1
|
Garmany A, Arrell DK, Yamada S, Jeon R, Behfar A, Park S, Terzic A. Decoded cardiopoietic cell secretome linkage to heart repair biosignature. Stem Cells Transl Med 2024; 13:1144-1159. [PMID: 39259666 PMCID: PMC11555478 DOI: 10.1093/stcltm/szae067] [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: 04/09/2024] [Accepted: 08/03/2024] [Indexed: 09/13/2024] Open
Abstract
Cardiopoiesis-primed human stem cells exert sustained benefit in treating heart failure despite limited retention following myocardial delivery. To assess potential paracrine contribution, the secretome of cardiopoiesis conditioned versus naïve human mesenchymal stromal cells was decoded by directed proteomics augmented with machine learning and systems interrogation. Cardiopoiesis doubled cellular protein output generating a distinct secretome that segregated the conditioned state. Altering the expression of 1035 secreted proteins, cardiopoiesis reshaped the secretome across functional classes. The resolved differential cardiopoietic secretome was enriched in mesoderm development and cardiac progenitor signaling processes, yielding a cardiovasculogenic profile bolstered by upregulated cardiogenic proteins. In tandem, cardiopoiesis enhanced the secretion of immunomodulatory proteins associated with cytokine signaling, leukocyte migration, and chemotaxis. Network analysis integrated the differential secretome within an interactome of 1745 molecules featuring prioritized regenerative processes. Secretome contribution to the repair signature of cardiopoietic cell-treated infarcted hearts was assessed in a murine coronary ligation model. Intramyocardial delivery of cardiopoietic cells improved the performance of failing hearts, with undirected proteomics revealing 50 myocardial proteins responsive to cell therapy. Pathway analysis linked the secretome to cardiac proteome remodeling, pinpointing 17 cardiopoiesis-upregulated secretome proteins directly upstream of 44% of the cell therapy-responsive cardiac proteome. Knockout, in silico, of this 22-protein secretome-dependent myocardial ensemble eliminated indices of the repair signature. Accordingly, in vivo, cell therapy rendered the secretome-dependent myocardial proteome of an infarcted heart indiscernible from healthy counterparts. Thus, the secretagogue effect of cardiopoiesis transforms the human stem cell secretome, endows regenerative competency, and upregulates candidate paracrine effectors of cell therapy-mediated molecular restitution.
Collapse
Affiliation(s)
- Armin Garmany
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
- Center for Regenerative Biotherapeutics, Mayo Clinic, Rochester, MN, United States
- Marriott Heart Disease Research Program, Mayo Clinic, Rochester, MN, United States
- Mayo Clinic Alix School of Medicine, Regenerative Sciences Track, Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN, United States
| | - D Kent Arrell
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
- Center for Regenerative Biotherapeutics, Mayo Clinic, Rochester, MN, United States
- Marriott Heart Disease Research Program, Mayo Clinic, Rochester, MN, United States
| | - Satsuki Yamada
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
- Center for Regenerative Biotherapeutics, Mayo Clinic, Rochester, MN, United States
- Marriott Heart Disease Research Program, Mayo Clinic, Rochester, MN, United States
- Section of Geriatric Medicine & Gerontology, Department of Medicine, Mayo Clinic, Rochester, MN, United States
| | - Ryounghoon Jeon
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
- Center for Regenerative Biotherapeutics, Mayo Clinic, Rochester, MN, United States
- Marriott Heart Disease Research Program, Mayo Clinic, Rochester, MN, United States
| | - Atta Behfar
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
- Center for Regenerative Biotherapeutics, Mayo Clinic, Rochester, MN, United States
- Van Cleve Cardiac Regenerative Medicine Program, Mayo Clinic, Rochester, MN, United States
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, United States
| | - Sungjo Park
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
- Center for Regenerative Biotherapeutics, Mayo Clinic, Rochester, MN, United States
- Marriott Heart Disease Research Program, Mayo Clinic, Rochester, MN, United States
| | - Andre Terzic
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
- Center for Regenerative Biotherapeutics, Mayo Clinic, Rochester, MN, United States
- Marriott Heart Disease Research Program, Mayo Clinic, Rochester, MN, United States
- Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, Rochester, MN, United States
- Department of Medical Genetics, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
2
|
Perretta‐Tejedor N, Price KL, Jafree DJ, Pomeranz G, Kolatsi‐Joannou M, Martínez‐Salgado C, Long DA, Vasilopoulou E. Cardiotrophin-1 therapy reduces disease severity in a murine model of glomerular disease. Physiol Rep 2024; 12:e16129. [PMID: 38955668 PMCID: PMC11219243 DOI: 10.14814/phy2.16129] [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: 02/22/2024] [Revised: 06/10/2024] [Accepted: 06/20/2024] [Indexed: 07/04/2024] Open
Abstract
Cardiotrophin-1 (CT-1), a member of the interleukin (IL)-6 cytokine family, has renoprotective effects in mouse models of acute kidney disease and tubulointerstitial fibrosis, but its role in glomerular disease is unknown. To address this, we used the mouse model of nephrotoxic nephritis to test the hypothesis that CT-1 also has a protective role in immune-mediated glomerular disease. Using immunohistochemistry and analysis of single-cell RNA-sequencing data of isolated glomeruli, we demonstrate that CT-1 is expressed in the glomerulus in male mice, predominantly in parietal epithelial cells and is downregulated in mice with nephrotoxic nephritis. Furthermore, analysis of data from patients revealed that human glomerular disease is also associated with reduced glomerular CT-1 transcript levels. In male mice with nephrotoxic nephritis and established proteinuria, administration of CT-1 resulted in reduced albuminuria, prevented podocyte loss, and sustained plasma creatinine, compared with mice administered saline. CT-1 treatment also reduced fibrosis in the kidney cortex, peri-glomerular macrophage accumulation and the kidney levels of the pro-inflammatory mediator complement component 5a. In conclusion, CT-1 intervention therapy delays the progression of glomerular disease in mice by preserving kidney function and inhibiting renal inflammation and fibrosis.
Collapse
Affiliation(s)
- Nuria Perretta‐Tejedor
- Developmental Biology and Cancer Research and Teaching DepartmentUCL Great Ormond Street Institute of Child HealthLondonUK
- UCL Centre for Kidney and Bladder HealthLondonUK
- Department of Physiology and Pharmacology, Translational Research on Renal and Cardiovascular Diseases (TRECARD)University of Salamanca, Institute of Biomedical Research of Salamanca (IBSAL)SalamancaSpain
| | - Karen L. Price
- Developmental Biology and Cancer Research and Teaching DepartmentUCL Great Ormond Street Institute of Child HealthLondonUK
- UCL Centre for Kidney and Bladder HealthLondonUK
| | - Daniyal J. Jafree
- Developmental Biology and Cancer Research and Teaching DepartmentUCL Great Ormond Street Institute of Child HealthLondonUK
- UCL Centre for Kidney and Bladder HealthLondonUK
- Specialised Foundation Programme in ResearchNHS East of EnglandCambridgeUK
| | - Gideon Pomeranz
- Developmental Biology and Cancer Research and Teaching DepartmentUCL Great Ormond Street Institute of Child HealthLondonUK
- UCL Centre for Kidney and Bladder HealthLondonUK
| | - Maria Kolatsi‐Joannou
- Developmental Biology and Cancer Research and Teaching DepartmentUCL Great Ormond Street Institute of Child HealthLondonUK
- UCL Centre for Kidney and Bladder HealthLondonUK
| | - Carlos Martínez‐Salgado
- Department of Physiology and Pharmacology, Translational Research on Renal and Cardiovascular Diseases (TRECARD)University of Salamanca, Institute of Biomedical Research of Salamanca (IBSAL)SalamancaSpain
| | - David A. Long
- Developmental Biology and Cancer Research and Teaching DepartmentUCL Great Ormond Street Institute of Child HealthLondonUK
- UCL Centre for Kidney and Bladder HealthLondonUK
| | - Elisavet Vasilopoulou
- Developmental Biology and Cancer Research and Teaching DepartmentUCL Great Ormond Street Institute of Child HealthLondonUK
- UCL Centre for Kidney and Bladder HealthLondonUK
- Comparative Biomedical SciencesThe Royal Veterinary CollegeLondonUK
| |
Collapse
|
3
|
Zaporowska-Stachowiak I, Springer M, Stachowiak K, Oduah M, Sopata M, Wieczorowska-Tobis K, Bryl W. Interleukin-6 Family of Cytokines in Cancers. J Interferon Cytokine Res 2024; 44:45-59. [PMID: 38232478 DOI: 10.1089/jir.2023.0103] [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] [Indexed: 01/19/2024] Open
Abstract
Nine soluble ligands [interleukin-6 (IL-6), interleukin-11 (IL-11), leukemia inhibitory factor (LIF), oncostatin M (OSM), ciliary neurotrophic factor (CNTF), cardiotrophin-1 (CT-1), cardiotrophin-like cytokine, interleukin-27 (IL-27), and interleukin-31] share the ubiquitously expressed transmembrane protein-glycoprotein-130 beta-subunit (gp130) and thus form IL-6 family cytokines. Proteins that may be important for cancerogenesis, CT-1, IL-11, IL-27, LIF, OSM, and CNTF, belong to the superfamily of IL-6. Cytokines such as IL-6, IL-11, and IL-27 are better investigated in comparison with other members of the same family of cytokines, eg, CT-1. Gp130 is one of the main receptors through which these cytokines exert their effects. The clinical implication of understanding the pathways of these cytokines in oncology is that targeted therapy to inhibit or potentiate cytokine activity may lead to remission in some cases.
Collapse
Affiliation(s)
- Iwona Zaporowska-Stachowiak
- Department and Clinic of Palliative Medicine, Poznan University of Medical Sciences, Poznan, Poland
- Palliative Medicine In-Patient Unit, University Hospital of Lord's Transfiguration, Poznan University of Medical Sciences, Poznan, Poland
| | - Michał Springer
- Department of Internal Diseases, Metabolic Disorders and Arterial Hypertension, Poznan University of Medical Sciences, Poznan, Poland
| | | | - Mary Oduah
- English Students' Research Association, Poznan University of Medical Sciences, Poznan, Poland
| | - Maciej Sopata
- Department and Clinic of Palliative Medicine, Poznan University of Medical Sciences, Poznan, Poland
- Palliative Medicine In-Patient Unit, University Hospital of Lord's Transfiguration, Poznan University of Medical Sciences, Poznan, Poland
| | - Katarzyna Wieczorowska-Tobis
- Department and Clinic of Palliative Medicine, Poznan University of Medical Sciences, Poznan, Poland
- Palliative Medicine In-Patient Unit, University Hospital of Lord's Transfiguration, Poznan University of Medical Sciences, Poznan, Poland
| | - Wiesław Bryl
- Department of Internal Diseases, Metabolic Disorders and Arterial Hypertension, Poznan University of Medical Sciences, Poznan, Poland
| |
Collapse
|
4
|
Kale İ, Dizdar M. Investigation of maternal serum cardiotrophin-1 concentrations in pregnant women with preeclampsia; a prospective case-control study. J Matern Fetal Neonatal Med 2023; 36:2229931. [PMID: 37369375 DOI: 10.1080/14767058.2023.2229931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 05/25/2023] [Accepted: 06/21/2023] [Indexed: 06/29/2023]
Abstract
OBJECTIVE We aimed to investigate the cardiotrophin-1 (CT-1) concentrations in the serum of pregnant women with preeclampsia. METHODS This cross-sectional study was conducted with 88 pregnant women who applied to the Umraniye Training and Research Hospital Gynecology and Obstetrics Clinic between May 2022 and September 2022. The preeclampsia group consisted of 44 pregnant women diagnosed with preeclampsia, and the control group consisted of 44 healthy pregnant women matched with the preeclampsia group in terms of age and body mass index. Demographic characteristics, ultrasound and laboratory findings, perinatal outcomes, and maternal serum CT-1 concentrations were recorded. RESULTS Both groups were similar in terms of demographic features and the gestational week at blood sampling for CT-1. Preeclampsia and control groups were compared in terms of maternal serum CT-1 concentrations and no significant difference was found between the two groups (2061.4 pg/ml, 2168.5 pg/ml, respectively, p = .516). The preeclampsia group was divided into subgroups as mild and severe preeclampsia according to the severity of the disease and early-onset and late-onset preeclampsia according to the time of onset and compared with the control group in terms of maternal serum CT-1 concentration, no significant difference was found between the groups (p > .005, for all). CONCLUSION The serum CT-1 concentration of women whose pregnancy was complicated with preeclampsia was found to be similar to that of healthy controls. Although it has been shown in the literature that high serum CT-1 concentrations are associated with hypertensive heart diseases, its role in the pathophysiology of preeclampsia remains unclear.
Collapse
Affiliation(s)
- İbrahim Kale
- Department of Obstetrics and Gynecology, Umraniye Training and Research Hospital, Ümraniye/İstanbul, Türkiye
| | - Merve Dizdar
- Department of Obstetrics and Gynecology, Umraniye Training and Research Hospital, Ümraniye/İstanbul, Türkiye
| |
Collapse
|
5
|
Battista MR, Grigoletto A, Tedeschini T, Cellucci A, Colaceci F, Laufer R, Pasut G, Di Marco A. Efficacy of PEGylated ciliary neurotrophic factor superagonist variant in diet-induced obesity mice. PLoS One 2022; 17:e0265749. [PMID: 35316287 PMCID: PMC8939829 DOI: 10.1371/journal.pone.0265749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 03/07/2022] [Indexed: 11/18/2022] Open
Abstract
Ciliary neurotrophic factor (CNTF) is a neurotrophic cytokine able to induce appetite reduction, weight loss and antidiabetic effects. However, its susceptibility to neutralizing anti-CNTF antibodies in patients hampered its use for treatment of human obesity and diabetes. In addition, CNTF has a very short plasma half-life, which limits its use as a therapeutic agent. Solutions, directed to prolong its in vivo effects, vary from the implantation of encapsulated secreting cells to identification of more active variants or chemical modification of the protein itself. PEGylation is a widely used modification for shielding proteins from circulating antibodies and for increasing their plasma half-life. Here, we have selected DH-CNTF, a CNTF variant which has a 40-fold higher affinity for the CNTF receptor α accompanied by an increased activity in cellular assays. The PEGylated DH-CNTF retained the biological activity of native protein in vitro and showed a significant improvement of pharmacokinetic parameters. In an acute model of glucose tolerance, the PEG-DH-CNTF was able to reduce the glycemia in diet-induced obese animals, with a performance equaled by a 10-fold higher dose of DH-CNTF. In addition, the PEGylated DH-CNTF analog demonstrated a more potent weight loss effect than the unmodified protein, opening to the use of CNTF as weight reducing agent with treatment regimens that can better meet patient compliance thanks to reduced dosing schedules.
