1
|
Ali MS, Hadda V, Verma S, Chopra A, Mittal S, Madan K, Tiwari P, Suri TM, Mohan A. Unravelling the transcriptomic characteristics of bronchoalveolar lavage in post-covid pulmonary fibrosis. BMC Med Genomics 2025; 18:54. [PMID: 40098116 PMCID: PMC11917041 DOI: 10.1186/s12920-025-02110-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Accepted: 02/20/2025] [Indexed: 03/19/2025] Open
Abstract
BACKGROUND Post-Covid Pulmonary Fibrosis (PCPF) has emerged as a significant global issue associated with a poor quality of life and significant morbidity. Currently, our understanding of the molecular pathways of PCPF is limited. Hence, in this study, we performed whole transcriptome sequencing of the RNA isolated from the bronchoalveolar lavage (BAL) samples of PCPF and compared it with idiopathic pulmonary fibrosis (IPF) and non-ILD (Interstitial Lung Disease) control to understand the gene expression profile and associated pathways. METHODS BAL samples from PCPF (n = 3), IPF (n = 3), and non-ILD Control (n = 3) (individuals with apparent healthy lung without interstitial lung disease) groups were obtained and RNA were isolated for whole transcriptomic sequencing. Differentially Expressed Genes (DEGs) were determined followed by functional enrichment analysis and qPCR validation. RESULTS A panel of differentially expressed genes were identified in bronchoalveolar lavage fluid cells (BALF) of PCPF as compare to control and IPF. Our analysis revealed dysregulated pathways associated with cell cycle regulation, immune responses, and neuroinflammatory processes. Real-time validation further supported these findings. The PPI network and module analysis shed light on potential biomarkers and underscore the complex interplay of molecular mechanisms in PCPF. The comparison of PCPF and IPF identified a significant downregulation of pathways that were more prominent in IPF. CONCLUSION This investigation provides crucial insights into the molecular mechanism of PCPF and also outlines avenues for prospective research and the development of therapeutic approaches.
Collapse
Affiliation(s)
- Mohammad Shadab Ali
- Department of Pulmonary, Critical Care, & Sleep Medicine, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| | - Vijay Hadda
- Department of Pulmonary, Critical Care, & Sleep Medicine, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India.
| | - Sonia Verma
- Division of Neuroscience and Ageing Biology, CSIR-Central Drug Research Institute, Lucknow, UP, 226031, India
| | - Anita Chopra
- Lab Oncology, Dr. BRAIRCH All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| | - Saurabh Mittal
- Department of Pulmonary, Critical Care, & Sleep Medicine, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| | - Karan Madan
- Department of Pulmonary, Critical Care, & Sleep Medicine, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| | - Pawan Tiwari
- Department of Pulmonary, Critical Care, & Sleep Medicine, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| | - Tejas Menon Suri
- Department of Pulmonary, Critical Care, & Sleep Medicine, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| | - Anant Mohan
- Department of Pulmonary, Critical Care, & Sleep Medicine, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| |
Collapse
|
2
|
Latayan J, Akkenapally SV, Madala SK. Emerging Concepts in Cytokine Regulation of Airway Remodeling in Asthma. Immunol Rev 2025; 330:e70020. [PMID: 40116139 PMCID: PMC11926778 DOI: 10.1111/imr.70020] [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: 08/24/2024] [Revised: 02/18/2025] [Accepted: 02/28/2025] [Indexed: 03/23/2025]
Abstract
Asthma, a chronic respiratory condition that has seen a dramatic rise in prevalence over the past few decades, now affects more than 300 million people globally and imposes a significant burden on healthcare systems. The key pathological features of asthma include inflammation, airway hyperresponsiveness, mucus cell metaplasia, smooth muscle hypertrophy, and subepithelial fibrosis. Cytokines released by lung epithelial cells, stromal cells, and immune cells during asthma are critical to pathological tissue remodeling in asthma. Over the past few decades, researchers have made great strides in understanding key cells involved in asthma and the cytokines that they produce. Epithelial cells as well as many adaptive and innate immune cells are activated by environmental signals to produce cytokines, namely, type 2 cytokines (IL-4, IL-5, IL-13), IFN-γ, IL-17, TGF-β, and multiple IL-6 family members. However, the precise mechanisms through which these cytokines contribute to airway remodeling remain elusive. Additionally, multiple cell types can produce the same cytokines, making it challenging to decipher how specific cell types and cytokines uniquely contribute to asthma pathogenesis. This review highlights recent advances and provides a comprehensive overview of the key cells involved in the production of cytokines and how these cytokines modulate airway remodeling in asthma.
Collapse
Affiliation(s)
- Jana Latayan
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal MedicineThe University of CincinnatiCincinnatiOhioUSA
- Immunology Graduate ProgramUniversity of CincinnatiCincinnatiOhioUSA
| | - Santhoshi V. Akkenapally
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal MedicineThe University of CincinnatiCincinnatiOhioUSA
| | - Satish K. Madala
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal MedicineThe University of CincinnatiCincinnatiOhioUSA
| |
Collapse
|
3
|
Jones LE, Ghassabian A, Yeung E, Mendola P, Kannan K, Bell EM. Maternal exposure to legacy PFAS compounds PFOA and PFOS is associated with disrupted cytokine homeostasis in neonates: The Upstate KIDS study (2008-2010). ENVIRONMENT INTERNATIONAL 2025; 196:109288. [PMID: 39848095 DOI: 10.1016/j.envint.2025.109288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 01/03/2025] [Accepted: 01/15/2025] [Indexed: 01/25/2025]
Abstract
There is growing concern that exposure to per/polyfluoroalkyl substances (PFAS), persistent chemicals used widely to make consumer products water- or grease-proof, may alter immune function, leading to reduced vaccine response or greater susceptibility to infections. We investigated associations between two legacy PFAS (PFOA and PFOS) and infant cytokine levels measured in newborn dried bloodspots (NDBS) from a large population-based birth cohort in Upstate New York, to determine whether exposure to legacy PFAS is associated with variability in cytokine profiles in newborns. We performed adjusted mixed effects regressions for each cytokine against PFOS and PFOA followed by exploratory factor analysis (EFA) on specific cytokine subsets selected via the prior regressions. Among 3448 neonates (2280 singletons and 1168 twins), significant cytokines were dominated by cytokines negatively associated with the given PFAS. Adjusted single-pollutant models with continuous log-transformed PFOA showed significant negative associations with IL-16 (-0.07, 95% CI: -0.3, -0.1), IL-5 (-0.05, 95%CI: -0.09, -0.02), IL-6 (-0.06, 95%CI: -0.1, -0.02), 6-Ckine (0.06, 95% CI: -0.10, -0.02) and significant positive associations with IL-1α (0.066, 95%CI: 0.03, 0.11), MCP-1 (0.06, 95%CI: 0.03, 0.10). Estimates for PFOS were slightly larger than estimates for PFOA but only significant for 6-Ckine (-0.21, 95%CI: -0.09, -0.33) after correction for multiplicity. Our data consistently suggest that legacy PFAS exposures are associated with disrupted, typically reduced, cytokine levels in neonates, with PFOA exposure resulting in more significant differences in individual cytokines and cytokine groupings than PFOS. Regression by PFAS quartile shows evidence of nonlinear dose-response relationships for most cytokines and cytokine groupings.
Collapse
Affiliation(s)
- Laura E Jones
- Center for Biostatistics, Bassett Research Institute, 1 Atwell Rd. Cooperstown NY USA.
| | - Akhgar Ghassabian
- Department of Pediatrics, New York University Grossman School of Medicine NY USA; Department of Population Health, New York University Grossman School of Medicine NY USA.
| | - Edwina Yeung
- Epidemiology Branch, Division of Population Health Research, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, 6710B Rockledge Drive Bethesda MD USA.
| | - Pauline Mendola
- Department of Epidemiology and Environmental Health, School of Public Health and Health Professions, University at Buffalo Buffalo NY USA.
| | - Kurunthachalam Kannan
- Wadsworth Center, New York State Department of Health Albany NY USA; Environmental Health Sciences, College of Integrated Health Sciences, University at Albany, Rensselaer, NY, USA.
| | - Erin M Bell
- Environmental Health Sciences, College of Integrated Health Sciences, University at Albany, Rensselaer, NY, USA; Department of Epidemiology and Biostatistics, College of Integrated Health Sciences, University at Albany, State University of New York, Rensselaer, NY, USA.
| |
Collapse
|
4
|
Park W, Lim W, Kim M, Jang H, Park SJ, Song G, Park S. Female reproductive disease, endometriosis: From inflammation to infertility. Mol Cells 2025; 48:100164. [PMID: 39617101 PMCID: PMC11760828 DOI: 10.1016/j.mocell.2024.100164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 11/21/2024] [Accepted: 11/26/2024] [Indexed: 01/11/2025] Open
Abstract
Despite the fact that endometriosis is a common gynecological disease that occurs in 10% of women of reproductive age, the pathogenesis and treatment strategy are not clear to date. Endometriosis patients are commonly characterized by adhesions in the pelvis or ovaries, which leads to prolonged inflammation in the abdominal cavity. To handle the chronic inflammation, changes of immune cells, including T cells, NK cells, and macrophage, are accompanied. Therefore, diverse cytokines and adhesions of the abdominal cavity lead to poor quality of ovarian follicles, inappropriate response to the hormone, and infertility. This review will guide researchers to summarize the molecular changes and identify new treatment strategies for endometriosis-mediated inflammation and pregnancy failure.
Collapse
Affiliation(s)
- Wonhyoung Park
- Department of Animal Science, Chungbuk National University, Cheongju 28644, Republic of Korea
| | - Whasun Lim
- Department of Biological Sciences, College of Science, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Miji Kim
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea
| | - Hyewon Jang
- Department of Biological Sciences, College of Science, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Soo Jin Park
- Department of Obstetrics and Gynecology, Seoul National University Hospital, Seoul 03080, Republic of Korea
| | - Gwonhwa Song
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea.
| | - Sunwoo Park
- Department of GreenBio Science, Gyeongsang National University, Jinju 52725, Republic of Korea.
| |
Collapse
|
5
|
Krygere L, Jukna P, Jariene K, Drejeriene E. Diagnostic Potential of Cytokine Biomarkers in Endometriosis: Challenges and Insights. Biomedicines 2024; 12:2867. [PMID: 39767772 PMCID: PMC11673701 DOI: 10.3390/biomedicines12122867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 12/15/2024] [Accepted: 12/16/2024] [Indexed: 01/11/2025] Open
Abstract
Endometriosis is a common gynecological condition affecting approximately 10% of women of reproductive age, characterized by the abnormal presence of endometrial-like tissue outside the uterus. Although endometriosis was first described over 300 years ago, its underlying mechanisms remain poorly understood, and accurate, prompt diagnosis continues to be challenging. Currently, there is a lack of effective, non-invasive diagnostic methods, and available treatments often come with significant side effects and high recurrence rates. This has spurred interest in investigating the role of pro- and anti-inflammatory molecules, particularly cytokines, in endometriosis, as these molecules play a key role in its progression by influencing cell growth and differentiation. Previous studies suggest that various cytokines could serve as potential biomarkers for diagnosing endometriosis, as they are detectable in both serum and peritoneal fluid. This review provides an overview of the expression, origin, function, and regulation of specific cytokines in endometriosis, along with a brief discussion on their potential clinical implications for diagnosis. Due to the complexity of endometriosis, a panel of multiple biomarkers may ultimately be necessary for accurate diagnosis. It is essential to consider factors such as patient selection, sample collection, and analytical variability when initiating or evaluating biomarker studies.
Collapse
Affiliation(s)
- Laura Krygere
- Department of Obstetrics and Gynaecology, Medical Academy, Lithuanian University of Health Sciences, LT-50161 Kaunas, Lithuania; (L.K.); (P.J.); (K.J.)
| | - Povilas Jukna
- Department of Obstetrics and Gynaecology, Medical Academy, Lithuanian University of Health Sciences, LT-50161 Kaunas, Lithuania; (L.K.); (P.J.); (K.J.)
- Department of Obstetrics and Gynaecology, Hospital of Lithuanian University of Health Sciences Kauno Klinikos, LT-50161 Kaunas, Lithuania
| | - Kristina Jariene
- Department of Obstetrics and Gynaecology, Medical Academy, Lithuanian University of Health Sciences, LT-50161 Kaunas, Lithuania; (L.K.); (P.J.); (K.J.)
