in vivo antitumor effects of the recombinant immunotoxin IL6(T23)-PE38KDEL in multiple myeloma. Oncol Lett 2012; 4:311-318. [PMID: 22844376 DOI: 10.3892/ol.2012.733] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2012] [Accepted: 05/23/2012] [Indexed: 01/10/2023] Open
Abstract
IL6(T23)-PE38KDEL is a chimeric molecule composed of interleukin 6 (IL6), missing the N-terminal 23 amino acids, and fused to a truncated mutant form of Pseudomonas exotoxin (PE38KDEL). The aim of this study was to evaluate this recombinant immunotoxin in terms of its specific cytotoxicity to IL6R-overexpressing multiple myeloma (MM) cells in vitro, as well as its antitumor effects and side effects in vivo. IL6(T23)-PE38KDEL was expressed in Escherichia coli, refolded and purified from inclusion bodies. The purified IL6(T23)-PE38KDEL was found to be selectively cytotoxic to IL6 receptor-positive tumor cells in vitro. IC(50) values of IL6(T23)-PE38KDEL were evaluated by MTS assay. Toxicity and maximum-tolerated dose of IL6(T23)-PE38KDEL were determined in mice. The antitumor activity of IL6(T23)-PE38KDEL was evaluated in mice with MM through intravenous injection and interventional therapy. Intravenous administration of IL6(T23)-PE38KDEL caused a significantly increased survival time in treated mice, and exhibited dose- and time-dependent antitumor effects against MM mice. Moreover, complete tumor regression was observed in 30 and 80% of mice treated intravenously and intraperitoneally, respectively, with 0.4 mg/kg/day for 10 days. These results demonstrated that the recombinant immunotoxin IL6(T23)-PE38KDEL kills IL6R-overexpressing cancer cells, and causes significant tumor regression.
Collapse
Affiliation(s)
- DE-Jun Guo
- College of Animal Science and Veterinary Medicine, Jilin University, Changchun, Jilin 130062
| | | | | | | | | | | |
Collapse
|
40
|
Rubio-Perez JM, Morillas-Ruiz JM. A review: inflammatory process in Alzheimer's disease, role of cytokines. ScientificWorldJournal 2012; 2012:756357. [PMID: 22566778 PMCID: PMC3330269 DOI: 10.1100/2012/756357] [Citation(s) in RCA: 536] [Impact Index Per Article: 44.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2011] [Accepted: 12/11/2011] [Indexed: 12/21/2022] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorder to date. Neuropathological hallmarks are β-amyloid (Aβ) plaques and neurofibrillary tangles, but the inflammatory process has a fundamental role in the pathogenesis of AD. Inflammatory components related to AD neuroinflammation include brain cells such as microglia and astrocytes, the complement system, as well as cytokines and chemokines. Cytokines play a key role in inflammatory and anti-inflammatory processes in AD. An important factor in the onset of inflammatory process is the overexpression of interleukin (IL)-1, which produces many reactions in a vicious circle that cause dysfunction and neuronal death. Other important cytokines in neuroinflammation are IL-6 and tumor necrosis factor (TNF)-α. By contrast, other cytokines such as IL-1 receptor antagonist (IL-1ra), IL-4, IL-10, and transforming growth factor (TGF)-β can suppress both proinflammatory cytokine production and their action, subsequently protecting the brain. It has been observed in epidemiological studies that treatment with nonsteroidal anti-inflammatory drugs (NSAIDs) decreases the risk for developing AD. Unfortunately, clinical trials of NSAIDs in AD patients have not been very fruitful. Proinflammatory responses may be countered through polyphenols. Supplementation of these natural compounds may provide a new therapeutic line of approach to this brain disorder.