Collapse
Affiliation(s)
| | - Antonella Grigoletto
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Tommaso Tedeschini
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | | | | | | | - Gianfranco Pasut
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
- * E-mail: (ADM); (GP)
| | | |
Collapse
|
6
|
Jablonka S, Hennlein L, Sendtner M. Therapy development for spinal muscular atrophy: perspectives for muscular dystrophies and neurodegenerative disorders. Neurol Res Pract 2022; 4:2. [PMID: 34983696 PMCID: PMC8725368 DOI: 10.1186/s42466-021-00162-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 10/21/2021] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Major efforts have been made in the last decade to develop and improve therapies for proximal spinal muscular atrophy (SMA). The introduction of Nusinersen/Spinraza™ as an antisense oligonucleotide therapy, Onasemnogene abeparvovec/Zolgensma™ as an AAV9-based gene therapy and Risdiplam/Evrysdi™ as a small molecule modifier of pre-mRNA splicing have set new standards for interference with neurodegeneration. MAIN BODY Therapies for SMA are designed to interfere with the cellular basis of the disease by modifying pre-mRNA splicing and enhancing expression of the Survival Motor Neuron (SMN) protein, which is only expressed at low levels in this disorder. The corresponding strategies also can be applied to other disease mechanisms caused by loss of function or toxic gain of function mutations. The development of therapies for SMA was based on the use of cell culture systems and mouse models, as well as innovative clinical trials that included readouts that had originally been introduced and optimized in preclinical studies. This is summarized in the first part of this review. The second part discusses current developments and perspectives for amyotrophic lateral sclerosis, muscular dystrophies, Parkinson's and Alzheimer's disease, as well as the obstacles that need to be overcome to introduce RNA-based therapies and gene therapies for these disorders. CONCLUSION RNA-based therapies offer chances for therapy development of complex neurodegenerative disorders such as amyotrophic lateral sclerosis, muscular dystrophies, Parkinson's and Alzheimer's disease. The experiences made with these new drugs for SMA, and also the experiences in AAV gene therapies could help to broaden the spectrum of current approaches to interfere with pathophysiological mechanisms in neurodegeneration.
Collapse
Affiliation(s)
- Sibylle Jablonka
- Institute of Clinical Neurobiology, University Hospital of Wuerzburg, Versbacher Str. 5, 97078, Wuerzburg, Germany.
| | - Luisa Hennlein
- Institute of Clinical Neurobiology, University Hospital of Wuerzburg, Versbacher Str. 5, 97078, Wuerzburg, Germany
| | - Michael Sendtner
- Institute of Clinical Neurobiology, University Hospital of Wuerzburg, Versbacher Str. 5, 97078, Wuerzburg, Germany.
| |
Collapse
|
7
|
Ozgu-Erdinc AS, Gozukara I, Kahyaoglu S, Yilmaz S, Yumusak OH, Yilmaz N, Erkaya S, Engin-Ustun Y. Is there any role of interleukin-6 and high sensitive C-reactive protein in predicting IVF/ICSI success? A prospective cohort study. Horm Mol Biol Clin Investig 2021; 43:35-40. [PMID: 34837488 DOI: 10.1515/hmbci-2021-0039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 11/10/2021] [Indexed: 11/15/2022]
Abstract
OBJECTIVES Studies have established a relationship between proinflammatory factors and implantation failure in IVF/ICSI cycles. Likewise, low-grade chronic inflammation is generally blamed for predisposing infertility. In the present study, we aimed to find a relationship between serum IL-6 and hs-CRP levels and IVF/ICSI cycle outcomes. METHODS A total of 129 patients who consented to participate and attended the IVF unit of our department for the treatment of infertility have been enrolled in this prospective cohort study. Serum levels of high sensitive C-reactive protein and interleukin 6 have been detected at the beginning of the IVF/ICSI ovulation induction cycle. Cycle outcomes have been compared between patients with and without clinical pregnancy achievement following ART treatments. IVF/ICSI cycle outcomes of these two groups were also comparable except the number of >14 mm follicles, retrieved oocytes, metaphase II oocytes, and fertilized oocytes (2 pronuclei) which were in favor of the clinical pregnancy group. RESULTS Mean serum hs-CRP levels were 3.08 mg/L (0.12-35.04) and 2.28 mg/L (0.09-22.52) patients with and without clinical pregnancy respectively. Mean serum IL-6 levels were 2 pg/mL (1-10.2) and 2 pg/mL (1-76.9) patients with and without clinical pregnancy respectively. Both tests were found to be statistically insignificant in predicting the success of the ART cycle in terms of implantation, clinical pregnancy, miscarriage, and live birth. CONCLUSIONS In the present study, we have not found any significant effect of hs-CRP and IL-6 levels in the IVF cycle. However, in the light of this and previous studies, large-scale research may prove the exact influence of these markers on IVF success.
Collapse
Affiliation(s)
| | - Ilay Gozukara
- University of Health Sciences, Ankara City Hospital, Ankara, Turkey
| | - Serkan Kahyaoglu
- University of Health Sciences, Ankara City Hospital, Ankara, Turkey
| | - Saynur Yilmaz
- University of Health Sciences, Ankara City Hospital, Ankara, Turkey
| | | | - Nafiye Yilmaz
- University of Health Sciences, Ankara City Hospital, Ankara, Turkey
| | - Salim Erkaya
- University of Health Sciences, Ankara City Hospital, Ankara, Turkey
| | | |
Collapse
|
8
|
Influences of the IL-6 cytokine family on bone structure and function. Cytokine 2021; 146:155655. [PMID: 34332274 DOI: 10.1016/j.cyto.2021.155655] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 07/08/2021] [Accepted: 07/12/2021] [Indexed: 01/12/2023]
Abstract
The IL-6 family of cytokines comprises a large group of cytokines that all act via the formation of a signaling complex that includes the glycoprotein 130 (gp130) receptor. Despite this, many of these cytokines have unique roles that regulate the activity of bone forming osteoblasts, bone resorbing osteoclasts, bone-resident osteocytes, and cartilage cells (chondrocytes). These include specific functions in craniofacial development, longitudinal bone growth, and the maintenance of trabecular and cortical bone structure, and have been implicated in musculoskeletal pathologies such as craniosynostosis, osteoporosis, rheumatoid arthritis, osteoarthritis, and heterotopic ossifications. This review will work systematically through each member of this family and provide an overview and an update on the expression patterns and functions of each of these cytokines in the skeleton, as well as their negative feedback pathways, particularly suppressor of cytokine signaling 3 (SOCS3). The specific cytokines described are interleukin 6 (IL-6), interleukin 11 (IL-11), oncostatin M (OSM), leukemia inhibitory factor (LIF), cardiotrophin 1 (CT-1), ciliary neurotrophic factor (CNTF), cardiotrophin-like cytokine factor 1 (CLCF1), neuropoietin, humanin and interleukin 27 (IL-27).
Collapse
|
9
|
Abdelhaffez AS, Abd El-Aziz EA, Tohamy MB, Ahmed AM. N-acetyl cysteine can blunt metabolic and cardiovascular effects via down-regulation of cardiotrophin-1 in rat model of fructose-induced metabolic syndrome. Arch Physiol Biochem 2021:1-16. [PMID: 33507837 DOI: 10.1080/13813455.2021.1876735] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
In this study, we investigated the ability of N-acetyl cysteine (NAC) to alleviate the metabolic disorders in fructose-induced metabolic syndrome (MS) in male rats and to examine its protective effect on aortic and cardiac tissues via its influence on cardiotrophin-1 (CT-1) expression. NAC (20 mg/kg b.w./day) was administered to fructose induced MS animals for 12 weeks. Chronic fructose consumption (20% w/v) increased body weight gain, relative heart weight, systolic blood pressure (SBP), diastolic blood pressure (DBP), insulin resistance (IR), and associated with metabolic alterations. Histological and immunohistochemical examination revealed aortic stiffness and myocardial degeneration and fibrosis together with increased CT-1 expression. Treatment with NAC improved IR, SBP, DBP, and mitigated dyslipidaemia and oxidative stress. Additionally, NAC down-regulated CT-1 expression in the heart and aorta. These findings demonstrated the protective effect of NAC against aortic and myocardial degeneration and fibrosis through down-regulation of CT-1 in fructose induced MS animal model.
Collapse
Affiliation(s)
- Azza S Abdelhaffez
- Faculty of Medicine, Department of Medical Physiology, Assiut University, Assiut, Egypt
| | - Ebtihal A Abd El-Aziz
- Faculty of Medicine, Department of Medical Physiology, Assiut University, Assiut, Egypt
| | - Maha B Tohamy
- Faculty of Medicine, Department of Medical Physiology, Assiut University, Assiut, Egypt
| | - Asmaa M Ahmed
- Faculty of Medicine, Department of Pathology, Assiut University, Assiut, Egypt
| |
Collapse
|
10
|
Ritter K, Rousseau J, Hölscher C. The Role of gp130 Cytokines in Tuberculosis. Cells 2020; 9:E2695. [PMID: 33334075 PMCID: PMC7765486 DOI: 10.3390/cells9122695] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 12/01/2020] [Accepted: 12/10/2020] [Indexed: 12/13/2022] Open
Abstract
Protective immune responses to Mycobacterium tuberculosis (Mtb) infection substantially depend on a delicate balance within cytokine networks. Thus, immunosuppressive therapy by cytokine blockers, as successfully used in the management of various chronic inflammatory diseases, is often connected with an increased risk for tuberculosis (TB) reactivation. Hence, identification of alternative therapeutics which allow the treatment of inflammatory diseases without compromising anti-mycobacterial immunity remains an important issue. On the other hand, in the context of novel therapeutic approaches for the management of TB, host-directed adjunct therapies, which combine administration of antibiotics with immunomodulatory drugs, play an increasingly important role, particularly to reduce the duration of treatment. In both respects, cytokines/cytokine receptors related to the common receptor subunit gp130 may serve as promising target candidates. Within the gp130 cytokine family, interleukin (IL)-6, IL-11 and IL-27 are most explored in the context of TB. This review summarizes the differential roles of these cytokines in protection and immunopathology during Mtb infection and discusses potential therapeutic implementations with respect to the aforementioned approaches.
Collapse
Affiliation(s)
- Kristina Ritter
- Infection Immunology, Research Centre Borstel, D-23845 Borstel, Germany; (K.R.); (J.R.)
| | - Jasmin Rousseau
- Infection Immunology, Research Centre Borstel, D-23845 Borstel, Germany; (K.R.); (J.R.)
| | - Christoph Hölscher
- Infection Immunology, Research Centre Borstel, D-23845 Borstel, Germany; (K.R.); (J.R.)
- German Centre for Infection Research (DZIF), Partner Site Hamburg-Borstel-Lübeck-Riems, D-23845 Borstel, Germany
| |
Collapse
|
11
|
Berezin AE, Berezin AA. Circulating Cardiac Biomarkers in Diabetes Mellitus: A New Dawn for Risk Stratification-A Narrative Review. Diabetes Ther 2020; 11:1271-1291. [PMID: 32430864 PMCID: PMC7261294 DOI: 10.1007/s13300-020-00835-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Indexed: 02/06/2023] Open
Abstract
The aim of this narrative review is to update the current knowledge on the differential choice of circulating cardiac biomarkers in patients with prediabetes and established type 2 diabetes mellitus (T2DM). There are numerous circulating biomarkers with unconfirmed abilities to predict clinical outcomes in pre-DM and DM individuals; the prognostication ability of the cardiac biomarkers reported here has been established, and they are still being studied. The conventional cardiac biomarkers, such as natriuretic peptides (NPs), soluble suppressor tumorigenisity-2, high-sensitivity circulating cardiac troponins and galectin-3, were useful to ascertain cardiovascular (CV) risk. Each cardiac biomarker has its strengths and weaknesses that affect the price of usage, specificity, sensitivity, predictive value and superiority in face-to-face comparisons. Additionally, there have been confusing reports regarding their abilities to be predictably relevant among patients without known CV disease. The large spectrum of promising cardiac biomarkers (growth/differential factor-15, heart-type fatty acid-binding protein, cardiotrophin-1, carboxy-terminal telopeptide of collagen type 1, apelin and non-coding RNAs) is discussed in the context of predicting CV diseases and events in patients with known prediabetes and T2DM. Various reasons have been critically discussed related to the variable findings regarding biomarker-based prediction of CV risk among patients with metabolic disease. It was found that NPs and hs-cTnT are still the most important tools that have an affordable price as well as high sensitivity and specificity to predict clinical outcomes among patients with pre-DM and DM in routine clinical practice, but other circulating biomarkers need to be carefully investigated in large trials in the future.
Collapse
Affiliation(s)
- Alexander E Berezin
- Internal Medicine Department, Ministry of Health of Ukraine, State Medical University, Zaporozhye, 69035, Ukraine.
| | - Alexander A Berezin
- Internal Medicine Department, Medical Academy of Post-Graduate Education, Ministry of Health of Ukraine, Zaporozhye, 69096, Ukraine
| |
Collapse
|
12
|
Omokehinde T, Johnson RW. GP130 Cytokines in Breast Cancer and Bone. Cancers (Basel) 2020; 12:cancers12020326. [PMID: 32023849 PMCID: PMC7072680 DOI: 10.3390/cancers12020326] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Revised: 01/24/2020] [Accepted: 01/29/2020] [Indexed: 12/14/2022] Open
Abstract
Breast cancer cells have a high predilection for skeletal homing, where they may either induce osteolytic bone destruction or enter a latency period in which they remain quiescent. Breast cancer cells produce and encounter autocrine and paracrine cytokine signals in the bone microenvironment, which can influence their behavior in multiple ways. For example, these signals can promote the survival and dormancy of bone-disseminated cancer cells or stimulate proliferation. The interleukin-6 (IL-6) cytokine family, defined by its use of the glycoprotein 130 (gp130) co-receptor, includes interleukin-11 (IL-11), leukemia inhibitory factor (LIF), oncostatin M (OSM), ciliary neurotrophic factor (CNTF), and cardiotrophin-1 (CT-1), among others. These cytokines are known to have overlapping pleiotropic functions in different cell types and are important for cross-talk between bone-resident cells. IL-6 cytokines have also been implicated in the progression and metastasis of breast, prostate, lung, and cervical cancer, highlighting the importance of these cytokines in the tumor–bone microenvironment. This review will describe the role of these cytokines in skeletal remodeling and cancer progression both within and outside of the bone microenvironment.
Collapse
Affiliation(s)
- Tolu Omokehinde
- Program in Cancer Biology, Vanderbilt University, Nashville, TN 37232, USA
- Vanderbilt Center for Bone Biology, Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Rachelle W. Johnson
- Program in Cancer Biology, Vanderbilt University, Nashville, TN 37232, USA
- Vanderbilt Center for Bone Biology, Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Correspondence: ; Tel.: +1-615-875-8965
| |
Collapse
|
13
|
Adipose Tissue-Derived Signatures for Obesity and Type 2 Diabetes: Adipokines, Batokines and MicroRNAs. J Clin Med 2019; 8:jcm8060854. [PMID: 31208019 PMCID: PMC6617388 DOI: 10.3390/jcm8060854] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 06/07/2019] [Accepted: 06/11/2019] [Indexed: 12/13/2022] Open
Abstract
: Obesity is one of the main risk factors for type 2 diabetes mellitus (T2DM). It is closely related to metabolic disturbances in the adipose tissue that primarily functions as a fat reservoir. For this reason, adipose tissue is considered as the primary site for initiation and aggravation of obesity and T2DM. As a key endocrine organ, the adipose tissue communicates with other organs, such as the brain, liver, muscle, and pancreas, for the maintenance of energy homeostasis. Two different types of adipose tissues-the white adipose tissue (WAT) and brown adipose tissue (BAT)-secrete bioactive peptides and proteins, known as "adipokines" and "batokines," respectively. Some of them have beneficial anti-inflammatory effects, while others have harmful inflammatory effects. Recently, "exosomal microRNAs (miRNAs)" were identified as novel adipokines, as adipose tissue-derived exosomal miRNAs can affect other organs. In the present review, we discuss the role of adipose-derived secretory factors-adipokines, batokines, and exosomal miRNA-in obesity and T2DM. It will provide new insights into the pathophysiological mechanisms involved in disturbances of adipose-derived factors and will support the development of adipose-derived factors as potential therapeutic targets for obesity and T2DM.