- Department of Obstetrics and Gynaecology, Hospital of Lithuanian University of Health Sciences Kauno Klinikos, LT-50161 Kaunas, Lithuania
| | - Egle Drejeriene
- Department of Obstetrics and Gynaecology, Medical Academy, Lithuanian University of Health Sciences, LT-50161 Kaunas, Lithuania; (L.K.); (P.J.); (K.J.)
- Department of Obstetrics and Gynaecology, Hospital of Lithuanian University of Health Sciences Kauno Klinikos, LT-50161 Kaunas, Lithuania
| |
Collapse
|
6
|
Omidvar-Mehrabadi A, Ebrahimi F, Shahbazi M, Mohammadnia-Afrouzi M. Cytokine and chemokine profiles in women with endometriosis, polycystic ovary syndrome, and unexplained infertility. Cytokine 2024; 178:156588. [PMID: 38555853 DOI: 10.1016/j.cyto.2024.156588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 03/19/2024] [Accepted: 03/22/2024] [Indexed: 04/02/2024]
Abstract
Numerous factors (including immunological, congenital, hormonal, and morphological disorders) can lead to infertility. In this regard, 3 specific diseases associated with infertility are discussed in this review study (i.e., polycystic ovary syndrome [PCOS], endometriosis [EMS], and unexplained infertility [UI]). PCOS is a common endocrine disorder characterized by chronic low-grade inflammation, and EMS is a benign disease characterized by the presence of ectopic endometrial tissue. UI refers to couples who are unable to conceive for no known reason. Conception and pregnancy are significantly affected by the immune system; in this regard, chemokines and cytokines play important roles in the regulation of immune responses. Patients with PCOS, EMS, and UI have altered cytokine and chemokine profiles, suggesting that dysregulation of these molecules may contribute to infertility in these conditions. Accordingly, the issue of infertility is addressed in this review study, a condition that affects approximately 16% of couples worldwide.
Collapse
Affiliation(s)
| | - Fateme Ebrahimi
- Department of Immunology, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Mehdi Shahbazi
- Department of Immunology, School of Medicine, Babol University of Medical Sciences, Babol, Iran.
| | | |
Collapse
|
7
|
Hashemi SF, Khorramdelazad H. The cryptic role of CXCL17/CXCR8 axis in the pathogenesis of cancers: a review of the latest evidence. J Cell Commun Signal 2023; 17:409-422. [PMID: 36352331 PMCID: PMC10409701 DOI: 10.1007/s12079-022-00699-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 08/17/2022] [Accepted: 09/12/2022] [Indexed: 11/10/2022] Open
Abstract
Chemokines are immune system mediators that mediate various activities and play a role in the pathogenesis of several cancers. Among these chemokines, C-X-C motif chemokine 17 (CXCL-17) is a relatively novel molecule produced along the airway epithelium in physiological and pathological conditions, and evidence shows that it plays a homeostatic role in most cases. CXCL17 has a protective role in some cancers and a pathological role in others, such as liver and lung cancer. This chemokine, along with its possible receptor termed G protein-coupled receptor 35 (GPR35) or CXCR8, are involved in recruiting myeloid cells, regulating angiogenesis, defending against pathogenic microorganisms, and numerous other mechanisms. Considering the few studies that have been performed on the dual role of CXCL17 in human malignancies, this review has investigated the possible pro-tumor and anti-tumor roles of this chemokine, as well as future treatment options in cancer therapy.
Collapse
Affiliation(s)
| | - Hossein Khorramdelazad
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.
| |
Collapse
|
8
|
Israr M, DeVoti JA, Papayannakos CJ, Bonagura VR. Role of chemokines in HPV-induced cancers. Semin Cancer Biol 2022; 87:170-183. [PMID: 36402301 DOI: 10.1016/j.semcancer.2022.11.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 11/10/2022] [Accepted: 11/14/2022] [Indexed: 11/18/2022]
Abstract
Human papillomaviruses (HPVs) cause cancers of the uterine cervix, oropharynx, anus, and vulvovaginal tract. Low-risk HPVs, such as HPV6 and 11, can also cause benign mucosal lesions including genital warts, and in patients with recurrent respiratory papillomatosis, lesions in the larynx, and on occasion, in the lungs. However, both high and less tumorigenic HPVs share a striking commonality in manipulating both innate and adaptive immune responses in HPV- infected keratinocytes, the natural host for HPV infection. In addition, immune/inflammatory cell infiltration into the tumor microenvironment influences cancer growth and prognosis, and this process is tightly regulated by different chemokines. Chemokines are small proteins and exert their biological effects by binding with G protein-coupled chemokine receptors (GPCRs) that are found on the surfaces of select target cells. Chemokines are not only involved in the establishment of a pro-tumorigenic microenvironment and organ-directed metastases but also involved in disease progression through enhancing tumor cell growth and proliferation. Therefore, having a solid grasp on chemokines and immune checkpoint modulators can help in the treatment of these cancers. In this review, we discuss the recent advances on the expression patterns and regulation of the main chemokines found in HPV-induced cancers, and their effects on both immune and non-immune cells in these lesions. Importantly, we also present the current knowledge of therapeutic interventions on the expression of specific chemokine and their receptors that have been shown to influence the development and progression of HPV-induced cancers.
Collapse
Affiliation(s)
- Mohd Israr
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States; The Department of Pediatrics, The Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, United States
| | - James A DeVoti
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States; The Department of Pediatrics, The Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, United States
| | - Christopher J Papayannakos
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States; The Department of Pediatrics, The Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, United States
| | - Vincent R Bonagura
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States; The Department of Pediatrics, The Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, United States.
| |
Collapse
|
9
|
The role of the Immune System in the Development of Endometriosis. Cells 2022; 11:cells11132028. [PMID: 35805112 PMCID: PMC9265783 DOI: 10.3390/cells11132028] [Citation(s) in RCA: 82] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 06/19/2022] [Accepted: 06/23/2022] [Indexed: 12/10/2022] Open
Abstract
Endometriosis is a chronic disease that affects about 10% of women of reproductive age. It can contribute to pelvic pain, infertility or other conditions such as asthma, cardiovascular disease, breast or ovarian cancer. Research has shown that one of the conditions for the development of endometrial lesions is the dysfunction of the immune system. It appears that immune cells, such as neutrophils, macrophages, NK cells and dendritic cells, may play a specific role in the angiogenesis, growth and invasion of endometriosis cells. Immune cells secrete cytokines and defensins that also affect the endometriosis environment. This review discusses the various components of the immune system that are involved in the formation of endometrial lesions in women.
Collapse
|
10
|
Bridges K, Miller-Jensen K. Mapping and Validation of scRNA-Seq-Derived Cell-Cell Communication Networks in the Tumor Microenvironment. Front Immunol 2022; 13:885267. [PMID: 35572582 PMCID: PMC9096838 DOI: 10.3389/fimmu.2022.885267] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 03/25/2022] [Indexed: 01/25/2023] Open
Abstract
Recent advances in single-cell technologies, particularly single-cell RNA-sequencing (scRNA-seq), have permitted high throughput transcriptional profiling of a wide variety of biological systems. As scRNA-seq supports inference of cell-cell communication, this technology has and continues to anchor groundbreaking studies into the efficacy and mechanism of novel immunotherapies for cancer treatment. In this review, we will highlight methods developed to infer inter- and intracellular signaling from scRNA-seq and discuss how they have contributed to studies of immunotherapeutic intervention in the tumor microenvironment (TME). However, a central challenge remains in validating the hypothesized cell-cell interactions. Therefore, this review will also cover strategies for integration of these scRNA-seq-derived interaction networks with existing experimental and computational approaches. Integration of these networks with imaging, protein secretion measurements, and network analysis and mathematical modeling tools addresses challenges that remain with scRNA-seq to enhance studies of immunosuppressive and immunotherapy-altered signaling in the TME.
Collapse
Affiliation(s)
- Kate Bridges
- Department of Biomedical Engineering, Yale University, New Haven, CT, United States
| | - Kathryn Miller-Jensen
- Department of Biomedical Engineering, Yale University, New Haven, CT, United States
- Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, CT, United States
- Systems Biology Institute, Yale University, New Haven, CT, United States
| |
Collapse
|
11
|
Progesterone and Inflammatory Response in the Oviduct during Physiological and Pathological Conditions. Cells 2022; 11:cells11071075. [PMID: 35406639 PMCID: PMC8997425 DOI: 10.3390/cells11071075] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 03/17/2022] [Accepted: 03/21/2022] [Indexed: 11/25/2022] Open
Abstract
Progesterone has been shown to be a potent suppressor of several inflammatory pathways. During pregnancy, progesterone levels increase, allowing for normal pregnancy establishment and maintenance. The dysregulation of progesterone, as well as inflammation, leads to poor pregnancy outcomes. However, it is unclear how progesterone imbalance could impact inflammatory responses in the oviduct and subsequently result in early pregnancy loss. Therefore, in this review, we describe the role of progesterone signaling in regulating the inflammatory response, with a focus on the oviduct and pathological conditions in the Fallopian tubes.
Collapse
|
12
|
AlAshqar A, Reschke L, Kirschen GW, Borahay MA. Role of inflammation in benign gynecologic disorders: from pathogenesis to novel therapies†. Biol Reprod 2021; 105:7-31. [PMID: 33739368 PMCID: PMC8256101 DOI: 10.1093/biolre/ioab054] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 03/15/2021] [Accepted: 03/17/2021] [Indexed: 12/16/2022] Open
Abstract
Emerging evidence supports the notion that inflammation fosters the development of common benign gynecologic disorders, including uterine leiomyoma, endometriosis, and adenomyosis. Numerous cytokines, chemokines, and growth and transcription factors have indisputable roles in the establishment and maintenance of benign gynecologic disorders by initiating complex cascades that promote proliferation, angiogenesis, and lesion progression. The interaction between inflammation and benign gynecologic disorders is orchestrated by a plethora of factors, including sex steroids, genetics, epigenetics, extracellular matrix, stem cells, cardiometabolic risk factors, diet, vitamin D, and the immune system. The role of inflammation in these disorders is not limited to local pathobiology but also extends to involve clinical sequelae that range from those confined to the reproductive tract, such as infertility and gynecologic malignancies, to systemic complications such as cardiovascular disease. Enhanced understanding of the intricate mechanisms of this association will introduce us to unvisited pathophysiological perspectives and guide future diagnostic and therapeutic implications aimed at reducing the burden of these disorders. Utilization of inflammatory markers, microRNA, and molecular imaging as diagnostic adjuncts may be valuable, noninvasive techniques for prompt detection of benign gynecologic disorders. Further, use of novel as well as previously established therapeutics, such as immunomodulators, hormonal treatments, cardiometabolic medications, and cyclooxygenase-2 and NF-κB inhibitors, can target inflammatory pathways involved in their pathogenesis. In this comprehensive review, we aim to dissect the existing literature on the role of inflammation in benign gynecologic disorders, including the proposed underlying mechanisms and complex interactions, its contribution to clinical sequelae, and the clinical implications this role entails.
Collapse
Affiliation(s)
- Abdelrahman AlAshqar
- Department of Gynecology and Obstetrics, Johns Hopkins University, Baltimore, MD, USA
- Department of Obstetrics and Gynecology, Kuwait University, Kuwait City, Kuwait
| | - Lauren Reschke
- Department of Gynecology and Obstetrics, Johns Hopkins University, Baltimore, MD, USA
| | - Gregory W Kirschen
- Department of Gynecology and Obstetrics, Johns Hopkins University, Baltimore, MD, USA
| | - Mostafa A Borahay
- Department of Gynecology and Obstetrics, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
13
|
Nisar S, Yousuf P, Masoodi T, Wani NA, Hashem S, Singh M, Sageena G, Mishra D, Kumar R, Haris M, Bhat AA, Macha MA. Chemokine-Cytokine Networks in the Head and Neck Tumor Microenvironment. Int J Mol Sci 2021; 22:ijms22094584. [PMID: 33925575 PMCID: PMC8123862 DOI: 10.3390/ijms22094584] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 04/03/2021] [Accepted: 04/05/2021] [Indexed: 02/07/2023] Open
Abstract
Head and neck squamous cell carcinomas (HNSCCs) are aggressive diseases with a dismal patient prognosis. Despite significant advances in treatment modalities, the five-year survival rate in patients with HNSCC has improved marginally and therefore warrants a comprehensive understanding of the HNSCC biology. Alterations in the cellular and non-cellular components of the HNSCC tumor micro-environment (TME) play a critical role in regulating many hallmarks of cancer development including evasion of apoptosis, activation of invasion, metastasis, angiogenesis, response to therapy, immune escape mechanisms, deregulation of energetics, and therefore the development of an overall aggressive HNSCC phenotype. Cytokines and chemokines are small secretory proteins produced by neoplastic or stromal cells, controlling complex and dynamic cell-cell interactions in the TME to regulate many cancer hallmarks. This review summarizes the current understanding of the complex cytokine/chemokine networks in the HNSCC TME, their role in activating diverse signaling pathways and promoting tumor progression, metastasis, and therapeutic resistance development.