Collapse
Affiliation(s)
- Jose Miguel Rubio-Perez
- Department of Food and Nutrition Technology, St. Anthony Catholic University, Campus de Los Jerónimos, s/n Guadalupe, 30107 Murcia, Spain
| | - Juana Maria Morillas-Ruiz
- Department of Food and Nutrition Technology, St. Anthony Catholic University, Campus de Los Jerónimos, s/n Guadalupe, 30107 Murcia, Spain
| |
Collapse
|
41
|
Phenotype-information-phenotype cycle for deconvolution of combinatorial antibody libraries selected against complex systems. Proc Natl Acad Sci U S A 2011; 108:13456-61. [PMID: 21825149 DOI: 10.1073/pnas.1111218108] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Use of large combinatorial antibody libraries and next-generation sequencing of nucleic acids are two of the most powerful methods in modern molecular biology. The libraries are screened using the principles of evolutionary selection, albeit in real time, to enrich for members with a particular phenotype. This selective process necessarily results in the loss of information about less-fit molecules. On the other hand, sequencing of the library, by itself, gives information that is mostly unrelated to phenotype. If the two methods could be combined, the full potential of very large molecular libraries could be realized. Here we report the implementation of a phenotype-information-phenotype cycle that integrates information and gene recovery. After selection for phage-encoded antibodies that bind to targets expressed on the surface of Escherichia coli, the information content of the selected pool is obtained by pyrosequencing. Sequences that encode specific antibodies are identified by a bioinformatic analysis and recovered by a stringent affinity method that is uniquely suited for gene isolation from a highly degenerate collection of nucleic acids. This approach can be generalized for selection of antibodies against targets that are present as minor components of complex systems.
Collapse
|
42
|
Mota BC, Pereira L, Souza MA, Silva LFA, Magni DV, Ferreira APO, Oliveira MS, Furian AF, Mazzardo-Martins L, Silva MDD, Santos ARS, Ferreira J, Fighera MR, Royes LFF. Exercise pre-conditioning reduces brain inflammation and protects against toxicity induced by traumatic brain injury: behavioral and neurochemical approach. Neurotox Res 2011; 21:175-84. [PMID: 21735317 DOI: 10.1007/s12640-011-9257-8] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2011] [Revised: 06/15/2011] [Accepted: 06/22/2011] [Indexed: 01/05/2023]
Abstract
Although the favorable effects of physical exercise in neurorehabilitation after traumatic brain injury (TBI) are well known, detailed pathologic and functional alterations exerted by previous physical exercise on post-traumatic cerebral inflammation have been limited. In the present study, it is showed that fluid percussion brain injury (FPI) induced motor function impairment, followed by increased plasma fluorescein extravasation and cerebral inflammation characterized by interleukin-1β, tumor necrosis factor-α (TNF-α) increase, and decreased IL-10. In addition, myeloperoxidase (MPO) increase and Na⁺,K⁺-ATPase activity inhibition after FPI suggest that the opening of blood-brain barrier (BBB) followed by neurtrophils infiltration and cerebral inflammation may contribute to the failure of selected targets leading to secondary damage. In fact, Pearson's correlation analysis revealed strong correlation of MPO activity increase with Na⁺,K⁺-ATPase activity inhibition in sedentary rats. Statistical analysis also revealed that previous running exercise (4 weeks) protected against FPI-induced motor function impairment and fluorescein extravasation. Previous physical training also induced IL-10 increase per se and protected against cerebral IL-1β, and TNF-α increase and IL-10 decrease induced by FPI. This protocol of physical training was effective against MPO activity increase and Na⁺,K⁺-ATPase activity inhibition after FPI. The present protection correlated with MPO activity decrease suggests that the alteration of cerebral inflammatory status profile elicited by previous physical training reduces initial damage and limits long-term secondary degeneration after TBI. This prophylactic effect may facilitate functional recovery in patients suffering from brain injury induced by TBI.