Collapse
|
14
|
Duraikannu A, Krishnan A, Chandrasekhar A, Zochodne DW. Beyond Trophic Factors: Exploiting the Intrinsic Regenerative Properties of Adult Neurons. Front Cell Neurosci 2019; 13:128. [PMID: 31024258 PMCID: PMC6460947 DOI: 10.3389/fncel.2019.00128] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 03/14/2019] [Indexed: 01/19/2023] Open
Abstract
Injuries and diseases of the peripheral nervous system (PNS) are common but frequently irreversible. It is often but mistakenly assumed that peripheral neuron regeneration is robust without a need to be improved or supported. However, axonal lesions, especially those involving proximal nerves rarely recover fully and injuries generally are complicated by slow and incomplete regeneration. Strategies to enhance the intrinsic growth properties of reluctant adult neurons offer an alternative approach to consider during regeneration. Since axons rarely regrow without an intimately partnered Schwann cell (SC), approaches to enhance SC plasticity carry along benefits to their axon partners. Direct targeting of molecules that inhibit growth cone plasticity can inform important regenerative strategies. A newer approach, a focus of our laboratory, exploits tumor suppressor molecules that normally dampen unconstrained growth. However several are also prominently expressed in stable adult neurons. During regeneration their ongoing expression “brakes” growth, whereas their inhibition and knockdown may enhance regrowth. Examples have included phosphatase and tensin homolog deleted on chromosome ten (PTEN), a tumor suppressor that inhibits PI3K/pAkt signaling, Rb1, the protein involved in retinoblastoma development, and adenomatous polyposis coli (APC), a tumor suppressor that inhibits β-Catenin transcriptional signaling and its translocation to the nucleus. The identification of several new targets to manipulate the plasticity of regenerating adult peripheral neurons is exciting. How they fit with canonical regeneration strategies and their feasibility require additional work. Newer forms of nonviral siRNA delivery may be approaches for molecular manipulation to improve regeneration.
Collapse
Affiliation(s)
- Arul Duraikannu
- Division of Neurology, Department of Medicine, and Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| | - Anand Krishnan
- Division of Neurology, Department of Medicine, and Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| | - Ambika Chandrasekhar
- Division of Neurology, Department of Medicine, and Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| | - Douglas W Zochodne
- Division of Neurology, Department of Medicine, and Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
15
|
Yildirim G, Ozcan KM, Keskin O, Tekeli F, Kaymaz AA. Effects of chronic REM sleep deprivation on lipocalin-2, nitric oxide synthase-3, interleukin-6 and cardiotrophin-1 levels: an experimental rat model. Sleep Biol Rhythms 2019. [DOI: 10.1007/s41105-019-00214-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
16
|
Lokau J, Garbers C. Activating mutations of the gp130/JAK/STAT pathway in human diseases. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2018; 116:283-309. [PMID: 31036294 DOI: 10.1016/bs.apcsb.2018.11.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Cytokines of the interleukin-6 (IL-6) family are involved in numerous physiological and pathophysiological processes. Dysregulated and increased activities of its members can be found in practically all human inflammatory diseases including cancer. All cytokines activate several intracellular signaling cascades, including the Jak/STAT, MAPK, PI3K, and Src/YAP signaling pathways. Additionally, several mutations in proteins involved in these signaling cascades have been identified in human patients, which render these proteins constitutively active and result in a hyperactivation of the signaling pathway. Interestingly, some of these mutations are associated with or even causative for distinct human diseases, making them interesting targets for therapy. This chapter describes the basic biology of the gp130/Jak/STAT pathway, summarizes what is known about the molecular mechanisms of the activating mutations, and gives an outlook how this knowledge can be exploited for targeted therapy in human diseases.
Collapse
Affiliation(s)
- Juliane Lokau
- Department of Pathology, Otto-von-Guericke-University Magdeburg, Medical Faculty, Magdeburg, Germany
| | - Christoph Garbers
- Department of Pathology, Otto-von-Guericke-University Magdeburg, Medical Faculty, Magdeburg, Germany.
| |
Collapse
|
17
|
Adrian-Segarra JM, Sreenivasan K, Gajawada P, Lörchner H, Braun T, Pöling J. The AB loop of oncostatin M (OSM) determines species-specific signaling in humans and mice. J Biol Chem 2018; 293:20181-20199. [PMID: 30373773 DOI: 10.1074/jbc.ra118.004375] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 10/12/2018] [Indexed: 11/06/2022] Open
Abstract
The pleiotropic interleukin-6 (IL-6)-type cytokine oncostatin M (OSM) signals in multiple cell types, affecting processes such as cell differentiation, hematopoiesis, and inflammation. In humans, OSM exerts its effects through activation of either of two different heterodimeric receptor complexes, formed by glycoprotein 130 (gp130) and either OSM receptor (OSMR) or leukemia inhibitory factor receptor (LIFR). In contrast, the mouse OSM orthologue acts mainly through dimers containing OSMR and gp130 and shows limited activity through mouse LIFR. Despite their structural similarity, neither human nor mouse OSM signal through the other species' OSMR. The molecular basis for such species-specific signaling, however, remains poorly understood. To identify key molecular features of OSM that determine receptor activation in humans and mice, we generated chimeric mouse-human cytokines. Replacing regions within binding site III of murine OSM with the human equivalents showed that the cytokine's AB loop was critical for receptor selection. Substitutions of individual amino acids within this region demonstrated that residues Asn-37, Thr-40, and Asp-42 of the murine cytokine were responsible for limited LIFR activation and absence of human OSMR/LIFR signaling. In human OSM, Lys-44 appeared to be the main residue preventing mouse OSMR activation. Our data reveal that individual amino acids within the AB loop of OSM determine species-specific activities. These mutations might reflect a key step in the evolutionary process of this cytokine, in which receptor promiscuity gives way to ligand-receptor specialization.
Collapse
Affiliation(s)
- Juan M Adrian-Segarra
- From the Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Ludwigstrasse 43, D-61231 Bad Nauheim, Germany and
| | - Krishnamoorthy Sreenivasan
- From the Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Ludwigstrasse 43, D-61231 Bad Nauheim, Germany and
| | - Praveen Gajawada
- From the Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Ludwigstrasse 43, D-61231 Bad Nauheim, Germany and
| | - Holger Lörchner
- From the Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Ludwigstrasse 43, D-61231 Bad Nauheim, Germany and; the German Centre for Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, Germany
| | - Thomas Braun
- From the Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Ludwigstrasse 43, D-61231 Bad Nauheim, Germany and; the German Centre for Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, Germany.
| | - Jochen Pöling
- From the Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Ludwigstrasse 43, D-61231 Bad Nauheim, Germany and; the German Centre for Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, Germany.
| |
Collapse
|
18
|
Hair Follicle Dermal Cells Support Expansion of Murine and Human Embryonic and Induced Pluripotent Stem Cells and Promote Haematopoiesis in Mouse Cultures. Stem Cells Int 2018; 2018:8631432. [PMID: 30154866 PMCID: PMC6098861 DOI: 10.1155/2018/8631432] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Accepted: 04/26/2018] [Indexed: 12/26/2022] Open
Abstract
In the hair follicle, the dermal papilla (DP) and dermal sheath (DS) support and maintain proliferation and differentiation of the epithelial stem cells that produce the hair fibre. In view of their regulatory properties, in this study, we investigated the interaction between hair follicle dermal cells (DP and DS) and embryonic stem cells (ESCs); induced pluripotent stem cells (iPSCs); and haematopoietic stem cells. We found that coculture of follicular dermal cells with ESCs or iPSCs supported their prolonged maintenance in an apparently undifferentiated state as established by differentiation assays, immunocytochemistry, and RT-PCR for markers of undifferentiated ESCs. We further showed that cytokines that are involved in ESC support are also expressed by cultured follicle dermal cells, providing a possible explanation for maintenance of ES cell stemness in cocultures. The same cytokines were expressed within follicles in situ in a pattern more consistent with a role in follicle growth activities than stem cell maintenance. Finally, we show that cultured mouse follicle dermal cells provide good stromal support for haematopoiesis in an established coculture model. Human follicular dermal cells represent an accessible and readily propagated source of feeder cells for pluripotent and haematopoietic cells and have potential for use in clinical applications.
Collapse
|
19
|
Cardiotrophin-1 attenuates experimental colitis in mice. Clin Sci (Lond) 2018; 132:985-1001. [PMID: 29572384 DOI: 10.1042/cs20171513] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 03/09/2018] [Accepted: 03/12/2018] [Indexed: 12/19/2022]
Abstract
Cardiotrophin-1 (CT-1) holds potent anti-inflammatory, cytoprotective, and anti-apoptotic effects in the liver, kidneys, and heart. In the present study, the role of endogenous CT-1 and the effect of exogenous CT-1 were evaluated in experimental ulcerative colitis. Colitis was induced in CT-1 knockout and wild-type (WT) mice by administration of dextran sulphate sodium (DSS) in the drinking water during 7 days. CT-1 knockout mice showed higher colon damage and disease severity than WT mice. In addition, CT-1 (200 µg/kg/day, iv) or vehicle (as control) was administered during 3 days to WT, colitic mice, starting on day 4 after initiation of DSS. Disease activity index (DAI), inflammatory markers (tumor necrosis factor α (TNF-α), INFγ, IL-17, IL-10, inducible nitric oxide synthase (iNOS)), colon damage, apoptosis (cleaved caspase 3), nuclear factor κB (NFκB) and STAT-3 activation, and bacterial translocation were measured. Compared with mice treated with DSS, mice also treated with exogenous CT-1 showed lower colon damage, DAI, plasma levels of TNFα, colon expression of TNF-α, INFγ, IL-17, iNOS and cleaved caspase 3, higher NFκB and signal transducer and activator of transcription 3 (STAT3) pathways activation, and absence of bacterial translocation. We conclude that endogenous CT-1 plays a role in the defense and repair response of the colon against ulcerative lesions through an anti-inflammatory and anti-apoptotic effect. Supplementation with exogenous CT-1 ameliorates disease symptoms, which opens a potentially new therapeutic strategy for ulcerative colitis.
Collapse
|
20
|
Forcina L, Miano C, Musarò A. The physiopathologic interplay between stem cells and tissue niche in muscle regeneration and the role of IL-6 on muscle homeostasis and diseases. Cytokine Growth Factor Rev 2018; 41:1-9. [PMID: 29778303 DOI: 10.1016/j.cytogfr.2018.05.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 05/03/2018] [Indexed: 12/11/2022]
Abstract
Skeletal muscle is a complex, dynamic tissue characterized by an elevated plasticity. Although the adult muscle is mainly composed of multinucleated fibers with post mitotic nuclei, it retains a remarkable ability to regenerate in response to traumatic events. The regenerative potential of the adult skeletal muscle relies in the activity of satellite cells, mononucleated cells residing within the muscle in intimate association with myofibers. Satellite cells normally remain quiescent in their sublaminar position, sporadically entering the cell cycle to guarantee an efficient cellular turnover, by fusing with pre-existing myofibers, and to maintain the stem cell pool. However, after muscle injury satellite cells undergo an extensive increase of their activity in response to environmental stimuli, thereby participating to the regeneration of a functional muscle tissue. Nevertheless, regeneration is affected in several pathologic conditions and by a wide range of environmental signals that are highly variable, not only through time, but also depending on the physiological or pathological conditions of the musculature. Among these factors, the interleukin-6 (IL-6) plays a critical physiopathologic role on muscle homeostasis and diseases. The basis of muscle regeneration and the impact of IL-6 on the physiopathology of skeletal muscle will be discussed.
Collapse
Affiliation(s)
- Laura Forcina
- DAHFMO-Unit of Histology and Medical Embryology, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Via A. Scarpa, 14, Rome 00161, Italy
| | - Carmen Miano
- DAHFMO-Unit of Histology and Medical Embryology, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Via A. Scarpa, 14, Rome 00161, Italy
| | - Antonio Musarò
- DAHFMO-Unit of Histology and Medical Embryology, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Via A. Scarpa, 14, Rome 00161, Italy; Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Rome 00161, Italy.
| |
Collapse
|
21
|
Adrian-Segarra JM, Schindler N, Gajawada P, Lörchner H, Braun T, Pöling J. The AB loop and D-helix in binding site III of human Oncostatin M (OSM) are required for OSM receptor activation. J Biol Chem 2018; 293:7017-7029. [PMID: 29511087 DOI: 10.1074/jbc.ra118.001920] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 02/16/2018] [Indexed: 01/11/2023] Open
Abstract
Oncostatin M (OSM) and leukemia inhibitory factor (LIF) are closely related members of the interleukin-6 (IL-6) cytokine family. Both cytokines share a common origin and structure, and both interact through a specific region, termed binding site III, to activate a dimeric receptor complex formed by glycoprotein 130 (gp130) and LIF receptor (LIFR) in humans. However, only OSM activates the OSM receptor (OSMR)-gp130 complex. The molecular features that enable OSM to specifically activate the OSMR are currently unknown. To define specific sequence motifs within OSM that are critical for initiating signaling via OSMR, here we generated chimeric OSM-LIF cytokines and performed alanine-scanning experiments. Replacement of the OSM AB loop within OSM's binding site III with that of LIF abrogated OSMR activation, measured as STAT3 phosphorylation at Tyr-705, but did not compromise LIFR activation. Correspondingly, substitution of the AB loop and D-helix in LIF with their OSM counterparts was sufficient for OSMR activation. The alanine-scanning experiments revealed that residues Tyr-34, Gln-38, Gly-39, and Leu-45 (in the AB loop) and Pro-153 (in the D-helix) had specific roles in activating OSMR but not LIFR signaling, whereas Leu-40 and Cys-49 (in the AB loop), and Phe-160 and Lys-163 (in the D-helix) were required for activation of both receptors. Because most of the key amino acid residues identified here are conserved between LIF and OSM, we concluded that comparatively minor differences in a few amino acid residues within binding site III account for the differential biological effects of OSM and LIF.
Collapse
Affiliation(s)
- Juan M Adrian-Segarra
- From the Department of Cardiac Development and Remodelling, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Natalie Schindler
- From the Department of Cardiac Development and Remodelling, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Praveen Gajawada
- From the Department of Cardiac Development and Remodelling, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Holger Lörchner
- From the Department of Cardiac Development and Remodelling, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Thomas Braun
- From the Department of Cardiac Development and Remodelling, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Jochen Pöling
- From the Department of Cardiac Development and Remodelling, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| |
Collapse
|
22
|
Human ISL1 + Ventricular Progenitors Self-Assemble into an In Vivo Functional Heart Patch and Preserve Cardiac Function Post Infarction. Mol Ther 2018; 26:1644-1659. [PMID: 29606507 PMCID: PMC6035340 DOI: 10.1016/j.ymthe.2018.02.012] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 02/09/2018] [Accepted: 02/11/2018] [Indexed: 11/26/2022] Open
Abstract
The generation of human pluripotent stem cell (hPSC)-derived ventricular progenitors and their assembly into a 3-dimensional in vivo functional ventricular heart patch has remained an elusive goal. Herein, we report the generation of an enriched pool of hPSC-derived ventricular progenitors (HVPs), which can expand, differentiate, self-assemble, and mature into a functional ventricular patch in vivo without the aid of any gel or matrix. We documented a specific temporal window, in which the HVPs will engraft in vivo. On day 6 of differentiation, HVPs were enriched by depleting cells positive for pluripotency marker TRA-1-60 with magnetic-activated cell sorting (MACS), and 3 million sorted cells were sub-capsularly transplanted onto kidneys of NSG mice where, after 2 months, they formed a 7 mm × 3 mm × 4 mm myocardial patch resembling the ventricular wall. The graft acquired several features of maturation: expression of ventricular marker (MLC2v), desmosomes, appearance of T-tubule-like structures, and electrophysiological action potential signature consistent with maturation, all this in a non-cardiac environment. We further demonstrated that HVPs transplanted into un-injured hearts of NSG mice remain viable for up to 8 months. Moreover, transplantation of 2 million HVPs largely preserved myocardial contractile function following myocardial infarction. Taken together, our study reaffirms the promising idea of using progenitor cells for regenerative therapy.