Collapse
Affiliation(s)
- Sabah Nisar
- Molecular and Metabolic Imaging Laboratory, Cancer Research Department, Sidra Medicine, Doha 26999, Qatar; (S.N.); (S.H.); (M.H.)
| | - Parvaiz Yousuf
- Department of Zoology, School of Life Sciences, Central University of Kashmir, Ganderbal 191201, India;
| | - Tariq Masoodi
- Department of Genomic Medicine, Genetikode 400102, India;
| | - Nissar A. Wani
- Department of Biotechnology, School of Life Sciences, Central University of Kashmir, Ganderbal 191201, India;
| | - Sheema Hashem
- Molecular and Metabolic Imaging Laboratory, Cancer Research Department, Sidra Medicine, Doha 26999, Qatar; (S.N.); (S.H.); (M.H.)
| | - Mayank Singh
- Departmental of Medical Oncology, Dr. B. R. Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi 110029, India;
| | | | - Deepika Mishra
- Centre for Dental Education and Research, Department of Oral Pathology and Microbiology, All India Institute of Medical Sciences, New Delhi 110029, India;
| | - Rakesh Kumar
- Centre for Advanced Research, School of Biotechnology and Indian Council of Medical Research, Shri Mata Vaishno Devi University, Katra 182320, India;
| | - Mohammad Haris
- Molecular and Metabolic Imaging Laboratory, Cancer Research Department, Sidra Medicine, Doha 26999, Qatar; (S.N.); (S.H.); (M.H.)
- Laboratory Animal Research Center, Qatar University, Doha 2713, Qatar
| | - Ajaz A. Bhat
- Molecular and Metabolic Imaging Laboratory, Cancer Research Department, Sidra Medicine, Doha 26999, Qatar; (S.N.); (S.H.); (M.H.)
- Correspondence: (A.A.B.); or (M.A.M.); Tel.: +974-40037703 (A.A.B.); +91-8082326900 (M.A.M.)
| | - Muzafar A. Macha
- Watson-Crick Centre for Molecular Medicine, Islamic University of Science and Technology, Awantipora 192122, India
- Correspondence: (A.A.B.); or (M.A.M.); Tel.: +974-40037703 (A.A.B.); +91-8082326900 (M.A.M.)
| |
Collapse
|
14
|
Donninelli G, Studer V, Brambilla L, Zecca C, Peluso D, Laroni A, Michelis D, Mantegazza R, Confalonieri P, Volpe E. Immune Soluble Factors in the Cerebrospinal Fluid of Progressive Multiple Sclerosis Patients Segregate Into Two Groups. Front Immunol 2021; 12:633167. [PMID: 33777018 PMCID: PMC7988186 DOI: 10.3389/fimmu.2021.633167] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 02/02/2021] [Indexed: 11/16/2022] Open
Abstract
Primary-progressive (PP) and secondary-progressive (SP) multiple sclerosis (MS) are characterized by neurological deficits caused by a permanent neuronal damage, clinically quantified by the expanded disability status scale (EDSS). Neuronal tissue damage is also mediated by immune infiltrates producing soluble factors, such as cytokines and chemokines, which are released in the cerebrospinal fluid (CSF). The mechanisms regulating the production of a soluble factor are not completely defined. Using multiplex bead-based assays, we simultaneously measured 27 immune soluble factors in the CSF collected from 38 patients, 26 with PP-MS and 12 with SP-MS. Then, we performed a correlation matrix of all soluble factors expressed in the CSF. The CSF from patients with PP-MS and SP-MS had similar levels of cytokines and chemokines; however, the stratification of patients according to active or inactive magnetic resonance imaging (MRI) unveils some differences. Correlative studies between soluble factors in the CSF of patients with PP-MS and SP-MS revealed two clusters of immune mediators with pro-inflammatory functions, namely IFN-γ, MCP-1, MIP-1α, MIP-1β, IL-8, IP-10, and TNF-α (group 1), and anti-inflammatory functions, namely IL-9, IL-15, VEGF, and IL-1ra (group 2). However, most of the significant correlations between cytokines of group 1 and of group 2 were lost in patients with more severe disability (EDSS ≥ 4) compared to patients with mild to moderate disability (EDSS < 4). These results suggest a common regulation of cytokines and chemokines belonging to the same group and indicate that, in patients with more severe disability, the production of those factors is less coordinated, possibly due to advanced neurodegenerative mechanisms that interfere with the immune response.
Collapse
Affiliation(s)
- Gloria Donninelli
- Molecular Neuroimmunology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione Santa Lucia, Rome, Italy
| | - Valeria Studer
- Neuroimmunology and Neuromuscular Diseases Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Neurologico Carlo Besta, Milan, Italy.,Neurology Department, Martini Hospital, Turin, Italy
| | - Laura Brambilla
- Neuroimmunology and Neuromuscular Diseases Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Neurologico Carlo Besta, Milan, Italy
| | - Chiara Zecca
- Neurocenter of Southern Switzerland, Ospedale Regionale di Lugano, Lugano, Switzerland.,Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
| | - Daniele Peluso
- Bioinformatics e Biostatistics Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione Santa Lucia, Rome, Italy
| | - Alice Laroni
- Department of Neuroscience, Rehabilitation, Opthalmology, Genetics, Maternal and Child Health, University of Genova, Genova, Italy.,Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale Policlinico San Martino, Genova, Italy
| | - Daniele Michelis
- Department of Neuroscience, Rehabilitation, Opthalmology, Genetics, Maternal and Child Health, University of Genova, Genova, Italy
| | - Renato Mantegazza
- Neuroimmunology and Neuromuscular Diseases Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Neurologico Carlo Besta, Milan, Italy
| | - Paolo Confalonieri
- Neuroimmunology and Neuromuscular Diseases Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Neurologico Carlo Besta, Milan, Italy
| | - Elisabetta Volpe
- Molecular Neuroimmunology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione Santa Lucia, Rome, Italy
| |
Collapse
|
15
|
Olbei M, Hautefort I, Modos D, Treveil A, Poletti M, Gul L, Shannon-Lowe CD, Korcsmaros T. SARS-CoV-2 Causes a Different Cytokine Response Compared to Other Cytokine Storm-Causing Respiratory Viruses in Severely Ill Patients. Front Immunol 2021; 12:629193. [PMID: 33732251 PMCID: PMC7956943 DOI: 10.3389/fimmu.2021.629193] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 01/29/2021] [Indexed: 12/21/2022] Open
Abstract
Hyper-induction of pro-inflammatory cytokines, also known as a cytokine storm or cytokine release syndrome (CRS), is one of the key aspects of the currently ongoing SARS-CoV-2 pandemic. This process occurs when a large number of innate and adaptive immune cells activate and start producing pro-inflammatory cytokines, establishing an exacerbated feedback loop of inflammation. It is one of the factors contributing to the mortality observed with coronavirus 2019 (COVID-19) for a subgroup of patients. CRS is not unique to the SARS-CoV-2 infection; it was prevalent in most of the major human coronavirus and influenza A subtype outbreaks of the past two decades (H5N1, SARS-CoV, MERS-CoV, and H7N9). With a comprehensive literature search, we collected changing the cytokine levels from patients upon infection with the viral pathogens mentioned above. We analyzed published patient data to highlight the conserved and unique cytokine responses caused by these viruses. Our curation indicates that the cytokine response induced by SARS-CoV-2 is different compared to other CRS-causing respiratory viruses, as SARS-CoV-2 does not always induce specific cytokines like other coronaviruses or influenza do, such as IL-2, IL-10, IL-4, or IL-5. Comparing the collated cytokine responses caused by the analyzed viruses highlights a SARS-CoV-2-specific dysregulation of the type-I interferon (IFN) response and its downstream cytokine signatures. The map of responses gathered in this study could help specialists identify interventions that alleviate CRS in different diseases and evaluate whether they could be used in the COVID-19 cases.
Collapse
Affiliation(s)
- Marton Olbei
- Earlham Institute, Norwich, United Kingdom
- Gut Microbes and Health Programme, Quadram Institute Bioscience, Norwich, United Kingdom
| | | | - Dezso Modos
- Earlham Institute, Norwich, United Kingdom
- Gut Microbes and Health Programme, Quadram Institute Bioscience, Norwich, United Kingdom
| | - Agatha Treveil
- Earlham Institute, Norwich, United Kingdom
- Gut Microbes and Health Programme, Quadram Institute Bioscience, Norwich, United Kingdom
| | - Martina Poletti
- Earlham Institute, Norwich, United Kingdom
- Gut Microbes and Health Programme, Quadram Institute Bioscience, Norwich, United Kingdom
| | - Lejla Gul
- Earlham Institute, Norwich, United Kingdom
| | - Claire D. Shannon-Lowe
- Institute of Immunology and Immunotherapy, The University of Birmingham, Birmingham, United Kingdom
| | - Tamas Korcsmaros
- Earlham Institute, Norwich, United Kingdom
- Gut Microbes and Health Programme, Quadram Institute Bioscience, Norwich, United Kingdom
| |
Collapse
|
16
|
Kobayashi H, Kimura M, Maruyama S, Nagayasu M, Imanaka S. Revisiting estrogen-dependent signaling pathways in endometriosis: Potential targets for non-hormonal therapeutics. Eur J Obstet Gynecol Reprod Biol 2020; 258:103-110. [PMID: 33421806 DOI: 10.1016/j.ejogrb.2020.12.044] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 12/21/2020] [Indexed: 12/22/2022]
Abstract
Endometriosis is an estrogen-dependent gynecologic disease. Endometriotic cells survive in oxidative stress and hypoxic environments. The aim of this review is to reconsider new therapeutic strategies for endometriosis by focusing on estrogen signaling, ROS production and scavenging, and mitochondrial metabolism. Each keyword alone or in combination was used to search from PubMed and Embase by applying the filters of the title and the publication years between January 2000 and May 2020. Abnormal epigenetic marks of estrogen receptors (ERs) in endometriosis cause overexpression of ERβ, progesterone resistance, inflammation, anti-apoptosis, and mitochondrial metabolic modification. In addition to hormonal action, estrogen is involved in various functions such as mitochondrial biosynthesis and energy metabolism. Estrogen works with its downstream target genes to modulate mitochondrial gene expression, regulate ROS production, and affect mitochondrial biology, including ATP production, antioxidant defenses, mitochondrial biosynthesis, quality control, and energy-transducing capacity. Endometriosis can shift mitochondrial metabolism from oxidative phosphorylation to aerobic glycolysis. This metabolic conversion suppresses ROS production and thus activates the survival signal of endometriotic cells. Therefore, molecules associated with aerobic glycolysis and mitochondrial metabolism are considered therapeutic targets for endometriosis. In conclusion, estrogen downstream target genes involved in mitochondrial metabolic biosynthesis may be potential targets for non-hormonal treatment of endometriosis.