Collapse
Affiliation(s)
- Bibiana Castagna Mota
- Laboratório de Bioquímica do Exercício, Departamento de Métodos e Técnicas Desportivas, Centro de Educação Física e Desportos, Universidade Federal de Santa Maria, Santa Maria, RS 97105-900, Brazil
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Ziebell JM, Morganti-Kossmann MC. Involvement of pro- and anti-inflammatory cytokines and chemokines in the pathophysiology of traumatic brain injury. Neurotherapeutics 2010; 7:22-30. [PMID: 20129494 PMCID: PMC5084109 DOI: 10.1016/j.nurt.2009.10.016] [Citation(s) in RCA: 497] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2009] [Revised: 10/27/2009] [Accepted: 10/28/2009] [Indexed: 12/12/2022] Open
Abstract
Despite dramatic improvements in the management of traumatic brain injury (TBI), to date there is no effective treatment available to patients, and morbidity and mortality remain high. The damage to the brain occurs in two phases, the initial primary phase being the injury itself, which is irreversible and amenable only to preventive measures to minimize the extent of damage, followed by an ongoing secondary phase, which begins at the time of injury and continues in the ensuing days to weeks. This delayed phase leads to a variety of physiological, cellular, and molecular responses aimed at restoring the homeostasis of the damaged tissue, which, if not controlled, will lead to secondary insults. The development of secondary brain injury represents a window of opportunity in which pharmaceutical compounds with neuroprotective properties could be administered. To establish effective treatments for TBI victims, it is imperative that the complex molecular cascades contributing to secondary injury be fully elucidated. One pathway known to be activated in response to TBI is cellular and humoral inflammation. Neuroinflammation within the injured brain has long been considered to intensify the damage sustained following TBI. However, the accumulated findings from years of clinical and experimental research support the notion that the action of inflammation may differ in the acute and delayed phase after TBI, and that maintaining limited inflammation is essential for repair. This review addresses the role of several cytokines and chemokines following focal and diffuse TBI, as well as the controversies around the use of therapeutic anti-inflammatory treatments versus genetic deletion of cytokine expression.
Collapse
Affiliation(s)
- Jenna M. Ziebell
- grid.1002.30000000419367857National Trauma Research Institute (NTRI), The Alfred Hospital, and Department of Medicine, Monash University, 3181 Melbourne, VIC Australia
| | - Maria Cristina Morganti-Kossmann
- grid.1002.30000000419367857National Trauma Research Institute (NTRI), The Alfred Hospital, and Department of Medicine, Monash University, 3181 Melbourne, VIC Australia
| |
Collapse
|
44
|
Dubinsky V, Junovich G, Gentile T, Gutiérrez G. ORIGINAL ARTICLE: IL-6 as a Regulatory Factor of the Humoral Response During Pregnancy. Am J Reprod Immunol 2008; 60:197-203. [DOI: 10.1111/j.1600-0897.2008.00614.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
45
|
Denessiouk KA, Denesyuk AI, Johnson MS. Negative modulation of signal transduction via interleukin splice variation. Proteins 2008; 71:751-70. [PMID: 17979192 DOI: 10.1002/prot.21756] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Interleukin 6 (IL-6) belongs to a large group of secreted proteins called cytokines functioning to mediate and regulate immunity, inflammation, and hematopoiesis with direct effects on cell proliferation, apoptosis, and differentiation. Along with the IL-6 protein, two of its splice variants, IL-6delta2 and IL-6delta4, were reported to be transcribed or expressed in vivo in human, and the mRNAs of IL-6delta3 and IL-6delta5 had been observed in mouse. While the existence of different splice variants of IL-6 has been shown, very little is known on how the structural modifications of IL-6 resulting from the formation of the different splice variants may alter cytokine functions. We have analyzed the potential effects splicing would have on interactions with the cell surface receptor complex. We (1) constructed three-dimensional structures of the IL-6 splice variants, IL-6delta2, IL-6delta3, and IL-6delta4, with the assumption that an interleukin splice variant as a folded protein should retain a functional hydrophobic core; (2) reconstructed the ternary structural complexes consisting of the modeled IL-6 splice variants, the IL-6 receptor molecule (IL-6R) and the dimeric signal-transducing protein, gp130, and (3) analyzed all complexes and made comparisons with the X-ray structure of the wild-type IL-6 complex. We identified three separate sites on IL-6 where interactions are made with IL-6R and with each of the two copies of gp130. The structural consequences of losing an exon lead to a unique pattern of lost interaction with different components of the receptor complex. Thus, in IL-6 and its splice variants, the exons appear to have compartmentalized roles contributing to the combined function of the cytokine. The modeled interactions suggest that splice variants could act as antagonists, and that IL-6delta2, missing the signal peptide, would be a cytoplasmic protein and be released and interact with nearby cell-surface receptors when cells are damaged. We argue that in the case of IL-6, helix E may act as a "silent secondary structure," which only has an active role when it substitutes for a part of the hydrophobic core, for example, replacing helix A in IL-6delta2.