Collapse
|
23
|
Dombert B, Balk S, Lüningschrör P, Moradi M, Sivadasan R, Saal-Bauernschubert L, Jablonka S. BDNF/trkB Induction of Calcium Transients through Ca v2.2 Calcium Channels in Motoneurons Corresponds to F-actin Assembly and Growth Cone Formation on β2-Chain Laminin (221). Front Mol Neurosci 2017; 10:346. [PMID: 29163025 PMCID: PMC5670157 DOI: 10.3389/fnmol.2017.00346] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 10/12/2017] [Indexed: 12/11/2022] Open
Abstract
Spontaneous Ca2+ transients and actin dynamics in primary motoneurons correspond to cellular differentiation such as axon elongation and growth cone formation. Brain-derived neurotrophic factor (BDNF) and its receptor trkB support both motoneuron survival and synaptic differentiation. However, in motoneurons effects of BDNF/trkB signaling on spontaneous Ca2+ influx and actin dynamics at axonal growth cones are not fully unraveled. In our study we addressed the question how neurotrophic factor signaling corresponds to cell autonomous excitability and growth cone formation. Primary motoneurons from mouse embryos were cultured on the synapse specific, β2-chain containing laminin isoform (221) regulating axon elongation through spontaneous Ca2+ transients that are in turn induced by enhanced clustering of N-type specific voltage-gated Ca2+ channels (Cav2.2) in axonal growth cones. TrkB-deficient (trkBTK-/-) mouse motoneurons which express no full-length trkB receptor and wildtype motoneurons cultured without BDNF exhibited reduced spontaneous Ca2+ transients that corresponded to altered axon elongation and defects in growth cone morphology which was accompanied by changes in the local actin cytoskeleton. Vice versa, the acute application of BDNF resulted in the induction of spontaneous Ca2+ transients and Cav2.2 clustering in motor growth cones, as well as the activation of trkB downstream signaling cascades which promoted the stabilization of β-actin via the LIM kinase pathway and phosphorylation of profilin at Tyr129. Finally, we identified a mutual regulation of neuronal excitability and actin dynamics in axonal growth cones of embryonic motoneurons cultured on laminin-221/211. Impaired excitability resulted in dysregulated axon extension and local actin cytoskeleton, whereas upon β-actin knockdown Cav2.2 clustering was affected. We conclude from our data that in embryonic motoneurons BDNF/trkB signaling contributes to axon elongation and growth cone formation through changes in the local actin cytoskeleton accompanied by increased Cav2.2 clustering and local calcium transients. These findings may help to explore cellular mechanisms which might be dysregulated during maturation of embryonic motoneurons leading to motoneuron disease.
Collapse
Affiliation(s)
- Benjamin Dombert
- Institute of Clinical Neurobiology, University of Wuerzburg, Wuerzburg, Germany
| | - Stefanie Balk
- Institute of Clinical Neurobiology, University of Wuerzburg, Wuerzburg, Germany
| | | | - Mehri Moradi
- Institute of Clinical Neurobiology, University of Wuerzburg, Wuerzburg, Germany
| | - Rajeeve Sivadasan
- Institute of Clinical Neurobiology, University of Wuerzburg, Wuerzburg, Germany
| | | | - Sibylle Jablonka
- Institute of Clinical Neurobiology, University of Wuerzburg, Wuerzburg, Germany
| |
Collapse
|
24
|
Sun S, Wang R, Song J, Guan M, Li N, Zhang X, Zhao Z, Zhang J. Blocking gp130 signaling suppresses autotaxin expression in adipocytes and improves insulin sensitivity in diet-induced obesity. J Lipid Res 2017; 58:2102-2113. [PMID: 28874440 DOI: 10.1194/jlr.m075655] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 08/17/2017] [Indexed: 12/20/2022] Open
Abstract
Autotaxin (ATX), which is highly expressed and secreted by adipocytes, functions as the key enzyme to generate lysophosphatidic acid (LPA) from lysophosphatidylcholine. Adipose tissue is the main source of circulating ATX that modulates plasma LPA levels. Upregulation of ATX expression in obese patients and mice is closely related with insulin resistance and impaired glucose tolerance. However, the mechanism of ATX expression in adipocytes remains largely unknown. In this study, we found that glycoprotein 130 (gp130)-mediated Janus kinase (JAK)-signal transducer and activator of transcription 3 (STAT3) activation was required for abundant ATX expression in adipocytes. Through gp130, the interleukin 6 (IL-6) family cytokines, such as IL-6, leukemia inhibitory factor, cardiotrophin-1, and ciliary neurotrophic factor, upregulated ATX expression in adipocytes. ATX contributes to the induction of insulin resistance and lipolysis in IL-6-stimulated adipocytes. Oral administration of gp130 inhibitor SC144 suppressed ATX expression in adipose tissue, decreased plasma ATX, LPA, and FFA levels, and significantly improved insulin sensitivity and glucose tolerance in high-fat diet-fed obese mice. In summary, our results indicate that the activation of gp130-JAK-STAT3 pathway by IL-6 family cytokines has an important role in regulating ATX expression in adipocytes and that gp130 is a promising target in the management of obesity-associated glucose metabolic diseases.
Collapse
Affiliation(s)
- Shuhong Sun
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Institute of Cell Biology, College of Life Sciences, Beijing Normal University, Beijing 100875, China
| | - Ran Wang
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Institute of Cell Biology, College of Life Sciences, Beijing Normal University, Beijing 100875, China
| | - Jianwen Song
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Institute of Cell Biology, College of Life Sciences, Beijing Normal University, Beijing 100875, China
| | - Ming Guan
- Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry Chinese Academy of Sciences, Beijing 100190, China
| | - Na Li
- Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry Chinese Academy of Sciences, Beijing 100190, China
| | - Xiaotian Zhang
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Institute of Cell Biology, College of Life Sciences, Beijing Normal University, Beijing 100875, China
| | - Zhenwen Zhao
- Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry Chinese Academy of Sciences, Beijing 100190, China
| | - Junjie Zhang
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Institute of Cell Biology, College of Life Sciences, Beijing Normal University, Beijing 100875, China
| |
Collapse
|
25
|
Li L, Lei QS, Kong LN, Zhang SJ, Qin B. Gene expression profile after knockdown of USP18 in Hepg2.2.15 cells. J Med Virol 2017; 89:1920-1930. [PMID: 28369997 DOI: 10.1002/jmv.24819] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Accepted: 03/13/2017] [Indexed: 01/18/2023]
Abstract
In our previous work, we found that the expression of ubiquitin-specific protease 18 (USP18), also known as UBP43, is associated with the efficiency of interferon alpha (IFN-α) treatment in patients with chronic hepatitis B (CHB). To elucidate the influence of USP18 on hepatitis B virus (HBV) replication and the mechanism of this activity, we silenced USP18 by introducing short hairpin RNA (shRNA) into Hepg2.2.15 cells. To identify the changed genes and pathways in Hepg2.2.15-shRNA-USP18 cells, we performed a microarray gene expression analysis to compare the Hepg2.2.15 stably expressing USP18-shRNA cells versus control cells using the Affymetrix Human Transcriptome Array (HTA) 2.0 microarrays. Microarray analysis indicated that genes involved in regulation of thyroid hormone signaling pathway, complement, and coagulation cascades, PERK-mediated unfolded protein response, and insulin-like growth factor-activated receptor activity were significantly altered after USP18 knockdown for 72 h. Furthermore, genes involved in hepatocyte proliferation, liver fibrosis, such as cell cycle regulatory gene CCND1, were also altered after USP18 knockdown in Hepg2.2.15 cells. In conclusion, USP18 is critical for regulating the replication of HBV in Hepg2.2.15 cells, which suggest that USP18 may be a candidate target for HBV treatment.
Collapse
Affiliation(s)
- Lin Li
- Department of Infectious Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qing-Song Lei
- Department of Infectious Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ling-Na Kong
- School of Nursing, Chongqing Medical University, Chongqing, China
| | - Shu-Jun Zhang
- Department of Infectious Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Bo Qin
- Department of Infectious Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
26
|
Brown S, Wilburn W, Martin T, Whalen M. Butyltin compounds alter secretion of interleukin 6 from human immune cells. J Appl Toxicol 2017; 38:201-218. [PMID: 28840599 DOI: 10.1002/jat.3514] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 07/18/2017] [Accepted: 07/19/2017] [Indexed: 01/13/2023]
Abstract
Butyltins (BTs), tributyltin (TBT) and dibutyltin (DBT) are organotin compounds that have been used in a variety of industrial applications; as a result, these compounds have been found in human blood. Interleukin (IL)-6 is a proinflammatory mediator that is produced by T lymphocytes and monocytes. It is responsible for immune response regulation as well as tissue repair and cellular growth. Both BTs decrease the ability of human natural killer cells to destroy tumor cells and alter the secretion of proinflammatory cytokines tumor necrosis factor alpha, interferon gamma and IL-1 beta (β) from human lymphocytes ex vivo. Here, we show that BTs alter the secretion of IL-6 from increasingly reconstituted preparations of human immune cells. IL-6 secretion was examined after 24 hour, 48 hour or 6 day exposures to TBT and DBT in highly enriched human natural killer cells, monocyte-depleted peripheral blood mononuclear cells (PBMCs), PBMCs, granulocytes and a preparation combining both PBMCs and granulocytes (PBMCs + granulocytes). The results indicated that both BTs altered IL-6 secretion from all cell preparations. Significant decreases of IL-6 secretion were seen at the highest concentration of TBT (200 nm) and DBT (5-2.5 μm) while the lower concentrations of DBT (0.05 and 0.1 μm) caused elevation of IL-6 secretion. The data indicate that BT-induced alterations of IL-6 secretion from immune cells may be a significant consequence of BT exposures that may potentially affect immune competence.
Collapse
Affiliation(s)
- Shyretha Brown
- Departments of Biological Sciences and Chemistry, Tennessee State University, Nashville, Tennessee, 37209, USA
| | - Wendy Wilburn
- Departments of Chemistry, Tennessee State University, Nashville, Tennessee, 37209, USA
| | - Tyesha Martin
- Departments of Chemistry, Tennessee State University, Nashville, Tennessee, 37209, USA
| | - Margaret Whalen
- Departments of Chemistry, Tennessee State University, Nashville, Tennessee, 37209, USA
| |
Collapse
|
27
|
Escoté X, Gómez-Zorita S, López-Yoldi M, Milton-Laskibar I, Fernández-Quintela A, Martínez JA, Moreno-Aliaga MJ, Portillo MP. Role of Omentin, Vaspin, Cardiotrophin-1, TWEAK and NOV/CCN3 in Obesity and Diabetes Development. Int J Mol Sci 2017; 18:ijms18081770. [PMID: 28809783 PMCID: PMC5578159 DOI: 10.3390/ijms18081770] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 08/09/2017] [Accepted: 08/10/2017] [Indexed: 01/22/2023] Open
Abstract
Adipose tissue releases bioactive mediators called adipokines. This review focuses on the effects of omentin, vaspin, cardiotrophin-1, Tumor necrosis factor-like Weak Inducer of Apoptosis (TWEAK) and nephroblastoma overexpressed (NOV/CCN3) on obesity and diabetes. Omentin is produced by the stromal-vascular fraction of visceral adipose tissue. Obesity reduces omentin serum concentrations and adipose tissue secretion in adults and adolescents. This adipokine regulates insulin sensitivity, but its clinical relevance has to be confirmed. Vaspin is produced by visceral and subcutaneous adipose tissues. Vaspin levels are higher in obese subjects, as well as in subjects showing insulin resistance or type 2 diabetes. Cardiotrophin-1 is an adipokine with a similar structure as cytokines from interleukin-6 family. There is some controversy regarding the regulation of cardiotrophin-1 levels in obese -subjects, but gene expression levels of cardiotrophin-1 are down-regulated in white adipose tissue from diet-induced obese mice. It also shows anti-obesity and hypoglycemic properties. TWEAK is a potential regulator of the low-grade chronic inflammation characteristic of obesity. TWEAK levels seem not to be directly related to adiposity, and metabolic factors play a critical role in its regulation. Finally, a strong correlation has been found between plasma NOV/CCN3 concentration and fat mass. This adipokine improves insulin actions.
Collapse
Affiliation(s)
- Xavier Escoté
- Department of Nutrition, Food Sciences and Physiology, University of Navarra, 31008 Pamplona, Spain.
- Centre for Nutrition Research, University of Navarra, 31008 Pamplona, Spain.
| | - Saioa Gómez-Zorita
- Nutrition and Obesity Group, Department of Nutrition and Food Science, University of the Basque Country (UPV/EHU) and Lucio Lascaray Research Institute, 01006 Vitoria, Spain.
- Spanish Biomedical Research Centre in Physiopathology of Obesity and Nutrition (CIBERobn), Institute of Health Carlos III, 01006 Vitoria, Spain.
| | - Miguel López-Yoldi
- Department of Nutrition, Food Sciences and Physiology, University of Navarra, 31008 Pamplona, Spain.
- Centre for Nutrition Research, University of Navarra, 31008 Pamplona, Spain.
| | - Iñaki Milton-Laskibar
- Nutrition and Obesity Group, Department of Nutrition and Food Science, University of the Basque Country (UPV/EHU) and Lucio Lascaray Research Institute, 01006 Vitoria, Spain.
- Spanish Biomedical Research Centre in Physiopathology of Obesity and Nutrition (CIBERobn), Institute of Health Carlos III, 01006 Vitoria, Spain.
| | - Alfredo Fernández-Quintela
- Nutrition and Obesity Group, Department of Nutrition and Food Science, University of the Basque Country (UPV/EHU) and Lucio Lascaray Research Institute, 01006 Vitoria, Spain.
- Spanish Biomedical Research Centre in Physiopathology of Obesity and Nutrition (CIBERobn), Institute of Health Carlos III, 01006 Vitoria, Spain.
| | - J Alfredo Martínez
- Department of Nutrition, Food Sciences and Physiology, University of Navarra, 31008 Pamplona, Spain.
- Centre for Nutrition Research, University of Navarra, 31008 Pamplona, Spain.
- Spanish Biomedical Research Centre in Physiopathology of Obesity and Nutrition (CIBERobn), Institute of Health Carlos III, 01006 Vitoria, Spain.
- Navarra Institute for Health Research (IdiSNa), 31008 Pamplona, Spain.
| | - María J Moreno-Aliaga
- Department of Nutrition, Food Sciences and Physiology, University of Navarra, 31008 Pamplona, Spain.