Collapse
Affiliation(s)
- Hiroshi Kobayashi
- Department of Obstetrics and Gynecology, Nara Medical University, Kashihara, Japan; Ms.Clinic MayOne, Kashihara, Japan.
| | - Mai Kimura
- Department of Obstetrics and Gynecology, Nara Medical University, Kashihara, Japan
| | - Sachiyo Maruyama
- Department of Obstetrics and Gynecology, Nara Medical University, Kashihara, Japan
| | - Mika Nagayasu
- Department of Obstetrics and Gynecology, Nara Medical University, Kashihara, Japan
| | - Shogo Imanaka
- Department of Obstetrics and Gynecology, Nara Medical University, Kashihara, Japan; Ms.Clinic MayOne, Kashihara, Japan
| |
Collapse
|
17
|
Krasnyi AM, Sadekova AA, Sefihanov TG, Vtorushina VV, Krechetova LV, Khilkevich EG, Arakelyan AS, Pavlovich SV. The Content of Cytokines IL-6, IL-8, TNF-α, IL-4 and the Level of CD86 and CD163 Expression in Peritoneal Fluid Macrophages Has a Reverse Correlation with the Degree of Severity of External Genital Endometriosis. BIOCHEMISTRY (MOSCOW), SUPPLEMENT SERIES B: BIOMEDICAL CHEMISTRY 2020. [DOI: 10.1134/s1990750820010096] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
18
|
Krasnyi AM, Sadekova AA, Sefihanov TG, Vtorushina VV, Krechetova EG, Khilkevich EG, Arakelyan AS, Pavlovich SV. [The content of cytokines IL-6, IL-8, TNF-α, IL-4 and the level of expression in macrophages CD86 and CD163 in peritoneal fluid has a reverse correlation with the degree of severity of external genital endometriosis]. BIOMEDIT︠S︡INSKAI︠A︡ KHIMII︠A︡ 2019; 65:432-436. [PMID: 31666417 DOI: 10.18097/pbmc20196505432] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Concentrations of eight different cytokines and the level of expression of CD86 and CD163 macrophages were studied in peritoneal fluid in women with endometriosis. It was found that the concentration of both inflammatory (IL-6, IL-8, TNF-α) and anti-inflammatory cytokines (IL-4) as well as the level of macrophage expression of the proinflammatory marker CD86 and anti-inflammatory marker CD163 increased in women with mild external genital endometriosis (1-2 stage), and did not differ from the control group in women with severe endometriosis (3-4 stage). The content of IL-2, IL-10, CM-CSF and IFN-γ in the peritoneal fluid of women with endometriosis did not differ significantly from the control group. The results of the study indicate that the development of external genital endometriosis may be based on insufficient both inflammatory and anti-inflammatory activity of macrophages in the peritoneal fluid.
Collapse
Affiliation(s)
- A M Krasnyi
- Academician Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow, Russia; Koltsov Institute of Developmental Biology, Moscow, Russia
| | - A A Sadekova
- Academician Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow, Russia
| | - T G Sefihanov
- Academician Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow, Russia
| | - V V Vtorushina
- Academician Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow, Russia
| | - E G Krechetova
- Academician Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow, Russia
| | - E G Khilkevich
- Academician Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow, Russia
| | - A S Arakelyan
- Academician Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow, Russia
| | - S V Pavlovich
- Academician Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow, Russia
| |
Collapse
|
19
|
Argilaguet J, Pedragosa M, Esteve-Codina A, Riera G, Vidal E, Peligero-Cruz C, Casella V, Andreu D, Kaisho T, Bocharov G, Ludewig B, Heath S, Meyerhans A. Systems analysis reveals complex biological processes during virus infection fate decisions. Genome Res 2019; 29:907-919. [PMID: 31138618 PMCID: PMC6581057 DOI: 10.1101/gr.241372.118] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 05/14/2019] [Indexed: 02/01/2023]
Abstract
The processes and mechanisms of virus infection fate decisions that are the result of a dynamic virus-immune system interaction with either an efficient effector response and virus elimination or an alleviated immune response and chronic infection are poorly understood. Here, we characterized the host response to acute and chronic lymphocytic choriomeningitis virus (LCMV) infections by gene coexpression network analysis of time-resolved splenic transcriptomes. First, we found an early attenuation of inflammatory monocyte/macrophage prior to the onset of T cell exhaustion, and second, a critical role of the XCL1-XCR1 communication axis during the functional adaptation of the T cell response to the chronic infection state. These findings not only reveal an important feedback mechanism that couples T cell exhaustion with the maintenance of a lower level of effector T cell response but also suggest therapy options to better control virus levels during the chronic infection phase.
Collapse
Affiliation(s)
- Jordi Argilaguet
- Infection Biology Laboratory, Department of Experimental and Health Sciences (DCEXS), Universitat Pompeu Fabra, Barcelona, Catalonia 08003, Spain
| | - Mireia Pedragosa
- Infection Biology Laboratory, Department of Experimental and Health Sciences (DCEXS), Universitat Pompeu Fabra, Barcelona, Catalonia 08003, Spain
| | - Anna Esteve-Codina
- CNAG-CRG, Center for Genomic Regulation (CRG), Barcelona Institute of Science and Technology, 08028 Barcelona, Spain.,Universitat Pompeu Fabra (UPF), Barcelona, Catalonia 08003, Spain
| | - Graciela Riera
- Infection Biology Laboratory, Department of Experimental and Health Sciences (DCEXS), Universitat Pompeu Fabra, Barcelona, Catalonia 08003, Spain
| | - Enric Vidal
- IRTA, Centre de Recerca en Sanitat Animal (CReSA-IRTA-UAB), Campus de la Universitat Autònoma de Barcelona, 08193 Bellaterra, Barcelona, Catalonia, Spain
| | - Cristina Peligero-Cruz
- Infection Biology Laboratory, Department of Experimental and Health Sciences (DCEXS), Universitat Pompeu Fabra, Barcelona, Catalonia 08003, Spain
| | - Valentina Casella
- Infection Biology Laboratory, Department of Experimental and Health Sciences (DCEXS), Universitat Pompeu Fabra, Barcelona, Catalonia 08003, Spain
| | - David Andreu
- Laboratory of Proteomics and Protein Chemistry, DCEXS, Universitat Pompeu Fabra, 08003 Barcelona, Spain
| | - Tsuneyasu Kaisho
- Department of Immunology, Institute of Advanced Medicine, Wakayama Medical University, Wakayama 641-8509, Japan.,Laboratory for Immune Regulation, World Premier International Research Center Initiative, Immunology Frontier Research Center, Osaka University, Osaka 565-0871, Japan
| | - Gennady Bocharov
- Marchuk Institute of Numerical Mathematics, Russian Academy of Sciences, Moscow, 119333, Russia.,Sechenov First Moscow State Medical University, Moscow, 119991, Russia
| | - Burkhard Ludewig
- Institute for Immunobiology, Kantonsspital St. Gallen, 9007 St. Gallen, Switzerland
| | - Simon Heath
- CNAG-CRG, Center for Genomic Regulation (CRG), Barcelona Institute of Science and Technology, 08028 Barcelona, Spain.,Universitat Pompeu Fabra (UPF), Barcelona, Catalonia 08003, Spain
| | - Andreas Meyerhans
- Infection Biology Laboratory, Department of Experimental and Health Sciences (DCEXS), Universitat Pompeu Fabra, Barcelona, Catalonia 08003, Spain.,Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, 08003, Spain
| |
Collapse
|
20
|
Inhibitory Effects of the Two Novel TSPO Ligands 2-Cl-MGV-1 and MGV-1 on LPS-induced Microglial Activation. Cells 2019; 8:cells8050486. [PMID: 31121852 PMCID: PMC6562711 DOI: 10.3390/cells8050486] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Revised: 05/16/2019] [Accepted: 05/17/2019] [Indexed: 02/06/2023] Open
Abstract
The 18 kDa translocator protein (TSPO) ligands 2-Cl-MGV-1 and MGV-1 can attenuate cell death of astrocyte-like cells (U118MG) and induce differentiation of neuronal progenitor cells (PC-12). Lipopolysaccharide (LPS) is a bacterial membrane endotoxin that activates cellular inflammatory pathways by releasing pro-inflammatory molecules, including cytokines and chemokines. The aim of the present study was to assess the immuno-modulatory effect of TSPO ligands in activated microglial cells. We demonstrated that the TSPO ligands 2-Cl-MGV-1 and MGV-1 can prevent LPS-induced activation of microglia (BV-2 cell line). Co-treatment of LPS (100 ng/mL) with these TSPO ligands (final concentration- 25 µM) reduces significantly the LPS-induced release of interleukin-6 (IL-6) from 16.9-fold to 2.5-fold, IL-β from 8.3-fold to 1.6-fold, interferon-γ from 16.0-fold to 2.2-fold, and tumor necrosis factor-α from 16.4-fold to 1.8-fold. This anti-inflammatory activity seems to be achieved by inhibition of NF-κB p65 activation. Assessment of initiation of ROS generation and cell metabolism shows significant protective effects of these two novel TSPO ligands. The IL-10 and IL-13 levels were not affected by any of the TSPO ligands. Thus, it appears that the ligands suppress the LPS-induced activation of some inflammatory responses of microglia. Such immunomodulatory effects may be relevant to the pharmacotherapy of neuro-inflammatory diseases.
Collapse
|
21
|
Tran HB, Lee YH, Guo JJ, Cheng TC. De novo transcriptome analysis of immune response on cobia (Rachycentron canadum) infected with Photobacterium damselae subsp. piscicida revealed inhibition of complement components and involvement of MyD88-independent pathway. FISH & SHELLFISH IMMUNOLOGY 2018; 77:120-130. [PMID: 29578048 DOI: 10.1016/j.fsi.2018.03.041] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 03/02/2018] [Accepted: 03/21/2018] [Indexed: 06/08/2023]
Abstract
Cobia, Rachycentron canadum, one of the most important aquatic species in Taiwan, has suffered heavy losses from Photobacterium damselae subsp. piscicida, which is the causal agent of photobacteriosis. In this study, the transcriptomic profiles of livers and spleens from Pdp-infected and non-infected cobia were obtained for the first time by Illumina-based paired-end sequencing method with a focus on immune-related genes. In total, 164,882 high quality unigenes were obtained in four libraries. Following Pdp infection, 7302 differentially expressed unigenes from liver and 8600 differentially expressed unigenes from spleen were identified. Twenty-seven of the differently expressed genes were further validated by RT-qPCR (average correlation coefficient 0.839, p-value <0.01). Results indicated a negative regulation of complement components and increased expression of genes involved in MyD88-independent pathway. Moreover, a remarkable finding was the increased expression of IL-10, implying an inadequacy of immune responses. This study not only characterized several putative immune pathways, but also provided a better understanding of the molecular responses to photobacteriosis in cobia.
Collapse
Affiliation(s)
- Hung Bao Tran
- Laboratory of Molecular Fish Immunology and Genetics, Department of Tropical Agriculture and International Cooperation, National Pingtung University of Science and Technology, Pingtung 91201, Taiwan; Research Center for Animal Biologics, National Pingtung University of Science and Technology, Pingtung 91201, Taiwan
| | - Yen-Hung Lee
- Tungkang Biotechnology Research Center, Fisheries Research Institute, Pingtung 92845, Taiwan
| | - Jiin-Ju Guo
- Tungkang Biotechnology Research Center, Fisheries Research Institute, Pingtung 92845, Taiwan
| | - Ta-Chih Cheng
- Laboratory of Molecular Fish Immunology and Genetics, Department of Tropical Agriculture and International Cooperation, National Pingtung University of Science and Technology, Pingtung 91201, Taiwan; Research Center for Animal Biologics, National Pingtung University of Science and Technology, Pingtung 91201, Taiwan.
| |
Collapse
|
22
|
The healthy donor profile of immunoregulatory soluble mediators is altered by stem cell mobilization and apheresis. Cytotherapy 2018; 20:740-754. [PMID: 29576502 DOI: 10.1016/j.jcyt.2018.02.366] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 02/08/2018] [Accepted: 02/11/2018] [Indexed: 11/23/2022]
Abstract
BACKGROUND Peripheral blood stem cells from healthy donors mobilized by granulocyte colony-stimulating factor (G-CSF) and thereafter harvested by leukapheresis are commonly used for allogeneic stem cell transplantation. METHODS Plasma levels of 38 soluble mediators (cytokines, soluble adhesion molecules, proteases, protease inhibitors) were analyzed in samples derived from healthy stem cell donors before G-CSF treatment and after 4 days, both immediately before and after leukapheresis. RESULTS Donors could be classified into two main subsets based on their plasma mediator profile before G-CSF treatment. Seventeen of 36 detectable mediators were significantly altered by G-CSF; generally an increase in mediator levels was seen, including pro-inflammatory cytokines, soluble adhesion molecules and proteases. Several leukocyte- and platelet-released mediators were increased during apheresis. Both plasma and graft mediator profiles were thus altered and showed correlations to graft concentrations of leukocytes and platelets; these concentrations were influenced by the apheresis device used. Finally, the mediator profile of the allotransplant recipients was altered by graft infusion, and based on their day +1 post-transplantation plasma profile our recipients could be divided into two major subsets that differed in overall survival. DISCUSSION G-CSF alters the short-term plasma mediator profile of healthy stem cell donors. These effects together with the leukocyte and platelet levels in the graft determine the mediator profile of the stem cell grafts. Graft infusion also alters the systemic mediator profile of the recipients, but further studies are required to clarify whether such graft-induced alterations have a prognostic impact.