Collapse
Affiliation(s)
- Konstantin A Denessiouk
- Department of Biochemistry and Pharmacy, Abo Akademi University, Artillerigatan 6 A, 20521 Turku, Finland
| | | | | |
Collapse
|
46
|
Silverman J, Liu Q, Lu Q, Bakker A, To W, Duguay A, Alba BM, Smith R, Rivas A, Li P, Le H, Whitehorn E, Moore KW, Swimmer C, Perlroth V, Vogt M, Kolkman J, Stemmer WPC. Multivalent avimer proteins evolved by exon shuffling of a family of human receptor domains. Nat Biotechnol 2005; 23:1556-61. [PMID: 16299519 DOI: 10.1038/nbt1166] [Citation(s) in RCA: 145] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2005] [Accepted: 10/11/2005] [Indexed: 11/09/2022]
Abstract
We have developed a class of binding proteins, called avimers, to overcome the limitations of antibodies and other immunoglobulin-based therapeutic proteins. Avimers are evolved from a large family of human extracellular receptor domains by in vitro exon shuffling and phage display, generating multidomain proteins with binding and inhibitory properties. Linking multiple independent binding domains creates avidity and results in improved affinity and specificity compared with conventional single-epitope binding proteins. Other potential advantages over immunoglobulin domains include simple and efficient production of multitarget-specific molecules in Escherichia coli, improved thermostability and resistance to proteases. Avimers with sub-nM affinities were obtained against five targets. An avimer that inhibits interleukin 6 with 0.8 pM IC50 in cell-based assays is biologically active in two animal models.
Collapse
Affiliation(s)
- Joshua Silverman
- Avidia, Inc., 2450 Bayshore Parkway, Mountain View, California 94043, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Abstract
The defense of the host from foreign pathogens is the commonly accepted function of the vertebrate immune system. A complex system consisting of many differing cells and structures communicating by both soluble and cell bound ligands, serves to protect the host from infection, and plays a role in preventing the development of certain types of tumours. Numerous signalling pathways are involved in the coordination of the immune system, serving both to activate and attenuate its responses to attack. The ability of the immune system, specifically those cells involved in acute inflammatory responses, to mediate the directed (and sometimes indirect) killing of cells and pathogens, make it a potential threat to host survival. Furthermore, the production and release of various survival factors such as the pleiotropic cytokine IL-6, a major mediator of inflammation and activator of signal transducer and activator of transcription 3, serves to block apoptosis in cells during the inflammatory process, keeping them alive in very toxic environments. Unfortunately, these same pathways serve also to maintain cells progressing towards neoplastic growth, protecting them from cellular apoptotic deletion and chemotherapeutic drugs. Here, we discuss the relationships between cancer and inflammation, and some of the molecular mechanisms involved in mediating the unintended consequences of host defense and tumour survival.
Collapse
Affiliation(s)
- David R Hodge
- Laboratory of Molecular Immunoregulation, Cytokine Molecular Mechanisms Section, Center for Cancer Research, The National Cancer Institute at Frederick, Frederick, MD 21702, USA
| | | | | |
Collapse
|
48
|
Hammacher A, Thompson EW, Williams ED. Interleukin-6 is a potent inducer of S100P, which is up-regulated in androgen-refractory and metastatic prostate cancer. Int J Biochem Cell Biol 2005; 37:442-50. [PMID: 15474988 DOI: 10.1016/j.biocel.2004.07.011] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2004] [Revised: 07/21/2004] [Accepted: 07/21/2004] [Indexed: 10/26/2022]
Abstract
Elevated circulating interleukin-6 (IL6) and up-regulated S100P in prostate cancer (PCa) specimens correlate independently with progression to androgen-independent and metastatic PCa. The cause of up-regulated S100P levels in advanced PCa remains to be determined. We investigated the possibility that IL6 is an inducer of S100P. Determination of mRNA and protein levels by real-time PCR and Western blotting revealed that IL6 is a more potent inducer of S100P than the synthetic androgen, R1881, in the LNCaP/C4-2B model of PCa progression. IL6 did not require androgen to induce S100P in these cells, which express a functional androgen receptor (AR). Like R1881, IL6 was unable to induce S100P in PC3 cells that lack a functional AR. IL6 did not strongly induce the AR-dependent genes PSA and KLK2 and, contrary to R1881, down-regulated Cyr61/CCN1, a potential marker that is down-regulated in PCa. Epidermal growth factor (EGF), which like IL6 is a non-androgen activator of the AR, did not induce S100P. The data identifies a unique gene-induction profile for IL6 and suggests that IL6 may require a functional AR for S100P induction. A link between elevated IL6 and up-regulated S100P in androgen-refractory and metastatic PCa is postulated.