- Centre for Nutrition Research, University of Navarra, 31008 Pamplona, Spain.
- Spanish Biomedical Research Centre in Physiopathology of Obesity and Nutrition (CIBERobn), Institute of Health Carlos III, 01006 Vitoria, Spain.
- Navarra Institute for Health Research (IdiSNa), 31008 Pamplona, Spain.
| | - María P Portillo
- Nutrition and Obesity Group, Department of Nutrition and Food Science, University of the Basque Country (UPV/EHU) and Lucio Lascaray Research Institute, 01006 Vitoria, Spain.
- Spanish Biomedical Research Centre in Physiopathology of Obesity and Nutrition (CIBERobn), Institute of Health Carlos III, 01006 Vitoria, Spain.
| |
Collapse
|
28
|
Morgani S, Nichols J, Hadjantonakis AK. The many faces of Pluripotency: in vitro adaptations of a continuum of in vivo states. BMC DEVELOPMENTAL BIOLOGY 2017; 17:7. [PMID: 28610558 PMCID: PMC5470286 DOI: 10.1186/s12861-017-0150-4] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 06/01/2017] [Indexed: 12/20/2022]
Abstract
Pluripotency defines the propensity of a cell to differentiate into, and generate, all somatic, as well as germ cells. The epiblast of the early mammalian embryo is the founder population of all germ layer derivatives and thus represents the bona fide in vivo pluripotent cell population. The so-called pluripotent state spans several days of development and is lost during gastrulation as epiblast cells make fate decisions towards a mesoderm, endoderm or ectoderm identity. It is now widely recognized that the features of the pluripotent population evolve as development proceeds from the pre- to post-implantation period, marked by distinct transcriptional and epigenetic signatures. During this period of time epiblast cells mature through a continuum of pluripotent states with unique properties. Aspects of this pluripotent continuum can be captured in vitro in the form of stable pluripotent stem cell types. In this review we discuss the continuum of pluripotency existing within the mammalian embryo, using the mouse as a model, and the cognate stem cell types that can be derived and propagated in vitro. Furthermore, we speculate on embryonic stage-specific characteristics that could be utilized to identify novel, developmentally relevant, pluripotent states.
Collapse
Affiliation(s)
- Sophie Morgani
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
- Wellcome Trust-Medical Research Council Centre for Stem Cell Research, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QR, UK
| | - Jennifer Nichols
- Wellcome Trust-Medical Research Council Centre for Stem Cell Research, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QR, UK
| | - Anna-Katerina Hadjantonakis
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA.
| |
Collapse
|
29
|
Peng L, Shu X, Lang C, Yu X. Cardiotrophin-1 stimulates the neural differentiation of human umbilical cord blood-derived mesenchymal stem cells and survival of differentiated cells through PI3K/Akt-dependent signaling pathways. Cytotechnology 2017; 69:933-941. [PMID: 28601931 DOI: 10.1007/s10616-017-0103-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 04/22/2017] [Indexed: 12/31/2022] Open
Abstract
Cardiotrophin-1 (CT1) plays an important role in the differentiation, development, and survival of neural stem cells. In this study, we analyzed its effects on the stimulation of human umbilical cord blood-derived mesenchymal stem cells in terms of their potential to differentiate into neuron-like cells, their survival characteristics, and the molecular mechanisms involved. The treatment of cells with neural induction medium (NIM) and CT1 generated more cells that were neuron-like and produced stronger expression of neural-lineage markers than cells treated with NIM and without CT1. Bcl-2 and Akt phosphorylation (p-Akt) expression levels increased significantly in cells treated with both NIM and CT1. This treatment also effectively blocked cell death following neural induction and decreased Bax, Bak and cleaved-caspase 3 expression compared with cells treated with NIM without CT1. In addition, the inhibition of phosphatidylinositol 3-kinase (PI3K) abrogated p-Akt and Bcl-2 expression. Thus, PI3K/Akt contribute to CT1-stimulated neural differentiation and to the survival of differentiated cells.
Collapse
Affiliation(s)
- Longying Peng
- Department of Pediatric, First Affiliated Hospital, Zunyi Medical College, Zunyi, 563003, Guizhou Province, China
| | - Xiaomei Shu
- Department of Pediatric, First Affiliated Hospital, Zunyi Medical College, Zunyi, 563003, Guizhou Province, China.
| | - Changhui Lang
- Department of Pediatric, First Affiliated Hospital, Zunyi Medical College, Zunyi, 563003, Guizhou Province, China
| | - Xiaohua Yu
- Department of Pediatric, First Affiliated Hospital, Zunyi Medical College, Zunyi, 563003, Guizhou Province, China
| |
Collapse
|
30
|
López-Yoldi M, Marcos-Gomez B, Romero-Lozano MA, Sáinz N, Prieto J, Martínez JA, Bustos M, Moreno-Aliaga MJ. Cardiotrophin-1 Regulates Adipokine Production in 3T3-L1 Adipocytes and Adipose Tissue From Obese Mice. J Cell Physiol 2017; 232:2469-2477. [PMID: 27608275 DOI: 10.1002/jcp.25590] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 09/06/2016] [Indexed: 12/19/2022]
Abstract
Cardiotrophin-1 (CT-1) belongs to the IL-6 family of cytokines. Previous studies of our group revealed that CT-1 is a key regulator of glucose and lipid metabolism. The aim of the present study was to analyze the in vitro and in vivo effects of CT-1 on the production of several adipokines involved in body weight regulation, nutrient metabolism, and inflammation. For this purpose, 3T3-L1 adipocytes were incubated with recombinant protein CT-1 (rCT-1) (1-40 ng/ml) for 1 and 18 h. Moreover, the acute effects of rCT-1 administration (0.2 mg/kg, i.v.) for 30 min and 3 h on adipokines levels were also evaluated in high-fat fed obese mice. In 3T3-L1 adipocytes, rCT-1 treatment downregulated the expression and secretion of leptin, resistin, and visfatin. However, rCT-1 significantly stimulated apelin mRNA and secretion. rCT-1 (18 h) also promoted the activation by phosphorylation of AKT, ERK 1/2, and STAT3. Interestingly, pre-treatment with the PI3K inhibitor LY294002 reversed the stimulatory effects of rCT-1 on apelin expression, suggesting that this pathway could be mediating the effects of rCT-1 on apelin production. In contrast, acute administration of rCT-1 (30 min and 3 h) to diet-induced obese mice downregulated leptin and resistin, without significantly modifying apelin or visfatin mRNA in adipose tissue. Furthermore, CT-1 null mice exhibited altered expression of adipokines in adipose tissue. The present study demonstrates that rCT-1 modulates the production of adipokines in vitro and in vivo, suggesting that the regulation of the secretory function of adipocytes could be involved in the metabolic actions of this cytokine. J. Cell. Physiol. 232: 2469-2477, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Miguel López-Yoldi
- Department of Nutrition, Food Science and Physiology, University of Navarra, Pamplona, Navarra, Spain.,Centre for Nutrition Research, University of Navarra, Pamplona, Navarra, Spain
| | - Beatriz Marcos-Gomez
- Department of Nutrition, Food Science and Physiology, University of Navarra, Pamplona, Navarra, Spain
| | | | - Neira Sáinz
- Department of Nutrition, Food Science and Physiology, University of Navarra, Pamplona, Navarra, Spain.,Centre for Nutrition Research, University of Navarra, Pamplona, Navarra, Spain
| | - Jesús Prieto
- Department of Hepatology and Gene Therapy, CIMA, University of Navarra, Pamplona, Navarra, Spain.,CIBERehd, Institute of Health Carlos III, Madrid, Spain
| | - Jose Alfredo Martínez
- Department of Nutrition, Food Science and Physiology, University of Navarra, Pamplona, Navarra, Spain.,Centre for Nutrition Research, University of Navarra, Pamplona, Navarra, Spain.,CIBERobn, Physiopathology of Obesity and Nutrition, Institute of Health Carlos III, Madrid, Spain.,IdiSNA, Navarra's Health Research Institute, Pamplona, Spain
| | - Matilde Bustos
- Department of Hepatology and Gene Therapy, CIMA, University of Navarra, Pamplona, Navarra, Spain
| | - Maria J Moreno-Aliaga
- Department of Nutrition, Food Science and Physiology, University of Navarra, Pamplona, Navarra, Spain.,Centre for Nutrition Research, University of Navarra, Pamplona, Navarra, Spain.,CIBERobn, Physiopathology of Obesity and Nutrition, Institute of Health Carlos III, Madrid, Spain.,IdiSNA, Navarra's Health Research Institute, Pamplona, Spain
| |
Collapse
|
31
|
Ngo D, Sinha S, Shen D, Kuhn EW, Keyes MJ, Shi X, Benson MD, O'Sullivan JF, Keshishian H, Farrell LA, Fifer MA, Vasan RS, Sabatine MS, Larson MG, Carr SA, Wang TJ, Gerszten RE. Aptamer-Based Proteomic Profiling Reveals Novel Candidate Biomarkers and Pathways in Cardiovascular Disease. Circulation 2016; 134:270-85. [PMID: 27444932 DOI: 10.1161/circulationaha.116.021803] [Citation(s) in RCA: 162] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 06/20/2016] [Indexed: 01/22/2023]
Abstract
BACKGROUND Single-stranded DNA aptamers are oligonucleotides of ≈50 base pairs in length selected for their ability to bind proteins with high specificity and affinity. Emerging DNA aptamer-based technologies may address limitations of existing proteomic techniques, including low sample throughput, which have hindered proteomic analyses of large cohorts. METHODS To identify early biomarkers of myocardial injury, we applied an aptamer-based proteomic platform that measures 1129 proteins to a clinically relevant perturbational model of planned myocardial infarction (PMI), patients undergoing septal ablation for hypertrophic cardiomyopathy. Blood samples were obtained before and at 10 and 60 minutes after PMI, and protein changes were assessed by repeated-measures analysis of variance. The generalizability of our PMI findings was evaluated in a spontaneous myocardial infarction cohort (Wilcoxon rank-sum). We then tested the platform's ability to detect associations between proteins and Framingham Risk Score components in the Framingham Heart Study, performing regression analyses for each protein versus each clinical trait. RESULTS We found 217 proteins that significantly changed in the peripheral vein blood after PMI in a derivation cohort (n=15; P<5.70E-5). Seventy-nine of these proteins were validated in an independent PMI cohort (n=15; P<2.30E-4); >85% were directionally consistent and reached nominal significance. We detected many protein changes that are novel in the context of myocardial injury, including Dickkopf-related protein 4, a WNT pathway inhibitor (peak increase 124%, P=1.29E-15) and cripto, a growth factor important in cardiac development (peak increase 64%, P=1.74E-4). Among the 40 validated proteins that increased within 1 hour after PMI, 23 were also elevated in patients with spontaneous myocardial infarction (n=46; P<0.05). Framingham Heart Study analyses revealed 156 significant protein associations with the Framingham Risk Score (n=899), including aminoacylase 1 (β=0.3386, P=2.54E-22) and trigger factor 2 (β=0.2846, P=5.71E-17). Furthermore, we developed a novel workflow integrating DNA-based immunoaffinity with mass spectrometry to analytically validate aptamer specificity. CONCLUSIONS Our results highlight an emerging proteomics tool capable of profiling >1000 low-abundance analytes with high sensitivity and high precision, applicable both to well-phenotyped perturbational studies and large human cohorts, as well.
Collapse
Affiliation(s)
- Debby Ngo
- From Division of Pulmonary and Critical Care Medicine, Department of Medicine (D.N.) and the Cardiovascular Research Center (D.N., S.S., D.S., M.J.K., X.S., M.D.B., J.F.O., L.A.F., R.E.G.), Massachusetts General Hospital, Harvard Medical School, Boston; Broad Institute of MIT and Harvard, Cambridge, MA (E.W.K., H.K., S.A.C., R.E.G.); Cardiovascular Division, Brigham and Women's Hospital, Boston, MA (M.D.B., M.S.S.); Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston (M.A.F., M.S.S., R.E.G.); Preventive Medicine Section, Department of Medicine, Boston University School of Medicine, MA (R.S.V.); Department of Biostatistics, Boston University School of Public Health, MA (M.G.L.); The National Heart, Lung, and Blood Institute's Framingham Heart Study, Framingham, MA (M.G.L.); Division of Cardiovascular Medicine, Vanderbilt University, Nashville, TN (T.J.W.); and Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA (R.E.G.)
| | - Sumita Sinha
- From Division of Pulmonary and Critical Care Medicine, Department of Medicine (D.N.) and the Cardiovascular Research Center (D.N., S.S., D.S., M.J.K., X.S., M.D.B., J.F.O., L.A.F., R.E.G.), Massachusetts General Hospital, Harvard Medical School, Boston; Broad Institute of MIT and Harvard, Cambridge, MA (E.W.K., H.K., S.A.C., R.E.G.); Cardiovascular Division, Brigham and Women's Hospital, Boston, MA (M.D.B., M.S.S.); Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston (M.A.F., M.S.S., R.E.G.); Preventive Medicine Section, Department of Medicine, Boston University School of Medicine, MA (R.S.V.); Department of Biostatistics, Boston University School of Public Health, MA (M.G.L.); The National Heart, Lung, and Blood Institute's Framingham Heart Study, Framingham, MA (M.G.L.); Division of Cardiovascular Medicine, Vanderbilt University, Nashville, TN (T.J.W.); and Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA (R.E.G.)
| | - Dongxiao Shen
- From Division of Pulmonary and Critical Care Medicine, Department of Medicine (D.N.) and the Cardiovascular Research Center (D.N., S.S., D.S., M.J.K., X.S., M.D.B., J.F.O., L.A.F., R.E.G.), Massachusetts General Hospital, Harvard Medical School, Boston; Broad Institute of MIT and Harvard, Cambridge, MA (E.W.K., H.K., S.A.C., R.E.G.); Cardiovascular Division, Brigham and Women's Hospital, Boston, MA (M.D.B., M.S.S.); Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston (M.A.F., M.S.S., R.E.G.); Preventive Medicine Section, Department of Medicine, Boston University School of Medicine, MA (R.S.V.); Department of Biostatistics, Boston University School of Public Health, MA (M.G.L.); The National Heart, Lung, and Blood Institute's Framingham Heart Study, Framingham, MA (M.G.L.); Division of Cardiovascular Medicine, Vanderbilt University, Nashville, TN (T.J.W.); and Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA (R.E.G.)
| | - Eric W Kuhn
- From Division of Pulmonary and Critical Care Medicine, Department of Medicine (D.N.) and the Cardiovascular Research Center (D.N., S.S., D.S., M.J.K., X.S., M.D.B., J.F.O., L.A.F., R.E.G.), Massachusetts General Hospital, Harvard Medical School, Boston; Broad Institute of MIT and Harvard, Cambridge, MA (E.W.K., H.K., S.A.C., R.E.G.); Cardiovascular Division, Brigham and Women's Hospital, Boston, MA (M.D.B., M.S.S.); Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston (M.A.F., M.S.S., R.E.G.); Preventive Medicine Section, Department of Medicine, Boston University School of Medicine, MA (R.S.V.); Department of Biostatistics, Boston University School of Public Health, MA (M.G.L.); The National Heart, Lung, and Blood Institute's Framingham Heart Study, Framingham, MA (M.G.L.); Division of Cardiovascular Medicine, Vanderbilt University, Nashville, TN (T.J.W.); and Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA (R.E.G.)