Collapse
|
23
|
Zhang YL, Li B, Zhou ZH. A cross-sectional study: serum CCL3/MIP-1α levels may reflect lumbar intervertebral disk degeneration in Han Chinese people. J Pain Res 2018; 11:497-503. [PMID: 29551909 PMCID: PMC5842780 DOI: 10.2147/jpr.s152349] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Background The macrophage inflammatory protein-1α (MIP-1α), also named chemokine cytokine ligand 3 (CCL3), has been detected in nucleus pulposus and increased following cytokine stimulation. Objective The current study was performed to explore the relationship between serum CCL3/MIP-1α levels with lumbar intervertebral disk degeneration (IDD). Patients and methods A total of 132 disk degeneration patients confirmed by magnetic resonance imaging and 126 healthy controls were enrolled in the current study. Radiological evaluation of the IDD was conducted using a 3.0-T magnetic resonance imaging scanner for entire lumbar vertebra region. Degeneration of intervertebral disk was assessed by Schneiderman criteria. Serum CCL3/MIP-1α levels were investigated using a sandwich enzyme-linked immunosorbent assay. The Visual Analog Scale scores and Oswestry Disability Index index were recorded for clinical severity. Results Elevated concentrations of CCL3 in serum were found in IDD patients compared with asymptomatic volunteers. The case group included 49 IDD patients with grade 1, 42 with grade 2, and 41 with grade 3. Grade 3 and 2 had significantly higher CCL3 concentrations in serum compared with those with grade 1. The serum CCL3 levels were positively related to the degree of disk degeneration. In addition, the serum CCL3 levels also demonstrated a significant correlation with the clinical severity determined by Visual Analog Scale scores and Oswestry Disability Index index. Conclusion Serum CCL3 may serve as a biomarker of IDD.
Collapse
Affiliation(s)
- Yi-Li Zhang
- School of Public Health, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China.,School of Health Services Management, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China
| | - Bei Li
- School of Public Health, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China.,School of Health Services Management, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China
| | - Zeng-Huan Zhou
- School of Public Health, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China
| |
Collapse
|
24
|
Hofmann SR, Laass MW, Fehrs A, Schuppan D, Zevallos VF, Salminger D, Mäbert K, Hedrich CM. IL10 promoter haplotypes may contribute to altered cytokine expression and systemic inflammation in celiac disease. Clin Immunol 2018; 190:15-21. [PMID: 29481982 DOI: 10.1016/j.clim.2018.02.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 02/22/2018] [Accepted: 02/22/2018] [Indexed: 01/06/2023]
Abstract
Celiac disease (CD) is an autoimmune/inflammatory condition triggered by dietary gluten intake in genetically predisposed individuals. Though associations with MHC class II HLA-DQ2 or -DQ8 are the primary and necessary genetic predisposition for CD, >97% of genetically predisposed individuals never develop CD. Cytokines were measured in the serum of CD patients and controls. Possible associations with IL10 promoter variants were investigated. Cytokine expression from PBMCs was monitored in response to gluten exposure, or CD3/TCR complex stimulation in the absence or presence of recombinant IL-10. Serum cytokines varied between patients with CD at the time of diagnosis, after dietary elimination of gluten, and healthy controls. Serum IL-17A reflected disease activity. Reduced IL-10 serum levels and altered IL-10 expression by PBMCs coincided with IL10 promoter haplotypes that encode for "low" IL-10 expression (ATA). Increased prevalence of ATA IL10 promoter haplotypes and subsequently reduced IL-10 expression may be an immunological cofactor in individuals genetically predisposed for the development of CD. Resulting cytokine imbalances may be utilized as disease biomarkers in CD.
Collapse
Affiliation(s)
- S R Hofmann
- Department of Pediatrics, Universitätsklinikum Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - M W Laass
- Department of Pediatrics, Universitätsklinikum Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - A Fehrs
- Department of Pediatrics, Universitätsklinikum Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - D Schuppan
- Institute of Translational Immunology, University Medical Center, Johannes Gutenberg University, Mainz, Germany; Research Center for Immunotherapy, University Medical Center, Johannes Gutenberg University, Mainz, Germany; Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - V F Zevallos
- Institute of Translational Immunology, University Medical Center, Johannes Gutenberg University, Mainz, Germany; Research Center for Immunotherapy, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - D Salminger
- Department of Pediatrics, Universitätsklinikum Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - K Mäbert
- Department of Pediatrics, Universitätsklinikum Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - C M Hedrich
- Department of Pediatrics, Universitätsklinikum Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Department of Women's & Children's Health, Institute of Translational Medicine, University of Liverpool, Liverpool, UK; Department of Paediatric Rheumatology, Alder Hey Children's NHS Foundation Trust Hospital, Liverpool, UK.
| |
Collapse
|
25
|
Involvement of TWEAK and the NF-κB signaling pathway in lupus nephritis. Exp Ther Med 2018; 15:2611-2619. [PMID: 29456665 DOI: 10.3892/etm.2018.5711] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 08/28/2017] [Indexed: 11/05/2022] Open
Abstract
Previous findings have identified that tumor necrosis factor-related weak inducer of apoptosis (TWEAK) is associated with lupus nephritis (LN) activity status; however, the mechanism involved remains unclear. The present study aimed to investigate the roles of TWEAK and the nuclear factor (NF)-κB signaling pathway in LN. TWEAK levels in the blood and urine of patients with LN or non-LN systemic lupus erythematosus were measured by ELISA and compared with those in healthy controls. TWEAK expression and NF-κB transcriptional activity in the kidney were detected by western blotting, and Ki-67 and cluster of differentiation (CD) 68 expression were assessed using immunofluorescence. Additionally, human mesangial cells (HMCs) were cultured in vitro and divided into five groups: Normal control, TWEAK stimulus group, TWEAK + TWEAK blocking antibody, TWEAK + NF-κB inhibitor (BAY 11-7082) and TWEAK + combined (blocking antibody + BAY 11-7082). Cell cycle activity and Ki-67 expression in the HMCs were evaluated using flow cytometry, and cell induction of macrophage chemotaxis was determined by a Transwell assay. Levels of the inflammation-associated factors interleukin (IL)-6, monocyte chemotactic protein 1 (MCP-1), chemokine ligand 5 (CCL5), IL-8 and IL-10 were also detected by reverse transcription-quantitative polymerase chain reaction. It was observed that the urine levels of TWEAK in patients with LN were significantly elevated compared with those in the other groups (P<0.05). LN kidneys exhibited markedly increased cell proliferative ability, macrophage infiltration, TWEAK expression and NF-κB transcriptional activity compared with normal kidneys. Furthermore, the results indicated that treatment with recombinant TWEAK notably enhanced NF-κB transcriptional activity and significantly promoted cell proliferation and cell cycle activity (P<0.05), induced macrophage chemotaxis (P<0.05), significantly increased the expression of the chemotactic factors IL-6, IL-8, MCP-1 and CCL5 (P<0.05), and significantly reduced anti-inflammatory cytokine IL-10 mRNA expression in HMCs (P<0.05), relative to normal controls. Accordingly, blocking TWEAK function or inhibiting NF-κB activity reversed these effects. Collectively these data indicate that urine TWEAK may be considered as a novel biomarker of LN activity, and that blocking TWEAK function or NF-κB activity may effectively alleviate glomerular mesangial cell proliferation and macrophage chemotaxis.
Collapse
|
26
|
Yang HL, Zhou WJ, Chang KK, Mei J, Huang LQ, Wang MY, Meng Y, Ha SY, Li DJ, Li MQ. The crosstalk between endometrial stromal cells and macrophages impairs cytotoxicity of NK cells in endometriosis by secreting IL-10 and TGF-β. Reproduction 2017; 154:815-825. [PMID: 28971893 DOI: 10.1530/rep-17-0342] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 08/31/2017] [Accepted: 09/29/2017] [Indexed: 02/04/2023]
Abstract
The dysfunction of NK cells in women with endometriosis (EMS) contributes to the immune escape of menstrual endometrial fragments refluxed into the peritoneal cavity. The reciprocal communications between endometrial stromal cells (ESCs) and lymphocytes facilitate the development of EMS. However, the mechanism of these communications on cytotoxicity of natural killer (NK) cells in endometriotic milieus is still largely unknown. To imitate the local immune microenvironment, the co-culture systems of ESCs from patients with EMS and monocyte-derived macrophages or of ESCs, macrophages and NK cells were constructed. The cytokine levels in the co-culture unit were evaluated by ELISA. The expression of functional molecules in NK cells was detected by flow cytometry (FCM). The NK cell behaviors in vitro were analyzed by cell counting kit-8 and cytotoxic activation assays. After incubation with ESCs and macrophages, the expression of CD16, NKG2D, perforin and IFN-γ, viability and cytotoxicity of NK cells were significantly downregulated. The secretion of interleukin (IL)-1β, IL-10 and transforming growth factor (TGF)-β in the co-culture system of ESCs and macrophages was increased. Exposure with anti-IL-10 receptor β neutralizing antibody (αhIL-10Rβ) or αTGF-β could partly reverse these effects of ESCs and macrophages on NK cells in vitro These results suggest that the interaction between macrophages and ESCs downregulates cytotoxicity of NK cells possibly by stimulating the secretion of IL-10 and TGF-β, and may further trigger the immune escape of ectopic fragments and promote the occurrence and the development of EMS.
Collapse
Affiliation(s)
- Hui-Li Yang
- Laboratory for Reproductive ImmunologyHospital of Obstetrics and Gynecology, Fudan University, Shanghai, People's Republic of China.,Key Laboratory of Reproduction Regulation of NPFPCSIPPR, IRD, Fudan University, Shanghai, People's Republic of China
| | - Wen-Jie Zhou
- Laboratory for Reproductive ImmunologyHospital of Obstetrics and Gynecology, Fudan University, Shanghai, People's Republic of China.,Key Laboratory of Reproduction Regulation of NPFPCSIPPR, IRD, Fudan University, Shanghai, People's Republic of China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related DiseasesShanghai, People's Republic of China
| | - Kai-Kai Chang
- Laboratory for Reproductive ImmunologyHospital of Obstetrics and Gynecology, Fudan University, Shanghai, People's Republic of China
| | - Jie Mei
- Reproductive Medicine CenterDepartment of Obstetrics and Gynecology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medicine School, Nanjing, People's Republic of China
| | - Li-Qing Huang
- Department of Statistics and PsychologyCollege of Letters and Science, University of California Davis, Davis, California, USA
| | - Ming-Yan Wang
- Laboratory for Reproductive ImmunologyHospital of Obstetrics and Gynecology, Fudan University, Shanghai, People's Republic of China
| | - Yi Meng
- Laboratory for Reproductive ImmunologyHospital of Obstetrics and Gynecology, Fudan University, Shanghai, People's Republic of China
| | - Si-Yao Ha
- Laboratory for Reproductive ImmunologyHospital of Obstetrics and Gynecology, Fudan University, Shanghai, People's Republic of China
| | - Da-Jin Li
- Laboratory for Reproductive ImmunologyHospital of Obstetrics and Gynecology, Fudan University, Shanghai, People's Republic of China.,Key Laboratory of Reproduction Regulation of NPFPCSIPPR, IRD, Fudan University, Shanghai, People's Republic of China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related DiseasesShanghai, People's Republic of China
| | - Ming-Qing Li
- Laboratory for Reproductive ImmunologyHospital of Obstetrics and Gynecology, Fudan University, Shanghai, People's Republic of China .,Key Laboratory of Reproduction Regulation of NPFPCSIPPR, IRD, Fudan University, Shanghai, People's Republic of China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related DiseasesShanghai, People's Republic of China
| |
Collapse
|
27
|
Vaithilingam V, Evans MDM, Lewy DM, Bean PA, Bal S, Tuch BE. Co-encapsulation and co-transplantation of mesenchymal stem cells reduces pericapsular fibrosis and improves encapsulated islet survival and function when allografted. Sci Rep 2017; 7:10059. [PMID: 28855611 PMCID: PMC5577272 DOI: 10.1038/s41598-017-10359-1] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 08/09/2017] [Indexed: 12/22/2022] Open
Abstract
Pericapsular fibrotic overgrowth (PFO) is associated with poor survival of encapsulated islets. A strategy to combat PFO is the use of mesenchymal stem cells (MSC). MSC have anti-inflammatory properties and their potential can be enhanced by stimulation with proinflammatory cytokines. This study investigated whether co-encapsulation or co-transplantation of MSC with encapsulated islets would reduce PFO and improve graft survival. Stimulating MSC with a cytokine cocktail of IFN-γ and TNF-α enhanced their immunosuppressive potential by increasing nitric oxide production and secreting higher levels of immunomodulatory cytokines. In vitro, co-encapsulation with MSC did not affect islet viability but significantly enhanced glucose-induced insulin secretion. In vivo, normoglycemia was achieved in 100% mice receiving islets co-encapsulated with stimulated MSC as opposed to 71.4% receiving unstimulated MSC and only 9.1% receiving encapsulated islets alone. Microcapsules retrieved from both unstimulated and stimulated MSC groups had significantly less PFO with improved islet viability and function compared to encapsulated islets alone. Levels of peritoneal immunomodulatory cytokines IL-4, IL-6, IL-10 and G-CSF were significantly higher in MSC co-encapsulated groups. Similar results were obtained when encapsulated islets and MSC were co-transplanted. In summary, co-encapsulation or co-transplantation of MSC with encapsulated islets reduced PFO and improved the functional outcome of allotransplants.