Collapse
Affiliation(s)
- Annet Hammacher
- Bernard O'Brien Institute of Microsurgery, University of Melbourne, 42 Fitzroy Street, Fitzroy, Vic. 3065, Australia.
| | | | | |
Collapse
|
49
|
Palmer J, Hertzog PJ, Hammacher A. Differential expression and effects of gp130 cytokines and receptors in prostate cancer cells. Int J Biochem Cell Biol 2005; 36:2258-69. [PMID: 15313471 DOI: 10.1016/j.biocel.2004.04.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2003] [Revised: 04/15/2004] [Accepted: 04/22/2004] [Indexed: 12/27/2022]
Abstract
High levels of circulating interleukin-6 (IL6), and possibly neuroendocrine (NE) differentiation, correlate with advanced prostate cancer (PCa). IL6 has many overlapping biological effects with the related gp130 cytokines LIF and OSM that can be explained by the shared usage of the signalling receptor, gp130. We set out to determine whether LIF and OSM can substitute for IL6 in PCa, particularly in relation to neuroendocrine differentiation. Expression analysis of the gp130 cytokines and receptors by RT-PCR, Southern blotting and immunohistochemistry showed that they are widely expressed in LNCaP, DU145 and PC3 cells, but not in normal prostate epithelial PZ-HPV-7 cells. IL6, but not LIF or OSM inhibited proliferation, induced NE differentiation and tyrosine phosphorylation of STAT3 in LNCaP cells. The data suggests that IL6 has a unique role in the progression of PCa.
Collapse
Affiliation(s)
- J Palmer
- Monash Institute of Reproduction and Development, Monash University, Clayton, Australia
| | | | | |
Collapse
|
50
|
Li H, Wang H, Nicholas J. Detection of direct binding of human herpesvirus 8-encoded interleukin-6 (vIL-6) to both gp130 and IL-6 receptor (IL-6R) and identification of amino acid residues of vIL-6 important for IL-6R-dependent and -independent signaling. J Virol 2001; 75:3325-34. [PMID: 11238858 PMCID: PMC114125 DOI: 10.1128/jvi.75.7.3325-3334.2001] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Human herpesvirus 8 (HHV-8) is associated with Kaposi's sarcoma, primary effusion lymphoma, and multicentric Castleman's disease; in all of these diseases, interleukin-6 (IL-6) has been implicated as a likely mitogenic and/or angiogenic factor. HHV-8 encodes a homologue of IL-6 (viral IL-6 [vIL-6]) that has been shown to be biologically active in several assays and whose activities mirror those of its mammalian counterparts. Like these proteins, vIL-6 mediates its effects through the gp130 signal transducer, but signaling is not dependent on the structurally related IL-6 receptor (IL-6R; gp80) subunit of the receptor-signal transducer complex. However, as we have shown previously, IL-6R can enhance vIL-6 signal transduction and can enable signaling through a gp130 variant (gp130.PM5) that is itself unable to support vIL-6 activity, indicating that IL-6R can form part of the signaling complex. Also, our analysis of a panel of vIL-6 mutants in transfection experiments in Hep3B cells (that express IL-6R and gp130) showed that most were able to function normally in this system. Here, we have used in vitro vIL-6-receptor binding assays to demonstrate direct binding of vIL-6 to both gp130 and IL-6R and vIL-6-induced gp130-IL-6R complex formation, and we have extended our functional analyses of the vIL-6 variants to identify residues important for IL-6R-independent and IL-6R-dependent signaling through native gp130 and gp130.PM5, respectively. These studies have identified residues in vIL-6 that are important for IL-6R-independent and IL-6R-mediated functional complex formation between vIL-6 and gp130 and that may be involved directly in binding to gp130 and IL-6R.
Collapse
Affiliation(s)
- H Li
- The Molecular Virology Laboratories, Department of Oncology, Johns Hopkins University, Baltimore, Maryland 21231, USA.
| | | | | |
Collapse
|