| | - Michelle J Keyes
- From Division of Pulmonary and Critical Care Medicine, Department of Medicine (D.N.) and the Cardiovascular Research Center (D.N., S.S., D.S., M.J.K., X.S., M.D.B., J.F.O., L.A.F., R.E.G.), Massachusetts General Hospital, Harvard Medical School, Boston; Broad Institute of MIT and Harvard, Cambridge, MA (E.W.K., H.K., S.A.C., R.E.G.); Cardiovascular Division, Brigham and Women's Hospital, Boston, MA (M.D.B., M.S.S.); Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston (M.A.F., M.S.S., R.E.G.); Preventive Medicine Section, Department of Medicine, Boston University School of Medicine, MA (R.S.V.); Department of Biostatistics, Boston University School of Public Health, MA (M.G.L.); The National Heart, Lung, and Blood Institute's Framingham Heart Study, Framingham, MA (M.G.L.); Division of Cardiovascular Medicine, Vanderbilt University, Nashville, TN (T.J.W.); and Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA (R.E.G.)
| | - Xu Shi
- From Division of Pulmonary and Critical Care Medicine, Department of Medicine (D.N.) and the Cardiovascular Research Center (D.N., S.S., D.S., M.J.K., X.S., M.D.B., J.F.O., L.A.F., R.E.G.), Massachusetts General Hospital, Harvard Medical School, Boston; Broad Institute of MIT and Harvard, Cambridge, MA (E.W.K., H.K., S.A.C., R.E.G.); Cardiovascular Division, Brigham and Women's Hospital, Boston, MA (M.D.B., M.S.S.); Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston (M.A.F., M.S.S., R.E.G.); Preventive Medicine Section, Department of Medicine, Boston University School of Medicine, MA (R.S.V.); Department of Biostatistics, Boston University School of Public Health, MA (M.G.L.); The National Heart, Lung, and Blood Institute's Framingham Heart Study, Framingham, MA (M.G.L.); Division of Cardiovascular Medicine, Vanderbilt University, Nashville, TN (T.J.W.); and Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA (R.E.G.)
| | - Mark D Benson
- From Division of Pulmonary and Critical Care Medicine, Department of Medicine (D.N.) and the Cardiovascular Research Center (D.N., S.S., D.S., M.J.K., X.S., M.D.B., J.F.O., L.A.F., R.E.G.), Massachusetts General Hospital, Harvard Medical School, Boston; Broad Institute of MIT and Harvard, Cambridge, MA (E.W.K., H.K., S.A.C., R.E.G.); Cardiovascular Division, Brigham and Women's Hospital, Boston, MA (M.D.B., M.S.S.); Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston (M.A.F., M.S.S., R.E.G.); Preventive Medicine Section, Department of Medicine, Boston University School of Medicine, MA (R.S.V.); Department of Biostatistics, Boston University School of Public Health, MA (M.G.L.); The National Heart, Lung, and Blood Institute's Framingham Heart Study, Framingham, MA (M.G.L.); Division of Cardiovascular Medicine, Vanderbilt University, Nashville, TN (T.J.W.); and Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA (R.E.G.)
| | - John F O'Sullivan
- From Division of Pulmonary and Critical Care Medicine, Department of Medicine (D.N.) and the Cardiovascular Research Center (D.N., S.S., D.S., M.J.K., X.S., M.D.B., J.F.O., L.A.F., R.E.G.), Massachusetts General Hospital, Harvard Medical School, Boston; Broad Institute of MIT and Harvard, Cambridge, MA (E.W.K., H.K., S.A.C., R.E.G.); Cardiovascular Division, Brigham and Women's Hospital, Boston, MA (M.D.B., M.S.S.); Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston (M.A.F., M.S.S., R.E.G.); Preventive Medicine Section, Department of Medicine, Boston University School of Medicine, MA (R.S.V.); Department of Biostatistics, Boston University School of Public Health, MA (M.G.L.); The National Heart, Lung, and Blood Institute's Framingham Heart Study, Framingham, MA (M.G.L.); Division of Cardiovascular Medicine, Vanderbilt University, Nashville, TN (T.J.W.); and Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA (R.E.G.)
| | - Hasmik Keshishian
- From Division of Pulmonary and Critical Care Medicine, Department of Medicine (D.N.) and the Cardiovascular Research Center (D.N., S.S., D.S., M.J.K., X.S., M.D.B., J.F.O., L.A.F., R.E.G.), Massachusetts General Hospital, Harvard Medical School, Boston; Broad Institute of MIT and Harvard, Cambridge, MA (E.W.K., H.K., S.A.C., R.E.G.); Cardiovascular Division, Brigham and Women's Hospital, Boston, MA (M.D.B., M.S.S.); Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston (M.A.F., M.S.S., R.E.G.); Preventive Medicine Section, Department of Medicine, Boston University School of Medicine, MA (R.S.V.); Department of Biostatistics, Boston University School of Public Health, MA (M.G.L.); The National Heart, Lung, and Blood Institute's Framingham Heart Study, Framingham, MA (M.G.L.); Division of Cardiovascular Medicine, Vanderbilt University, Nashville, TN (T.J.W.); and Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA (R.E.G.)
| | - Laurie A Farrell
- From Division of Pulmonary and Critical Care Medicine, Department of Medicine (D.N.) and the Cardiovascular Research Center (D.N., S.S., D.S., M.J.K., X.S., M.D.B., J.F.O., L.A.F., R.E.G.), Massachusetts General Hospital, Harvard Medical School, Boston; Broad Institute of MIT and Harvard, Cambridge, MA (E.W.K., H.K., S.A.C., R.E.G.); Cardiovascular Division, Brigham and Women's Hospital, Boston, MA (M.D.B., M.S.S.); Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston (M.A.F., M.S.S., R.E.G.); Preventive Medicine Section, Department of Medicine, Boston University School of Medicine, MA (R.S.V.); Department of Biostatistics, Boston University School of Public Health, MA (M.G.L.); The National Heart, Lung, and Blood Institute's Framingham Heart Study, Framingham, MA (M.G.L.); Division of Cardiovascular Medicine, Vanderbilt University, Nashville, TN (T.J.W.); and Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA (R.E.G.)
| | - Michael A Fifer
- From Division of Pulmonary and Critical Care Medicine, Department of Medicine (D.N.) and the Cardiovascular Research Center (D.N., S.S., D.S., M.J.K., X.S., M.D.B., J.F.O., L.A.F., R.E.G.), Massachusetts General Hospital, Harvard Medical School, Boston; Broad Institute of MIT and Harvard, Cambridge, MA (E.W.K., H.K., S.A.C., R.E.G.); Cardiovascular Division, Brigham and Women's Hospital, Boston, MA (M.D.B., M.S.S.); Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston (M.A.F., M.S.S., R.E.G.); Preventive Medicine Section, Department of Medicine, Boston University School of Medicine, MA (R.S.V.); Department of Biostatistics, Boston University School of Public Health, MA (M.G.L.); The National Heart, Lung, and Blood Institute's Framingham Heart Study, Framingham, MA (M.G.L.); Division of Cardiovascular Medicine, Vanderbilt University, Nashville, TN (T.J.W.); and Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA (R.E.G.)
| | - Ramachandran S Vasan
- From Division of Pulmonary and Critical Care Medicine, Department of Medicine (D.N.) and the Cardiovascular Research Center (D.N., S.S., D.S., M.J.K., X.S., M.D.B., J.F.O., L.A.F., R.E.G.), Massachusetts General Hospital, Harvard Medical School, Boston; Broad Institute of MIT and Harvard, Cambridge, MA (E.W.K., H.K., S.A.C., R.E.G.); Cardiovascular Division, Brigham and Women's Hospital, Boston, MA (M.D.B., M.S.S.); Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston (M.A.F., M.S.S., R.E.G.); Preventive Medicine Section, Department of Medicine, Boston University School of Medicine, MA (R.S.V.); Department of Biostatistics, Boston University School of Public Health, MA (M.G.L.); The National Heart, Lung, and Blood Institute's Framingham Heart Study, Framingham, MA (M.G.L.); Division of Cardiovascular Medicine, Vanderbilt University, Nashville, TN (T.J.W.); and Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA (R.E.G.)
| | - Marc S Sabatine
- From Division of Pulmonary and Critical Care Medicine, Department of Medicine (D.N.) and the Cardiovascular Research Center (D.N., S.S., D.S., M.J.K., X.S., M.D.B., J.F.O., L.A.F., R.E.G.), Massachusetts General Hospital, Harvard Medical School, Boston; Broad Institute of MIT and Harvard, Cambridge, MA (E.W.K., H.K., S.A.C., R.E.G.); Cardiovascular Division, Brigham and Women's Hospital, Boston, MA (M.D.B., M.S.S.); Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston (M.A.F., M.S.S., R.E.G.); Preventive Medicine Section, Department of Medicine, Boston University School of Medicine, MA (R.S.V.); Department of Biostatistics, Boston University School of Public Health, MA (M.G.L.); The National Heart, Lung, and Blood Institute's Framingham Heart Study, Framingham, MA (M.G.L.); Division of Cardiovascular Medicine, Vanderbilt University, Nashville, TN (T.J.W.); and Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA (R.E.G.)
| | - Martin G Larson
- From Division of Pulmonary and Critical Care Medicine, Department of Medicine (D.N.) and the Cardiovascular Research Center (D.N., S.S., D.S., M.J.K., X.S., M.D.B., J.F.O., L.A.F., R.E.G.), Massachusetts General Hospital, Harvard Medical School, Boston; Broad Institute of MIT and Harvard, Cambridge, MA (E.W.K., H.K., S.A.C., R.E.G.); Cardiovascular Division, Brigham and Women's Hospital, Boston, MA (M.D.B., M.S.S.); Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston (M.A.F., M.S.S., R.E.G.); Preventive Medicine Section, Department of Medicine, Boston University School of Medicine, MA (R.S.V.); Department of Biostatistics, Boston University School of Public Health, MA (M.G.L.); The National Heart, Lung, and Blood Institute's Framingham Heart Study, Framingham, MA (M.G.L.); Division of Cardiovascular Medicine, Vanderbilt University, Nashville, TN (T.J.W.); and Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA (R.E.G.)
| | - Steven A Carr
- From Division of Pulmonary and Critical Care Medicine, Department of Medicine (D.N.) and the Cardiovascular Research Center (D.N., S.S., D.S., M.J.K., X.S., M.D.B., J.F.O., L.A.F., R.E.G.), Massachusetts General Hospital, Harvard Medical School, Boston; Broad Institute of MIT and Harvard, Cambridge, MA (E.W.K., H.K., S.A.C., R.E.G.); Cardiovascular Division, Brigham and Women's Hospital, Boston, MA (M.D.B., M.S.S.); Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston (M.A.F., M.S.S., R.E.G.); Preventive Medicine Section, Department of Medicine, Boston University School of Medicine, MA (R.S.V.); Department of Biostatistics, Boston University School of Public Health, MA (M.G.L.); The National Heart, Lung, and Blood Institute's Framingham Heart Study, Framingham, MA (M.G.L.); Division of Cardiovascular Medicine, Vanderbilt University, Nashville, TN (T.J.W.); and Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA (R.E.G.).
| | - Thomas J Wang
- From Division of Pulmonary and Critical Care Medicine, Department of Medicine (D.N.) and the Cardiovascular Research Center (D.N., S.S., D.S., M.J.K., X.S., M.D.B., J.F.O., L.A.F., R.E.G.), Massachusetts General Hospital, Harvard Medical School, Boston; Broad Institute of MIT and Harvard, Cambridge, MA (E.W.K., H.K., S.A.C., R.E.G.); Cardiovascular Division, Brigham and Women's Hospital, Boston, MA (M.D.B., M.S.S.); Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston (M.A.F., M.S.S., R.E.G.); Preventive Medicine Section, Department of Medicine, Boston University School of Medicine, MA (R.S.V.); Department of Biostatistics, Boston University School of Public Health, MA (M.G.L.); The National Heart, Lung, and Blood Institute's Framingham Heart Study, Framingham, MA (M.G.L.); Division of Cardiovascular Medicine, Vanderbilt University, Nashville, TN (T.J.W.); and Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA (R.E.G.).
| | - Robert E Gerszten
- From Division of Pulmonary and Critical Care Medicine, Department of Medicine (D.N.) and the Cardiovascular Research Center (D.N., S.S., D.S., M.J.K., X.S., M.D.B., J.F.O., L.A.F., R.E.G.), Massachusetts General Hospital, Harvard Medical School, Boston; Broad Institute of MIT and Harvard, Cambridge, MA (E.W.K., H.K., S.A.C., R.E.G.); Cardiovascular Division, Brigham and Women's Hospital, Boston, MA (M.D.B., M.S.S.); Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston (M.A.F., M.S.S., R.E.G.); Preventive Medicine Section, Department of Medicine, Boston University School of Medicine, MA (R.S.V.); Department of Biostatistics, Boston University School of Public Health, MA (M.G.L.); The National Heart, Lung, and Blood Institute's Framingham Heart Study, Framingham, MA (M.G.L.); Division of Cardiovascular Medicine, Vanderbilt University, Nashville, TN (T.J.W.); and Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA (R.E.G.)
| |
Collapse
|
32
|
López-Yoldi M, Castilla-Madrigal R, Lostao MP, Barber A, Prieto J, Martínez JA, Bustos M, Moreno-Aliaga MJ. Cardiotrophin-1 decreases intestinal sugar uptake in mice and in Caco-2 cells. Acta Physiol (Oxf) 2016; 217:217-26. [PMID: 26972986 DOI: 10.1111/apha.12674] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Revised: 10/12/2015] [Accepted: 03/04/2016] [Indexed: 12/15/2022]
Abstract
AIM Cardiotrophin-1 (CT-1) is a member of the IL-6 family of cytokines with a key role in glucose and lipid metabolism. In the current investigation, we examined the in vivo and in vitro effects of CT-1 treatment on intestinal sugar absorption in different experimental models. METHODS rCT-1 effects on α-Methyl-D-glucoside uptake were assessed in everted intestinal rings from wild-type and CT-1(-/-) mice and in Caco-2 cells. rCT-1 actions on SGLT-1 expression in brush border membrane vesicles and the identification of the potential signalling pathways involved were determined by Western blot. RESULTS In vivo administration (0.2 mg kg(-1) ) of rCT-1 caused a significant decrease on α-Methyl-D-glucoside uptake in everted intestinal rings from wild-type and CT-1(-/-) mice after short-term and long-term treatments. Similarly, in vitro treatment (1-50 ng mL(-1) ) with rCT-1 reduced α-Methyl-D-glucoside uptake in everted intestinal rings. In Caco-2 cells, rCT-1 treatment (20 ng mL(-1) , 1 and 24 h) lowered apical uptake of α-Methyl-D-glucoside in parallel with a decrease on SGLT-1 protein expression. rCT-1 promoted the phosphorylation of STAT-3 after 5 and 15 min treatment, but inhibited the activation by phosphorylation of AMPK after 30 and 60 min. Interestingly, pre-treatment with the JAK/STAT inhibitor (AG490) and with the AMPK activator (AICAR) reversed the inhibitory effects of rCT-1 on α-Methyl-D-glucoside uptake. AICAR also prevented the inhibition of SGLT-1 observed in rCT-1-treated cells. CONCLUSIONS CT-1 inhibits intestinal sugar absorption by the reduction of SGLT-1 levels through the AMPK pathway, which could also contribute to explain the hypoglycaemic and anti-obesity properties of CT-1.