Collapse
Affiliation(s)
- Vijayaganapathy Vaithilingam
- Biomedical Manufacturing Research Program, Commonwealth Scientific and Industrial Research Organization (CSIRO), Manufacturing Flagship, North Ryde, New South Wales, Australia.
| | - Margaret D M Evans
- Biomedical Manufacturing Research Program, Commonwealth Scientific and Industrial Research Organization (CSIRO), Manufacturing Flagship, North Ryde, New South Wales, Australia
| | - Denise M Lewy
- Biomedical Manufacturing Research Program, Commonwealth Scientific and Industrial Research Organization (CSIRO), Manufacturing Flagship, North Ryde, New South Wales, Australia
| | - Penelope A Bean
- Biomedical Manufacturing Research Program, Commonwealth Scientific and Industrial Research Organization (CSIRO), Manufacturing Flagship, North Ryde, New South Wales, Australia
| | - Sumeet Bal
- Biomedical Manufacturing Research Program, Commonwealth Scientific and Industrial Research Organization (CSIRO), Manufacturing Flagship, North Ryde, New South Wales, Australia
| | - Bernard E Tuch
- Biomedical Manufacturing Research Program, Commonwealth Scientific and Industrial Research Organization (CSIRO), Manufacturing Flagship, North Ryde, New South Wales, Australia
- Australian Foundation for Diabetes Research, Sydney, New South Wales, Australia, previously at CSIRO Manufacturing Flagship, North Ryde, New South Wales, Australia
| |
Collapse
|
28
|
Richter KR, Nasr AN, Mexas AM. Cytokine Concentrations Measured by Multiplex Assays in Canine Peripheral Blood Samples. Vet Pathol 2017; 55:53-67. [PMID: 28812527 DOI: 10.1177/0300985817725388] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Cytokines are known to play important roles in a wide range of pathologic conditions spanning all organ systems in every species studied. As our knowledge of the physiology of individual cytokines expands and our ability to measure multiple cytokines in smaller biological samples increases, we gain more insight into the significance and function of each cytokine and the importance of cytokine networks. Previous studies that reported measurements of cytokine concentrations from serum or plasma in dogs with infectious, autoimmune, metabolic, endocrine, and neoplastic diseases yield an appreciation for the complexity of cytokine control and potential applications for cytokine measurements in the diagnosis, prognosis, and therapy of a variety of disease conditions. In this review, we highlight the benefits of multiplex cytokine analysis, summarize clinical and experimental reports that have used this technology in dogs, and discuss the strengths and limitations of data analysis for the interpretation of results in these studies. We describe how differences in technical acuity, data reporting tactics, statistical analysis, study population selection criteria, and cross-sectional experimental design methods may affect interpretation of results from this technology. We also suggest methods for analysis in future studies, such as reporting median fluorescence intensity values, analyzing the proportion of patients above population medians, and performing longitudinal studies.
Collapse
Affiliation(s)
- Kristen R Richter
- 1 College of Veterinary Medicine, Midwestern University, Glendale, AZ, USA
| | - Amirah N Nasr
- 1 College of Veterinary Medicine, Midwestern University, Glendale, AZ, USA
| | - Angela M Mexas
- 1 College of Veterinary Medicine, Midwestern University, Glendale, AZ, USA
| |
Collapse
|
29
|
Shill DD, Polley KR, Willingham TB, Call JA, Murrow JR, McCully KK, Jenkins NT. Experimental intermittent ischemia augments exercise-induced inflammatory cytokine production. J Appl Physiol (1985) 2017; 123:434-441. [DOI: 10.1152/japplphysiol.01006.2016] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 05/16/2017] [Accepted: 05/30/2017] [Indexed: 12/13/2022] Open
Abstract
Acute exercise-induced inflammation is implicated in mediating the beneficial adaptations to regular exercise. Evidence suggests that reduced oxygen and/or blood flow to contracting muscle alters cytokine appearance. However, the acute inflammatory responses to hypoxic/ischemic exercise have been documented with inconsistent results and may not accurately reflect the ischemia produced during exercise in patients with ischemic cardiovascular diseases. Therefore, we determined the extent to which local inflammation is involved in the response to ischemic exercise. Fourteen healthy males performed unilateral isometric forearm contractions for 30 min with and without experimental ischemia. Blood was drawn at baseline, 5 and 10 min into exercise, at the end of exercise, and 30, 60, and 120 min after exercise. Oxygen saturation levels, as measured by near-infrared spectroscopy, were reduced by 10% and 41% during nonischemic and ischemic exercise, respectively. Nonischemic exercise did not affect cytokine values. Ischemia enhanced concentrations of basic fibroblast growth factor, interleukin (IL)-6, IL-10, tumor necrosis factor-alpha, and vascular endothelial growth factor during exercise, but IL-8 was not influenced by ischemic exercise. In conclusion, the present study demonstrates that ischemic, small-muscle endurance exercise elicits local inflammatory cytokine production compared with nonischemic exercise. NEW & NOTEWORTHY We demonstrate that ischemic, small-muscle endurance exercise elicits local inflammatory cytokine production compared with nonischemic exercise. The present study advances our knowledge of the inflammatory response to exercise in a partial ischemic state, which may be relevant for understanding the therapeutic effects of exercise training for people with ischemic cardiovascular disease-associated comorbidities.
Collapse
Affiliation(s)
- Daniel D. Shill
- Department of Kinesiology, University of Georgia, Athens, Georgia
| | - Kristine R. Polley
- Department of Foods and Nutrition, University of Georgia, Athens, Georgia
| | | | - Jarrod A. Call
- Department of Kinesiology, University of Georgia, Athens, Georgia
- Regenerative Bioscience Center, University of Georgia, Athens, Georgia; and
| | - Jonathan R. Murrow
- Department of Kinesiology, University of Georgia, Athens, Georgia
- Augusta University-University of Georgia Medical Partnership, Athens, Georgia
| | - Kevin K. McCully
- Department of Kinesiology, University of Georgia, Athens, Georgia
| | | |
Collapse
|
30
|
Poon K, Barson JR, Shi H, Chang GQ, Leibowitz SF. Involvement of the CXCL12 System in the Stimulatory Effects of Prenatal Exposure to High-Fat Diet on Hypothalamic Orexigenic Peptides and Behavior in Offspring. Front Behav Neurosci 2017; 11:91. [PMID: 28567007 PMCID: PMC5434113 DOI: 10.3389/fnbeh.2017.00091] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 04/28/2017] [Indexed: 01/09/2023] Open
Abstract
Exposure to a high fat diet (HFD) during gestation stimulates neurogenesis and expression of hypothalamic orexigenic neuropeptides that affect consummatory and emotional behaviors. With recent studies showing a HFD to increase inflammation, this report investigated the neuroinflammatory chemokine, CXCL12, and compared the effects of prenatal CXCL12 injection to those of prenatal HFD exposure, first, by testing whether the HFD affects circulating CXCL12 in the dam and the CXCL12 system in the offspring brain, and then by examining whether prenatal exposure to CXCL12 itself mimics the effects of a HFD on hypothalamic neuropeptides and emotional behaviors. Our results showed that prenatal exposure to a HFD significantly increased circulating levels of CXCL12 in the dam, and that daily injections of CXCL12 induced a similar increase in CXCL12 levels as the HFD. In addition, prenatal HFD exposure significantly increased the expression of CXCL12 and its receptors, CXCR4 and CXCR7, in the hypothalamic paraventricular nucleus (PVN) of the offspring. Finally, the results revealed strong similarities in the effects of prenatal HFD and CXCL12 administration, which both stimulated neurogenesis and enkephalin (ENK) expression in the PVN, while having inconsistent or no effect in other regions of the hypothalamus, and also increased anxiety as measured by several behavioral tests. These results focus attention specifically on the CXCL12 chemokine system in the PVN of the offspring as being possibly involved in the stimulatory effects of prenatal HFD exposure on ENK-expressing neurons in the PVN and their associated changes in emotional behavior.
Collapse
Affiliation(s)
- Kinning Poon
- Laboratory of Behavioral Neurobiology, Rockefeller UniversityNew York, NY, USA
| | - Jessica R Barson
- Laboratory of Behavioral Neurobiology, Rockefeller UniversityNew York, NY, USA.,Department of Neurobiology and Anatomy, Drexel University College of MedicinePhiladelphia, PA, USA
| | - Huanzhi Shi
- Laboratory of Behavioral Neurobiology, Rockefeller UniversityNew York, NY, USA
| | - Guo Qing Chang
- Laboratory of Behavioral Neurobiology, Rockefeller UniversityNew York, NY, USA
| | - Sarah F Leibowitz
- Laboratory of Behavioral Neurobiology, Rockefeller UniversityNew York, NY, USA
| |
Collapse
|
31
|
Andersson NW, Li Q, Mills CW, Ly J, Nomura Y, Chen J. Influence of prenatal maternal stress on umbilical cord blood cytokine levels. Arch Womens Ment Health 2016; 19:761-7. [PMID: 26846778 PMCID: PMC5032828 DOI: 10.1007/s00737-016-0607-7] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2015] [Accepted: 01/09/2016] [Indexed: 12/19/2022]
Abstract
PURPOSE Prenatal maternal stress (PNMS) is known to influence fetal programming and development. Thus far, the effects of PNMS on the developing immune system have mainly been documented in animal studies. This study aimed to examine the association between PNMS and immune cytokine profiles in the umbilical cord blood of newborn human infants. METHODS PNMS, including perceived stress, numbers of stressful life events experiences (both partner and health related), and state and trait anxiety, was assessed with five questionnaires and interviews from 43 pregnant women during the second trimester. Seven key cytokines important for immune function, i.e., IL-12, IL-1β, IL-4, IL-5, IL-6, IL-8, and TNF-α, were analyzed in cord blood by bead-based ELISA method (Luminex 200). Logistic regression was used to estimate the associations of PNMS scores and cytokine levels. RESULTS Increased levels of IL-1β, IL-4, IL-5, IL-6, and IL-8 were significantly associated with at least one of the maternal stress assessments, while the levels of IL-12 and TNF-α were not significantly associated with any of the PNMS measurements examined. CONCLUSION These preliminary findings suggest that PNMS may influence cytokine levels in newborn infants, in particular Th2-related cytokines. This report supports previous findings in animal studies and could suggest that newborns born to mothers with elevated PNMS have a predisposition to immune-related disorders.
Collapse
Affiliation(s)
- Niklas W. Andersson
- Department of Epidemiology Research, Statens Serum Institut, Artillerivej 5, 2300 Copenhagen S, Denmark, Sydney Medical School, Faculty of Medicine, University of Sydney, Concord Hospital, Sydney, Australia, Department of Psychology, Queens College, the City University of New York (CUNY), Flushing, New York, USA
| | - Qian Li
- Department of Preventive Medicine, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
| | | | - Jenny Ly
- Department of Psychology, Queens College, the City University of New York (CUNY), Flushing, New York, USA, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yoko Nomura
- Department of Psychology, Queens College, the City University of New York (CUNY), Flushing, New York, USA. .,Department of Preventive Medicine, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA. .,The Graduate Center, CUNY, New York, NY, USA. .,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Jia Chen
- Department of Preventive Medicine, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA, Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA, Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
32
|
Liu SQ, Ma XL, Qin G, Liu Q, Li YC, Wu YH. Trans-system mechanisms against ischemic myocardial injury. Compr Physiol 2015; 5:167-92. [PMID: 25589268 DOI: 10.1002/cphy.c140026] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
A mammalian organism possesses a hierarchy of naturally evolved protective mechanisms against ischemic myocardial injury at the molecular, cellular, and organ levels. These mechanisms comprise regional protective processes, including upregulation and secretion of paracrine cell-survival factors, inflammation, angiogenesis, fibrosis, and resident stem cell-based cardiomyocyte regeneration. There are also interactive protective processes between the injured heart, circulation, and selected remote organs, defined as trans-system protective mechanisms, including upregulation and secretion of endocrine cell-survival factors from the liver and adipose tissue as well as mobilization of bone marrow, splenic, and hepatic cells to the injury site to mediate myocardial protection and repair. The injured heart and activated remote organs exploit molecular and cellular processes, including signal transduction, gene expression, cell proliferation, differentiation, migration, mobilization, and/or extracellular matrix production, to establish protective mechanisms. Both regional and trans-system cardioprotective mechanisms are mediated by paracrine and endocrine messengers and act in coordination and synergy to maximize the protective effect, minimize myocardial infarction, and improve myocardial function, ensuring the survival and timely repair of the injured heart. The concept of the trans-system protective mechanisms may be generalized to other organ systems-injury in one organ may initiate regional as well as trans-system protective responses, thereby minimizing injury and ensuring the survival of the entire organism. Selected trans-system processes may serve as core protective mechanisms that can be exploited by selected organs in injury. These naturally evolved protective mechanisms are the foundation for developing protective strategies for myocardial infarction and injury-induced disorders in other organ systems.
Collapse
Affiliation(s)
- Shu Q Liu
- Biomedical Engineering Department, Northwestern University, Evanston, Illinois Department of Emergency Medicine, Thomas Jefferson University Hospitals, Philadelphia, Pennsylvania Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, Illinois Carbohydrate and Lipid Metabolism Research Laboratory, College of Life Science and Technology, Dalian University, Dalian, China Department of Medicine, Division of Biological Sciences, The University of Chicago, Chicago, Illinois
| | | | | | | | | | | |
Collapse
|
33
|
Chapman JC, Chapman FM, Michael SD. The production of alpha/beta and gamma/delta double negative (DN) T-cells and their role in the maintenance of pregnancy. Reprod Biol Endocrinol 2015; 13:73. [PMID: 26164866 PMCID: PMC4499209 DOI: 10.1186/s12958-015-0073-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Accepted: 07/08/2015] [Indexed: 01/19/2023] Open
Abstract
The ability of the thymus gland to convert bone marrow-derived progenitor cells into single positive (SP) T-cells is well known. In this review we present evidence that the thymus, in addition to producing SP T-cells, also has a pathway for the production of double negative (DN) T-cells. The existence of this pathway was noted during our examination of relevant literature to determine the cause of sex steroid-induced thymocyte loss. In conducting this search our objective was to answer the question of whether thymocyte loss is the end product of a typical interaction between the reproductive and immune systems, or evidence that the two systems are incompatible. We can now report that "thymocyte loss" is a normal process that occurs during the production of DN T-cells. The DN T-cell pathway is unique in that it is mediated by thymic mast cells, and becomes functional following puberty. Sex steroids initiate the development of the pathway by binding to an estrogen receptor alpha located in the outer membrane of the mast cells, causing their activation. This results in their uptake of extracellular calcium, and the production and subsequent release of histamine and serotonin. Lymphatic vessels, located in the subcapsular region of the thymus, respond to the two vasodilators by undergoing a substantial and preferential uptake of gamma/delta and alpha/beta DN T- cells. These T- cells exit the thymus via efferent lymphatic vessels and enter the lymphatic system.The DN pathway is responsible for the production of three subsets of gamma/delta DN T-cells and one subset of alpha/beta DN T-cells. In postpubertal animals approximately 35 % of total thymocytes exit the thymus as DN T-cells, regardless of sex. In pregnant females, their levels undergo a dramatic increase. Gamma/delta DN T-cells produce cytokines that are essential for the maintenance of pregnancy.
Collapse
Affiliation(s)
- John C Chapman
- Department of Biological Sciences, Binghamton University, Binghamton, New York, 13902-6000, USA.
| | - Fae M Chapman
- Department of Biological Sciences, Binghamton University, Binghamton, New York, 13902-6000, USA.
| | - Sandra D Michael
- Department of Biological Sciences, Binghamton University, Binghamton, New York, 13902-6000, USA.
| |
Collapse
|
34
|
Ahn SH, Monsanto SP, Miller C, Singh SS, Thomas R, Tayade C. Pathophysiology and Immune Dysfunction in Endometriosis. BIOMED RESEARCH INTERNATIONAL 2015; 2015:795976. [PMID: 26247027 PMCID: PMC4515278 DOI: 10.1155/2015/795976] [Citation(s) in RCA: 213] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Accepted: 11/18/2014] [Indexed: 12/30/2022]
Abstract
Endometriosis is an estrogen-dependent, chronic, proinflammatory disease prevalent in 10% of women of reproductive age worldwide. Characterized by the growth of endometrium-like tissue in aberrant locations outside of the uterus, it is responsible for symptoms including chronic pelvic pain, dysmenorrhea, and subfertility that degrade quality of life of women significantly. In Canada, direct and indirect economic cost of endometriosis amounts to 1.8 billion dollars, and this is elevated to 20 billion dollars in the United States. Despite decades of research, the etiology and pathophysiology of endometriosis still remain to be elucidated. This review aims to bring together the current understanding regarding the pathogenesis of endometriosis with specific focus on mechanisms behind vascularization of the lesions and the contribution of immune factors in facilitating lesion establishment and development. The role of hormones, immune cells, and cytokine signaling is highlighted, in addition to discussing the current pharmaceutical options available for management of pain symptoms in women with endometriosis.
Collapse
Affiliation(s)
- Soo Hyun Ahn
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada K7L 3N6
| | - Stephany P. Monsanto
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada K7L 3N6
| | - Caragh Miller
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada K7L 3N6
| | - Sukhbir S. Singh
- Department of Obstetrics and Gynecology, University of Ottawa, The Ottawa Hospital, ON, Canada K1H 7W9
| | - Richard Thomas
- Department of Obstetrics and Gynecology, Kingston General Hospital, Kingston, ON, Canada K7L 3N6
| | - Chandrakant Tayade
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada K7L 3N6
| |
Collapse
|
35
|
The potential of cytokines as safety biomarkers for drug-induced liver injury. Eur J Clin Pharmacol 2010; 66:961-76. [PMID: 20694460 DOI: 10.1007/s00228-010-0862-x] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2010] [Accepted: 06/23/2010] [Indexed: 12/12/2022]
|
36
|
Danesh A, Seneviratne C, Cameron CM, Banner D, Devries ME, Kelvin AA, Xu L, Ran L, Bosinger SE, Rowe T, Czub M, Jonsson CB, Cameron MJ, Kelvin DJ. Cloning, expression and characterization of ferret CXCL10. Mol Immunol 2007; 45:1288-97. [PMID: 18006061 PMCID: PMC5653245 DOI: 10.1016/j.molimm.2007.09.018] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2007] [Revised: 09/12/2007] [Accepted: 09/13/2007] [Indexed: 11/19/2022]
Abstract
Chemokines and their receptors function in the recruitment and activation of cells of the immune system to sites of inflammation. As such, chemokines play an important role in mediating pathophysiological events during microbial infection. In particular, CXCL9, CXCL10 and CXCL11 and their cognate receptor CXCR3 have been associated with the clinical course of several infectious diseases, including severe acute respiratory syndrome (SARS) and influenza. While CXCL9, CXCL10 and CXCL11 share the same receptor and have overlapping functions, each can also have unique activity in host defense. The lack of a preferred characterized animal model for SARS has brought our attention to ferrets, which have been used for years in influenza studies. The lack of immunological reagents for ferrets prompted us to clone CXCL9, CXCL10, CXCL11 and CXCR3 and, in the case of CXCL10, to express the gene as a recombinant protein. In this study we demonstrate that endogenous ferret CXCL10 exhibits similar mRNA expression patterns in the lungs of deceased SARS patients and ferrets experimentally infected with SARS coronavirus. This study therefore represents an important step towards development of the ferret as a model for the role of CXCL9, CXCL10 and CXCL11:CXCR3 axis in severe viral infections.
Collapse
Affiliation(s)
- Ali Danesh
- Division of Experimental Therapeutics, Toronto General Research Institute, University Health Network, 101 College Street, Toronto, Ontario, Canada M5G 1L7
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Charit Seneviratne
- Division of Experimental Therapeutics, Toronto General Research Institute, University Health Network, 101 College Street, Toronto, Ontario, Canada M5G 1L7
| | - Cheryl M. Cameron
- Division of Experimental Therapeutics, Toronto General Research Institute, University Health Network, 101 College Street, Toronto, Ontario, Canada M5G 1L7
| | - David Banner
- Division of Experimental Therapeutics, Toronto General Research Institute, University Health Network, 101 College Street, Toronto, Ontario, Canada M5G 1L7
| | - Mark E. Devries
- Division of Experimental Therapeutics, Toronto General Research Institute, University Health Network, 101 College Street, Toronto, Ontario, Canada M5G 1L7
| | - Alyson A. Kelvin
- Division of Experimental Therapeutics, Toronto General Research Institute, University Health Network, 101 College Street, Toronto, Ontario, Canada M5G 1L7
| | - Luoling Xu
- Division of Experimental Therapeutics, Toronto General Research Institute, University Health Network, 101 College Street, Toronto, Ontario, Canada M5G 1L7
| | - Longsi Ran
- Division of Experimental Therapeutics, Toronto General Research Institute, University Health Network, 101 College Street, Toronto, Ontario, Canada M5G 1L7
| | - Steven E. Bosinger
- Division of Experimental Therapeutics, Toronto General Research Institute, University Health Network, 101 College Street, Toronto, Ontario, Canada M5G 1L7
| | - Thomas Rowe
- Department of Biochemistry and Molecular Biology, Southern Research Institute, Birmingham, AL 35205, USA
| | - Marcus Czub
- National Microbiology Laboratory, Canadian Science Center for Human and Animal Health, 1015 Arlington Street, Winnipeg, Manitoba, Canada R3E 3R2
| | - Colleen B. Jonsson
- Department of Biochemistry and Molecular Biology, Southern Research Institute, Birmingham, AL 35205, USA
| | - Mark J. Cameron
- Division of Experimental Therapeutics, Toronto General Research Institute, University Health Network, 101 College Street, Toronto, Ontario, Canada M5G 1L7
| | - David J. Kelvin
- Division of Experimental Therapeutics, Toronto General Research Institute, University Health Network, 101 College Street, Toronto, Ontario, Canada M5G 1L7
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
- Department of Biochemistry and Molecular Biology, Southern Research Institute, Birmingham, AL 35205, USA
- Corresponding author at: Toronto General Research Institute, Division of Experimental Therapeutics, Toronto General Hospital, TMDT, 101 College Street, 3rd Floor, Room 913, Toronto, Ontario, Canada M5G 1L7. Tel.: +1 416 581 7608; fax: +1 416 581 7606.
| |
Collapse
|
37
|
Breathnach RM, Fanning S, Mulcahy G, Bassett HF, Jones BR, Daly P. Evaluation of Th1-like, Th2-like and immunomodulatory cytokine mRNA expression in the skin of dogs with immunomodulatory-responsive lymphocytic-plasmacytic pododermatitis. Vet Dermatol 2007; 17:313-21. [PMID: 16961816 DOI: 10.1111/j.1365-3164.2006.00534.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The term immunomodulatory-responsive lymphocytic-plasmacytic pododermatitis (ImR-LPP) has previously been proposed to denote a subpopulation of dogs with idiopathic pododermatitis. The objective of this study was to quantify the expression of mRNA encoding Th(1)-like [interferon (IFN)-gamma, interleukin (IL)-2 and IL-12], Th(2)-like [IL-4 and IL-6] and immunomodulatory cytokines [IL-10 and transforming growth factor (TGF)-beta] in lesional ImR-LPP, nonlesional ImR-LPP and healthy control pedal skin. Gene transcripts were quantified using TaqMan real-time reverse transcriptase-polymerase chain reaction assays. The skin of dogs with ImR-LPP had significant overexpression of IL-6 mRNA (P < 0.05) and significant underexpression of IL-12 mRNA (P < 0.01) compared to healthy controls. In addition, lesional ImR-LPP skin had significantly higher levels of IL-10 transcripts compared to healthy control pedal skin (P < 0.05). Although not attaining significance (P = 0.07), a trend towards reduced TGF-beta mRNA expression in lesional ImR-LPP skin was also evident. There were no significant differences in the levels of IFN-gamma or IL-2 mRNA transcripts among the three skin sample sources. IL-4 mRNA was detected in only one lesional sample. These results suggest that the pathogenesis of ImR-LPP may be associated with a T-cell-mediated inflammatory response characterized by impaired Th(1)-like, but enhanced Th(2)-like cytokine expression.