Collapse
Affiliation(s)
- M. López-Yoldi
- Department of Nutrition, Food Science and Physiology; University of Navarra; Pamplona Navarra Spain
- Centre for Nutrition Research; University of Navarra; Pamplona Navarra Spain
| | - R. Castilla-Madrigal
- Department of Nutrition, Food Science and Physiology; University of Navarra; Pamplona Navarra Spain
- Centre for Nutrition Research; University of Navarra; Pamplona Navarra Spain
| | - M. P. Lostao
- Department of Nutrition, Food Science and Physiology; University of Navarra; Pamplona Navarra Spain
- Centre for Nutrition Research; University of Navarra; Pamplona Navarra Spain
- IdiSNA; Navarra Institute for Health Research; Pamplona Spain
| | - A. Barber
- Department of Nutrition, Food Science and Physiology; University of Navarra; Pamplona Navarra Spain
| | - J. Prieto
- Department of Gene Therapy and Hepatology; CIMA; University of Navarra; Pamplona Navarra Spain
- CIBERehd; Institute of Health Carlos III; Madrid Spain
| | - J. A. Martínez
- Department of Nutrition, Food Science and Physiology; University of Navarra; Pamplona Navarra Spain
- Centre for Nutrition Research; University of Navarra; Pamplona Navarra Spain
- IdiSNA; Navarra Institute for Health Research; Pamplona Spain
- CIBERobn; Physiopathology of Obesity and Nutrition; Institute of Health Carlos III; Madrid Spain
| | - M. Bustos
- Department of Gene Therapy and Hepatology; CIMA; University of Navarra; Pamplona Navarra Spain
| | - M. J. Moreno-Aliaga
- Department of Nutrition, Food Science and Physiology; University of Navarra; Pamplona Navarra Spain
- Centre for Nutrition Research; University of Navarra; Pamplona Navarra Spain
- IdiSNA; Navarra Institute for Health Research; Pamplona Spain
- CIBERobn; Physiopathology of Obesity and Nutrition; Institute of Health Carlos III; Madrid Spain
| |
Collapse
|
33
|
Hunt LC, White J. The Role of Leukemia Inhibitory Factor Receptor Signaling in Skeletal Muscle Growth, Injury and Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 900:45-59. [DOI: 10.1007/978-3-319-27511-6_3] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
34
|
Guild J, Haque A, Gheibi P, Gao Y, Son KJ, Foster E, Dumont S, Revzin A. Embryonic Stem Cells Cultured in Microfluidic Chambers Take Control of Their Fate by Producing Endogenous Signals Including LIF. Stem Cells 2016; 34:1501-12. [DOI: 10.1002/stem.2324] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Accepted: 12/23/2015] [Indexed: 01/09/2023]
Affiliation(s)
- Joshua Guild
- Department of Biomedical Engineering; University of California, Davis; Davis California USA
- Department of Cell and Tissue Biology; University of California San Francisco; San Francisco California USA
| | - Amranul Haque
- Department of Biomedical Engineering; University of California, Davis; Davis California USA
| | - Pantea Gheibi
- Department of Biomedical Engineering; University of California, Davis; Davis California USA
| | - Yandong Gao
- Department of Biomedical Engineering; University of California, Davis; Davis California USA
| | - Kyung Jin Son
- Department of Biomedical Engineering; University of California, Davis; Davis California USA
| | - Elena Foster
- Department of Biomedical Engineering; University of California, Davis; Davis California USA
| | - Sophie Dumont
- Department of Cell and Tissue Biology; University of California San Francisco; San Francisco California USA
- Department of Cellular and Molecular Pharmacology; University of California; San Francisco, San Francisco California USA
| | - Alexander Revzin
- Department of Biomedical Engineering; University of California, Davis; Davis California USA
| |
Collapse
|
35
|
Cardiotrophin-1 promotes cardiomyocyte differentiation from mouse induced pluripotent stem cells via JAK2/STAT3/Pim-1 signaling pathway. JOURNAL OF GERIATRIC CARDIOLOGY : JGC 2016; 12:591-9. [PMID: 26788034 PMCID: PMC4712363 DOI: 10.11909/j.issn.1671-5411.2015.06.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
BACKGROUND The induced pluripotent stem cell (iPSC) has shown great potential in cellular therapy of myocardial infarction (MI), while its application is hampered by the low efficiency of cardiomyocyte differentiation. The present study was designed to investigate the effects of cardiotrophin-1 (CT-1) on cardiomyocyte differentiation from mouse induced pluripotent stem cells (miPSCs) and the underlying mechanisms involved. METHODS The optimal treatment condition for cardiomyocyte differentiation from miPSCs was established with ideal concentration (10 ng/mL) and duration (from day 3 to day 14) of CT-1 administration. Up-regulated expression of cardiac specific genes that accounted for embryonic cardiogenesis was observed by quantitative RT-PCR. Elevated amount of α-myosin heavy chain (α-MHC) and cardiac troponin I (cTn I) positive cells were detected by immunofluorescence staining and flow cytometry analysis in CT-1 group. RESULTS Transmission electron microscopic analysis revealed that cells treated with CT-1 showed better organized sacromeric structure and more mitochondria, which are morphological characteristic of matured cardiomyocytes. Western blot demonstrated that CT-1 promotes cardiomyocyte differentiation from miPSCs partly via JAK2/STAT3/Pim-1 pathway as compared with control group. CONCLUSIONS These findings suggested that CT-1 could enhance the cardiomyocyte differentiation as well as the maturation of mouse induced pluripotent stem cell derived cardiomyocytes by regulating JAK2/STAT3/Pim-1signaling pathway.
Collapse
|
36
|
Hung HC, Lu FH, Ou HY, Wu HT, Wu JS, Yang YC, Chang CJ. Cardiotrophin-1 is associated with increased risk of arterial stiffness. Biomark Med 2015; 9:123-30. [PMID: 25689900 DOI: 10.2217/bmm.14.87] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
AIM Cardiotrophin-1 null mice presented decreased arterial stiffness. The aim of this study is to investigate the relationship between cardiotrophin-1 and arterial stiffness, assessed by brachial-ankle pulse-wave velocity (baPWV). METHODS We enrolled 300 subjects, 200 with baPWV >1400 and 100 with baPWV ≤1400 cm/s. RESULTS Cardiotrophin-1 levels were significantly higher in subjects with baPWV >1400 than those with baPWV ≤1400 cm/s. The multivariate logistic regression analysis showed that age, prehypertension, hypertension and cardiotrophin-1 were independently associated with baPWV >1400 cm/s after adjusting for gender, obesity, diabetes, homeostasis model assessment-insulin resistance, total cholesterol, high-density lipoprotein cholesterol, triglyceride, high-sensitivity C-reactive protein, creatinine, smoking and habitual exercise. CONCLUSION Cardiotrophin-1 is positively related to baPWV independent of traditional cardiometabolic risk factors for arterial stiffness.
Collapse
Affiliation(s)
- Hao-Chang Hung
- Division of Endocrinology & Metabolism, Department of Internal Medicine, National Cheng Kung University Hospital, Tainan, Taiwan
| | | | | | | | | | | | | |
Collapse
|
37
|
Ohtsuka S, Nakai-Futatsugi Y, Niwa H. LIF signal in mouse embryonic stem cells. JAKSTAT 2015; 4:e1086520. [PMID: 27127728 PMCID: PMC4802755 DOI: 10.1080/21623996.2015.1086520] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 08/18/2015] [Indexed: 12/22/2022] Open
Abstract
Since the establishment of mouse embryonic stem cells (mESCs) in the 1980s, a number of important notions on the self-renewal of pluripotent stem cells in vitro have been found. In serum containing conventional culture, an exogenous cytokine, leukemia inhibitory factor (LIF), is absolutely essential for the maintenance of pluripotency. In contrast, in serum-free culture with simultaneous inhibition of Map-kinase and Gsk3 (so called 2i-culture), LIF is no longer required. However, recent findings also suggest that LIF may have a role not covered by the 2i for the maintenance of naïve pluripotency. These suggest that LIF functions for the maintenance of naïve pluripotency in a context dependent manner. We summarize how LIF-signal pathway is converged to maintain the naïve state of pluripotency.
Collapse
Affiliation(s)
- Satoshi Ohtsuka
- Laboratory for Pluripotent Stem Cell Studies; Center for Developmental Biology (CDB) RIKEN ; Kobe, Japan
| | - Yoko Nakai-Futatsugi
- Laboratory for Pluripotent Stem Cell Studies; Center for Developmental Biology (CDB) RIKEN ; Kobe, Japan
| | - Hitoshi Niwa
- Laboratory for Pluripotent Stem Cell Studies; Center for Developmental Biology (CDB) RIKEN; Kobe, Japan; Department of Pluripotent Stem Cell Biology; Institute of Molecular Embryology and Genetics (IMEG); Kumamoto University; Kumamoto, Japan
| |
Collapse
|
38
|
Hu Q, Huang C, Wang Y, Wu R. Expression of leukemia inhibitory factor in the rat retina following acute ocular hypertension. Mol Med Rep 2015; 12:6577-83. [PMID: 26352383 PMCID: PMC4626123 DOI: 10.3892/mmr.2015.4287] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 05/27/2015] [Indexed: 12/25/2022] Open
Abstract
The aim of the present study was to investigate the expression of leukemia inhibitory factor (LIF) and its downstream signaling pathways in the rat retina following acute ocular hypertension. The intraocular pressure of the rats was elevated to 110 mmHg for 1 h by infusing the anterior chamber with normal saline. The retinal tissues were obtained 12 h, 24 h, and 2, 3 and 7 days after termination of the ocular hypertension. Hematoxylin and eosin and terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining were performed to assess the morphological changes and the apoptosis of retinal cells, respectively. Quantification of the retinal ganglion cells (RGCs) was performed using fluorogold retrograde (FG) staining. The expression levels of LIF, LIF receptor (LIFR), signal transducers and activators of transcription 3 (STAT3), phosphorylated STAT3 (P-STAT3), Akt, phosphorylated-Akt (P-Akt), extracellular signal-regulated kinase (ERK) and phosphorylated ERK (P-ERK) were determined at different time-points following acute ocular hypertension using western blot analysis. Reverse transcription-quantitative polymerase chain reaction was performned to detect the mRNA expression levels of LIF and LIFR. The results revealed that 12 h, 24 h, 2, 3 and 7 days after reperfusion, the thickness of the inner nuclear layer and the inner plexiform layer was decreased, with a significant reduction in the number of RGCs, as determined using TUNEL and FG staining. The expression levels of LIF and LIFR were increased following acute ocular hypertension. At 12 h post-retinal reperfusion, the expression levels of P-STAT3 and P-Akt were significantly upregulated, while the expression of P-ERK was decreased. The changes in the expression levels of LIF and LIFR suggested that LIF may be important in the process of degeneration/protection following retinal ischemia induced by acute ocular hypertension, via activation of the Janus kinase/STAT and Akt signaling pathways.
Collapse
Affiliation(s)
- Qianqian Hu
- Department of Glaucoma, Xiamen University Affiliated Eye Center, Xiamen, Fujian 361001, P.R. China
| | - Changquan Huang
- Department of Glaucoma, Xiamen University Affiliated Eye Center, Xiamen, Fujian 361001, P.R. China
| | - Yao Wang
- Department of Glaucoma, Xiamen University Affiliated Eye Center, Xiamen, Fujian 361001, P.R. China
| | - Renyi Wu
- Department of Glaucoma, Xiamen University Affiliated Eye Center, Xiamen, Fujian 361001, P.R. China
| |
Collapse
|
39
|
López-Yoldi M, Moreno-Aliaga MJ, Bustos M. Cardiotrophin-1: A multifaceted cytokine. Cytokine Growth Factor Rev 2015; 26:523-32. [PMID: 26188636 DOI: 10.1016/j.cytogfr.2015.07.009] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 07/01/2015] [Indexed: 10/23/2022]
Abstract
Cardiotrophin-1 (CT-1) is a member of the gp130 family of cytokines that have pleiotropic functions on different tissues and cell types. Although many effects of CT-1 have been described on the heart, there is an extensive research showing important protective effects in other organs such as liver, kidney or nervous system. Recently, several studies have pointed out that CT-1 might also play a key role in the regulation of body weight and intermediate metabolism. This paper will review many aspects of CT-1 physiological role in several organs and discuss data for consideration in therapeutic approaches.
Collapse
Affiliation(s)
- Miguel López-Yoldi
- Department of Nutrition, Food Science and Physiology, University of Navarra, Pamplona, Spain; Centre for Nutrition Research, University of Navarra, Pamplona, Spain
| | - María J Moreno-Aliaga
- Department of Nutrition, Food Science and Physiology, University of Navarra, Pamplona, Spain; Centre for Nutrition Research, University of Navarra, Pamplona, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III (ISCIII), Madrid, Spain; Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Matilde Bustos
- Area of Hepatology and Gene Therapy, CIMA (Center for Applied Medical Research) University of Navarra, Pamplona, Spain.
| |
Collapse
|
40
|
Stimulation of cardiomyogenesis from mouse embryonic stem cells by nuclear translocation of cardiotrophin-1. Int J Cardiol 2015; 193:23-33. [PMID: 26005169 DOI: 10.1016/j.ijcard.2015.05.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 05/05/2015] [Accepted: 05/06/2015] [Indexed: 01/07/2023]
Abstract
BACKGROUND Cardiotrophin-1 (CT-1) controls cardiomyogenesis of mouse embryonic stem (ES) cells. OBJECTIVES To investigate the signaling pathway underlying the action of CT-1 on cardiac cell differentiation. METHODS Protein expression was analyzed by western blot technique and cardiac areas by immunohistochemistry. Calcium, reactive oxygen species (ROS) and nitric oxide (NO) were assessed by microfluorometry using fluo-4, H2DCF, and DAF-2DA, respectively. Gene inactivation of CT-1 was achieved by siRNA technology. RESULTS CT-1 as well as its receptor gp 130 were transiently upregulated during differentiation of ES cells. Exogenous CT-1 enhanced cardiomyogenesis, increased the cardiac transcription factors MEF2c, Nkx-2.5, TEAD3 and GATA4, the cardiac proteins α-actinin, MLC2a, MYH7, MLC1a, MLC2v and HCN4 as well as vascular endothelial growth factor (VEGF), platelet-derived growth factor-BB (PDGF-BB), fibroblast growth factor-2 (FGF-2) and atrial natriuretic peptide (ANP). CT-1 downregulation by small interfering RNA (siRNA) inhibited cardiomyogenesis and decreased VEGF, PDGF-BB, FGF-2 and ANP expression. CT-1 raised intracellular calcium which was abolished by the intracellular calcium chelator BAPTA, AM and thapsigargin. Moreover, CT-1 treatment increased ROS, followed by NO generation and NOS3 activation. During ES cell differentiation CT-1 was translocated to the cell nucleus. Exogenous CT-1 induced nuclear translocation of endogenous CT-1, which was inhibited by BAPTA, the NOS inhibitor L-N(G)-Nitroarginine methyl ester (l-NAME), the radical scavenger N-(2-mercaptopropionyl)-glycine (NMPG) as well as the janus kinase 2 (JAK2) inhibitor AG490 and the PI3 kinase (PI3K) inhibitor LY294002. CONCLUSIONS Nuclear translocation of CT-1 regulates cardiomyogenesis of ES cells and involves calcium, NO, ROS as well as CT-1 regulated signaling pathways.