Collapse
Affiliation(s)
- Rory M Breathnach
- Department of Small Animal Clinical Studies, Faculty of Veterinary Medicine, University College Dublin, Dublin 4, Ireland.
| | | | | | | | | | | |
Collapse
|
38
|
Stefanovic L, Stefanovic B. Mechanism of direct hepatotoxic effect of KC chemokine: sequential activation of gene expression and progression from inflammation to necrosis. J Interferon Cytokine Res 2006; 26:760-70. [PMID: 17032170 DOI: 10.1089/jir.2006.26.760] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
Abstract
This work aimed to show that an important, yet unrecognized, role of KC chemokine in the liver is regulation of gene expression. KC expression in the liver stimulated three classes of genes in this temporal order: immediate-early genes, proinflammatory genes, and profibrotic genes. Transcription factors E2F5 and early growth response 1 (EGR1), Ca(2+) signaling molecules S100A8 and S100A9, and two oxidative stress-induced genes were identified as immediate-early genes of KC. Expression of these genes was stimulated at 3-5-fold increased KC concentrations. Expression of proinflammatory genes was activated 6 h after the immediateearly genes, and they included interleukin-1alpha (IL-1alpha) and IL-1beta. KC receptor gene CXCR2 was also upregulated, suggesting that KC may act through a positive feedback loop. Stimulation of expression of profibrotic genes, including type I collagen, was seen only after the proinflammatory genes were highly expressed for 12 h. KC is a potent regulator of gene expression that proceeds in a sequential manner. Immediate-early genes of KC stimulation were identified. The positive feedback regulation and an increased oxidative stress induced by KC may explain the poor prognosis in liver patients with elevated levels of CXC chemokines.
Collapse
Affiliation(s)
- Lela Stefanovic
- Department of Biomedical Science, College of Medicine, Florida State University, Tallahassee, FL 32306-4300, USA
| | | |
Collapse
|
39
|
Jha P, Sohn JH, Xu Q, Nishihori H, Wang Y, Nishihori S, Manickam B, Kaplan HJ, Bora PS, Bora NS. The complement system plays a critical role in the development of experimental autoimmune anterior uveitis. Invest Ophthalmol Vis Sci 2006; 47:1030-8. [PMID: 16505038 PMCID: PMC1975680 DOI: 10.1167/iovs.05-1062] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
PURPOSE The role of complement in ocular autoimmunity was explored in a experimental autoimmune anterior uveitis (EAAU) animal model. METHODS EAAU was induced in Lewis rats by immunization with bovine melanin-associated antigen. Complement activation in the eye was monitored by Western blot for iC3b. The importance of complement to the development of EAAU was studied by comparing the course of intraocular inflammation in normal Lewis rats (complement-sufficient) with cobra venom factor-treated rats (complement-depleted). Eyes were harvested from both complement-sufficient and complement-depleted rats for mRNA and protein analysis for IFN-gamma, IL-10, and interferon-inducible protein (IP)-10. Intracellular adhesion molecule (ICAM)-1 and leukocyte-endothelial cell adhesion molecule (LECAM)-1 were detected by immunofluorescent staining. OX-42 was used to investigate the importance of iC3b and CR3 interaction in EAAU. RESULTS There was a correlation between ocular complement activation and disease progression in EAAU. The incidence, duration, and severity of disease were dramatically reduced after active immunization in complement-depleted rats. Complement depletion also completely suppressed adoptive transfer EAAU. The presence of complement was critical for local production of cytokines (IFN-gamma and IL-10), chemokines (IP-10), and adhesion molecules (ICAM-1 and LECAM-1) during EAAU. Furthermore, intraocular complement activation, specifically iC3b production and engagement of complement receptor 3 (CR3), had a significant impact on disease activity in EAAU. CONCLUSIONS The study provided the novel finding that complement activation plays a central role in the pathogenesis of ocular autoimmunity and may serve as a potential target for therapeutic intervention.
Collapse
Affiliation(s)
- Purushottam Jha
- Department of Ophthalmology, Jones Eye Institute, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Jeong-Hyeon Sohn
- Department of Ophthalmology and Visual Sciences, Kentucky Lions Eye Center, University of Louisville, Louisville, Kentucky
| | - Qin Xu
- Department of Ophthalmology, Jones Eye Institute, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Hiroki Nishihori
- Department of Ophthalmology and Visual Sciences, Kentucky Lions Eye Center, University of Louisville, Louisville, Kentucky
| | - Yali Wang
- Department of Ophthalmology and Visual Sciences, Kentucky Lions Eye Center, University of Louisville, Louisville, Kentucky
| | - Saori Nishihori
- Department of Ophthalmology and Visual Sciences, Kentucky Lions Eye Center, University of Louisville, Louisville, Kentucky
| | - Balasubramanian Manickam
- Department of Ophthalmology, Jones Eye Institute, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Henry J. Kaplan
- Department of Ophthalmology and Visual Sciences, Kentucky Lions Eye Center, University of Louisville, Louisville, Kentucky
| | - Puran S. Bora
- Department of Ophthalmology, Jones Eye Institute, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Nalini S. Bora
- Department of Ophthalmology, Jones Eye Institute, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| |
Collapse
|
40
|
Stefanovic L, Brenner DA, Stefanovic B. Direct hepatotoxic effect of KC chemokine in the liver without infiltration of neutrophils. Exp Biol Med (Maywood) 2005; 230:573-86. [PMID: 16118408 DOI: 10.1177/153537020523000809] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
KC is a mouse homolog of human chemokine gro-alpha (CXCL1), expression of which is increased in liver diseases. We show that activated, but not quiescent, hepatic stellate cells (HSCs) express KC. Hepatic stellate cells constitutively express the KC receptor, CXCR2. Addition of recombinant KC to HSCs undergoing activation in culture increases secretion and processing of Type I collagen. Overexpression of endogenous KC in the mouse liver could be achieved by an intraperitoneal injection of CCl(4), followed after 24 hrs by an injection of recombinant KC into circulation. This protocol resulted in about a 14-fold increase in concentration of KC protein in the liver. Overexpression of KC was associated with upregulation of the mRNA for CXCR2 and MIP-2 and with necrosis and increased synthesis of Type I collagen. This suggests that KC has a direct hepatotoxic effect, which led to a massive liver necrosis after 48 hrs. No accumulation of neutrophils was seen in the livers as judged by histology and reverse transcriptase-polymerase chain reaction analysis of myeloperoxidase mRNA. Autostimulation of KC and CXCR2 expression by recombinant KC protein in the mice with preexisting liver injury indicates a positive feedback regulation. Such regulation and direct hepatotoxicity of KC with increased collagen synthesis represent novel findings about the role of KC/ gro-alpha in liver pathology.
Collapse
Affiliation(s)
- Lela Stefanovic
- Department of Biomedical Science, Florida State University College of Medicine, Tallahassee, FL 32306, USA
| | | | | |
Collapse
|
41
|
Abstract
A characteristic feature of all inflammatory disorders is the excessive recruitment of leukocytes to the site of inflammation. The loss of control in trafficking these cells contributes to inflammatory diseases. Leukocyte recruitment is a well-orchestrated process that includes several protein families including the large cytokine subfamily of chemotactic cytokines, the chemokines. Chemokines and their receptors are involved in the pathogenesis of several diseases. Acute lung injury that clinically manifests as acute respiratory distress syndrome (ARDS) is caused by an uncontrolled systemic inflammatory response resulting from clinical events including major surgery, trauma, multiple transfusions, severe burns, pancreatitis, and sepsis. Systemic inflammatory response syndrome involves activation of alveolar macrophages and sequestered neutrophils in the lung. The clinical hallmarks of ARDS are severe hypoxemia, diffuse bilateral pulmonary infiltrates, and normal intracardiac filling pressures. The magnitude and duration of the inflammatory process may ultimately determine the outcome in patients with ARDS. Recent evidence shows that activated leukocytes and chemokines play a key role in the pathogenesis of ARDS. The expanding number of antagonists of chemokine receptors for inflammatory disorders may hold promise for new medicines to combat ARDS.
Collapse
Affiliation(s)
- Padmam Puneet
- Dept. of Pharmacology, National University of Singapore, Singapore 117597
| | | | | |
Collapse
|
42
|
Wallace GR, John Curnow S, Wloka K, Salmon M, Murray PI. The role of chemokines and their receptors in ocular disease. Prog Retin Eye Res 2004; 23:435-48. [PMID: 15219876 DOI: 10.1016/j.preteyeres.2004.04.004] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
The migration and infiltration of cells into the eye whether blood-borne leucocytes, endothelial or epithelial cells occurs in many ocular diseases. Dysregulation of this process is apparent in chronic inflammation, corneal graft rejection, allergic eye disease and other sight-threatening conditions. Under normal and inflammatory conditions, chemokines and their receptors are important contributors to cell migration. To date, 47 chemokines and 19 chemokine receptors have been identified and characterised. In recent years, investigations into the role of chemokines and their receptors in ocular disease have generated an increasing number of publications. In the eye, the best understood action of these molecules has arisen from the study of their ability to control the infiltration of leucocytes in uveitis. However, the involvement of chemokines in angiogenesis in several ocular conditions and in the survival of corneal transplants demonstrates the multifaceted nature of their effects. Interestingly, the constitutive expression of chemokines and their receptors in ocular tissues suggests that certain chemokines have a homeostatic function. In this review, we discuss the nature and function of chemokines in health and disease, and describe the role of chemokines in the pathogenesis of different ocular conditions.
Collapse
Affiliation(s)
- Graham R Wallace
- The Birmingham and Midland Eye Centre, Sandwell and West Birmingham Hospitals NHS Trust, The University of Birmingham, City Hospital, Dudley Road, Birmingham B18 7QU, UK.
| | | | | | | | | |
Collapse
|
43
|
Chisholm D, Libet L, Hayashi T, Horner AA. Airway peptidoglycan and immunostimulatory DNA exposures have divergent effects on the development of airway allergen hypersensitivities. J Allergy Clin Immunol 2004; 113:448-54. [PMID: 15007346 DOI: 10.1016/j.jaci.2003.12.011] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Environmental exposures to toll-like receptor (TLR) ligands have been suggested to provide immunologic protection against allergic diseases. However, some TLRs use unique intracellular signaling pathways, suggesting that ambient TLR ligand exposures might induce a range of host responses. OBJECTIVE These investigations compared peptidoglycan (PGN; TLR2)-induced and immunostimulatory sequence DNA oligodeoxynucleotide (ISS-ODN; TLR9)-induced innate responses and determined how airway exposures to these TLR ligands affect adaptive immunity and the asthmatic phenotype. METHODS In in vitro and in vivo studies innate responses to PGN and ISS-ODN were compared. Alternatively, mice were intranasally immunized with ovalbumin (OVA) alone or OVA plus PGN or ISS-ODN, and adaptive immune profiles and responses to airway OVA challenge were assessed. RESULTS PGN and ISS-ODN induced divergent innate responses predictive of their having TH2- and TH1-biasing adjuvant potential, respectively. Consistent with these findings, mice intranasally immunized with OVA alone had relatively weak adaptive responses, whereas intranasal OVA/PGN- and OVA/ISS-ODN-coimmunized mice had much stronger humoral and cellular responses that were TH2 and TH1 biased, respectively. Finally, on airway allergen challenge, mice intranasally immunized with OVA alone had modest TH2-biased airway hypersensitivity responses, whereas airway responses were greatly exaggerated for intranasal OVA/PGN-immunized mice. In contrast, intranasal OVA/ISS-ODN-immunized mice had little evidence of airway hypersensitivity after airway allergen challenge. CONCLUSIONS Considered in a larger context, these results suggest that inspired air is likely to contain TLR ligands capable of both preventing and precipitating the asthmatic phenotype.
Collapse
Affiliation(s)
- Dugald Chisholm
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093-0663, USA
| | | | | | | |
Collapse
|