Collapse
|
41
|
Hua X, Shan Y, Li D, Xu D, Zhang J, Yang T, Han L, Shen C, Xia Y, Chen Q, Ma X, Zhang J, Xia Q. A Potential Profibrogenic Role of Biliary Epithelium-Derived Cardiotrophin-1 in Pediatric Cholestatic Liver Disease. J Interferon Cytokine Res 2015; 35:606-12. [PMID: 25919795 DOI: 10.1089/jir.2014.0128] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
As a cytokine of the interleukin-6 family, cardiotrophin-1 (CT-1) has been shown to be an important endogenous protector in liver injury. Our study aimed to investigate the role of CT-1 in liver fibrosis in pediatric cholestatic liver disease (PCLD). CT-1 mRNA and protein expression levels were upregulated in PCLD liver biopsy tissues compared with controls. Immunohistochemistry and confocal microscopy of liver sections showed that CT-1 was predominantly expressed by biliary epithelium cells. Serum CT-1 was elevated significantly in the children with PCLD compared with controls. Serum CT-1 levels exhibited a moderate positive correlation with the Scheuer stage of hepatic fibrosis and serum TB levels and a weak correlation with serum ALP levels. In vitro analysis indicated that LX-2 cells preconditioned with CT-1 exhibited significant increments in proliferation and accumulation of extracellular matrix components, while also positively regulating the STAT3 and p38MAPK pathways. In conclusion, biliary epithelium-derived CT-1 may exert a profibrogenic potential in PCLD.
Collapse
Affiliation(s)
- Xiangwei Hua
- 1 Department of Liver Surgery and Liver Transplantation Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University , Shanghai, People's Republic of China
| | - Yuhua Shan
- 1 Department of Liver Surgery and Liver Transplantation Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University , Shanghai, People's Republic of China
| | - Dawei Li
- 1 Department of Liver Surgery and Liver Transplantation Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University , Shanghai, People's Republic of China
| | - Dongwei Xu
- 1 Department of Liver Surgery and Liver Transplantation Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University , Shanghai, People's Republic of China
| | - Jiang Zhang
- 1 Department of Liver Surgery and Liver Transplantation Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University , Shanghai, People's Republic of China
| | - Taihua Yang
- 1 Department of Liver Surgery and Liver Transplantation Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University , Shanghai, People's Republic of China
| | - Longzhi Han
- 1 Department of Liver Surgery and Liver Transplantation Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University , Shanghai, People's Republic of China
| | - Conghuan Shen
- 1 Department of Liver Surgery and Liver Transplantation Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University , Shanghai, People's Republic of China
| | - Yun Xia
- 1 Department of Liver Surgery and Liver Transplantation Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University , Shanghai, People's Republic of China
| | - Qimin Chen
- 2 Department of Urology, Shanghai Children's Medical Center Affiliated to Shanghai Jiao Tong University School of Medicine , Shanghai, People's Republic of China
| | - Xiong Ma
- 3 Division of Gastroenterology and Hepatology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University , Shanghai Institute of Digestive Disease, Shanghai, People's Republic of China .,4 Key Laboratory of Gastroenterology & Hepatology, Ministry of Health (Shanghai Jiao-Tong University) , Shanghai, People's Republic of China
| | - Jianjun Zhang
- 1 Department of Liver Surgery and Liver Transplantation Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University , Shanghai, People's Republic of China
| | - Qiang Xia
- 1 Department of Liver Surgery and Liver Transplantation Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University , Shanghai, People's Republic of China
| |
Collapse
|
42
|
Fontes JA, Rose NR, Čiháková D. The varying faces of IL-6: From cardiac protection to cardiac failure. Cytokine 2015; 74:62-8. [PMID: 25649043 DOI: 10.1016/j.cyto.2014.12.024] [Citation(s) in RCA: 233] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Revised: 12/19/2014] [Accepted: 12/22/2014] [Indexed: 02/08/2023]
Abstract
IL6 is a pleiotropic cytokine that is made in response to perturbations in homeostasis. IL6 becomes elevated in the acute response to host injury and can activate immune cells, direct immune cell trafficking, signal protective responses in local tissue, initial the acute phase response or initiate wound healing. In the short term this proinflammatory response is protective and limits host damage. It is when this acute response remains chronically activated that IL6 becomes pathogenic to the host. Chronically elevated IL6 levels lead to chronic inflammation and fibrotic disorders. The heart is a tissue where this temporal regulation of IL6 is very apparent. Studies from myocardial infarction show how short-term IL6 signaling can protect and preserve the heart tissue in response to acute damage, where long term IL6 signaling or an over-production of IL6R protein plays a causal role in cardiovascular disease. Thus, IL6 can be both protective and pathogenic, depending on the kinetics of the host response.
Collapse
Affiliation(s)
- Jillian A Fontes
- William H. Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, United States
| | - Noel R Rose
- William H. Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, United States; Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
| | - Daniela Čiháková
- William H. Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, United States; Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States.
| |
Collapse
|
43
|
Morgani SM, Brickman JM. LIF supports primitive endoderm expansion during pre-implantation development. Development 2015; 142:3488-99. [DOI: 10.1242/dev.125021] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 08/19/2015] [Indexed: 01/31/2023]
Abstract
Embryonic stem cells (ESCs) are pluripotent cell lines that can be maintained indefinitely in an early developmental state. ESC culture conditions almost all require the cytokine LIF to maintain self-renewal. As ESCs are not homogeneous, but contain multiple populations reminiscent of the blastocyst, identifying the target cells of LIF is necessary to understand the propagation of pluripotency. We recently found that LIF acts under self-renewing conditions to stimulate the fraction of ESCs that express extraembryonic markers, but has little impact on pluripotent gene expression. Here we report that LIF has two distinct roles. It blocks early epiblast differentiation and supports the expansion of primitive endoderm (PrE) primed ESCs and PrE in vivo. We find that activation of JAK/STAT signalling downstream of LIF occurs initially throughout the pre-implantation embryo, but later marks the PrE. Moreover, the addition of LIF to cultured embryos increases the GATA6+ PrE population while inhibition of JAK/STAT reduces both NANOG+ epiblast (Epi) and GATA6+ PrE. The reduction of the NANOG+ Epi may be explained by its precocious differentiation to later Epi derivatives, while the increase in PrE is mediated both by an increase in proliferation and inhibition of PrE apoptosis that is normally triggered in embryos with an excess of GATA6+ cells. Thus, it appears that the relative size of the PrE is determined by the number of LIF-producing cells in the embryo. This suggests a mechanism by which the embryo adjusts the relative ratio of the primary lineages in response to experimental manipulation.
Collapse
Affiliation(s)
- Sophie M. Morgani
- The Danish Stem Cell Centre - DanStem, University of Copenhagen, 3B Blegdamsvej, DK-2200 Copenhagen N, Denmark
| | - Joshua M. Brickman
- The Danish Stem Cell Centre - DanStem, University of Copenhagen, 3B Blegdamsvej, DK-2200 Copenhagen N, Denmark
| |
Collapse
|
44
|
López-Yoldi M, Fernández-Galilea M, Laiglesia LM, Larequi E, Prieto J, Martínez JA, Bustos M, Moreno-Aliaga MJ. Cardiotrophin-1 stimulates lipolysis through the regulation of main adipose tissue lipases. J Lipid Res 2014; 55:2634-43. [PMID: 25351614 DOI: 10.1194/jlr.m055335] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cardiotrophin-1 (CT-1) is a cytokine with antiobesity properties and with a role in lipid metabolism regulation and adipose tissue function. The aim of this study was to analyze the molecular mechanisms involved in the lipolytic actions of CT-1 in adipocytes. Recombinant CT-1 (rCT-1) effects on the main proteins and signaling pathways involved in the regulation of lipolysis were evaluated in 3T3-L1 adipocytes and in mice. rCT-1 treatment stimulated basal glycerol release in a concentration- and time-dependent manner in 3T3-L1 adipocytes. rCT-1 (20 ng/ml for 24 h) raised cAMP levels, and in parallel increased protein kinase (PK)A-mediated phosphorylation of perilipin and hormone sensitive lipase (HSL) at Ser660. siRNA knock-down of HSL or PKA, as well as pretreatment with the PKA inhibitor H89, blunted the CT-1-induced lipolysis, suggesting that the lipolytic action of CT-1 in adipocytes is mainly mediated by activation of HSL through the PKA pathway. In ob/ob mice, acute rCT-1 treatment also promoted PKA-mediated phosphorylation of perilipin and HSL at Ser660 and Ser563, and increased adipose triglyceride lipase (desnutrin) content in adipose tissue. These results showed that the ability of CT-1 to regulate the activity of the main lipases underlies the lipolytic action of this cytokine in vitro and in vivo, and could contribute to CT-1 antiobesity effects.
Collapse
Affiliation(s)
- Miguel López-Yoldi
- Departments of Nutrition, Food Science, and Physiology University of Navarra, Pamplona, Navarra, Spain Centre for Nutrition Research, University of Navarra, Pamplona, Navarra, Spain
| | - Marta Fernández-Galilea
- Departments of Nutrition, Food Science, and Physiology University of Navarra, Pamplona, Navarra, Spain
| | - Laura M Laiglesia
- Departments of Nutrition, Food Science, and Physiology University of Navarra, Pamplona, Navarra, Spain Centre for Nutrition Research, University of Navarra, Pamplona, Navarra, Spain
| | - Eduardo Larequi
- Gene Therapy and Hepatology, CIMA, University of Navarra, Pamplona, Navarra, Spain
| | - Jesús Prieto
- Gene Therapy and Hepatology, CIMA, University of Navarra, Pamplona, Navarra, Spain CIBERehd Institute of Health Carlos III, Madrid, Spain
| | - J Alfredo Martínez
- Departments of Nutrition, Food Science, and Physiology University of Navarra, Pamplona, Navarra, Spain Centre for Nutrition Research, University of Navarra, Pamplona, Navarra, Spain CIBERobn, Physiopathology of Obesity and Nutrition, Institute of Health Carlos III, Madrid, Spain
| | - Matilde Bustos
- Gene Therapy and Hepatology, CIMA, University of Navarra, Pamplona, Navarra, Spain
| | - Maria J Moreno-Aliaga
- Departments of Nutrition, Food Science, and Physiology University of Navarra, Pamplona, Navarra, Spain Centre for Nutrition Research, University of Navarra, Pamplona, Navarra, Spain CIBERobn, Physiopathology of Obesity and Nutrition, Institute of Health Carlos III, Madrid, Spain
| |
Collapse
|
45
|
Lutz SZ, Franck O, Böhm A, Machann J, Schick F, Machicao F, Fritsche A, Häring HU, Staiger H. Common genetic variation in the human CTF1 locus, encoding cardiotrophin-1, determines insulin sensitivity. PLoS One 2014; 9:e100391. [PMID: 25025664 PMCID: PMC4099130 DOI: 10.1371/journal.pone.0100391] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Accepted: 05/23/2014] [Indexed: 12/02/2022] Open
Abstract
Aims/Hypothesis Recently, cardiotrophin-1, a member of the interleukin-6 family of cytokines was described to protect beta-cells from apoptosis, to improve glucose-stimulated insulin secretion and insulin resistance, and to prevent streptozotocin-induced diabetes in mice. Here, we studied whether common single nucleotide polymorphisms (SNPs) in the CTF1 locus, encoding cardiotrophin-1, influence insulin secretion and insulin sensitivity in humans. Methods We genotyped 1,771 German subjects for three CTF1 tagging SNPs (rs1046276, rs1458201, and rs8046707). The subjects were metabolically characterized by an oral glucose tolerance test. Subgroups underwent magnetic resonance (MR) imaging/spectroscopy and hyperinsulinaemic-euglycaemic clamps. Results After appropriate adjustment, the minor allele of CTF1 SNP rs8046707 was significantly associated with decreased in vivo measures of insulin sensitivity. The other tested SNPs were not associated with OGTT-derived sensitivity parameters, nor did the three tested SNPs show any association with OGTT-derived parameters of insulin release. In the MR subgroup, SNP rs8046707 was nominally associated with lower visceral adipose tissue. Furthermore, the SNP rs1458201 showed a nominal association with increased VLDL levels. Conclusions In conclusion, this study, even though preliminary and awaiting further confirmation by independent replication, provides first evidence that common genetic variation in CTF1 could contribute to insulin sensitivity in humans. Our SNP data indicate an insulin-desensitizing effect of cardiotrophin-1 and underline that cardiotrophin-1 represents an interesting target to influence insulin sensitivity.
Collapse
Affiliation(s)
- Stefan Z. Lutz
- Department of Internal Medicine, Division of Endocrinology, Diabetology, Vascular Disease, Nephrology and Clinical Chemistry, University of Tübingen, Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Centre Munich at the University of Tübingen, Tübingen, Germany
- German Centre for Diabetes Research (DZD), Tübingen, Germany
| | - Olga Franck
- Department of Internal Medicine, Division of Endocrinology, Diabetology, Vascular Disease, Nephrology and Clinical Chemistry, University of Tübingen, Tübingen, Germany
| | - Anja Böhm
- Department of Internal Medicine, Division of Endocrinology, Diabetology, Vascular Disease, Nephrology and Clinical Chemistry, University of Tübingen, Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Centre Munich at the University of Tübingen, Tübingen, Germany
- German Centre for Diabetes Research (DZD), Tübingen, Germany
| | - Jürgen Machann
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Centre Munich at the University of Tübingen, Tübingen, Germany
- German Centre for Diabetes Research (DZD), Tübingen, Germany
- Department of Diagnostic and Interventional Radiology, Section on Experimental Radiology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Fritz Schick
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Centre Munich at the University of Tübingen, Tübingen, Germany
- German Centre for Diabetes Research (DZD), Tübingen, Germany
- Department of Diagnostic and Interventional Radiology, Section on Experimental Radiology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Fausto Machicao
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Centre Munich at the University of Tübingen, Tübingen, Germany
- German Centre for Diabetes Research (DZD), Tübingen, Germany
| | - Andreas Fritsche
- Department of Internal Medicine, Division of Endocrinology, Diabetology, Vascular Disease, Nephrology and Clinical Chemistry, University of Tübingen, Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Centre Munich at the University of Tübingen, Tübingen, Germany
- German Centre for Diabetes Research (DZD), Tübingen, Germany
- Department of Internal Medicine, Division of Nutritional and Preventive Medicine, University of Tübingen, Tübingen, Germany
| | - Hans-Ulrich Häring
- Department of Internal Medicine, Division of Endocrinology, Diabetology, Vascular Disease, Nephrology and Clinical Chemistry, University of Tübingen, Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Centre Munich at the University of Tübingen, Tübingen, Germany
- German Centre for Diabetes Research (DZD), Tübingen, Germany
- * E-mail:
| | - Harald Staiger
- Department of Internal Medicine, Division of Endocrinology, Diabetology, Vascular Disease, Nephrology and Clinical Chemistry, University of Tübingen, Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Centre Munich at the University of Tübingen, Tübingen, Germany
- German Centre for Diabetes Research (DZD), Tübingen, Germany
| |
Collapse
|
46
|
Wagner MA, Siddiqui MAQ. The JAK-STAT pathway in hypertrophic stress signaling and genomic stress response. JAKSTAT 2014; 1:131-41. [PMID: 24058762 PMCID: PMC3670293 DOI: 10.4161/jkst.20702] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The JAK-STAT signaling pathway plays a central role in transducing stress and growth signals in the hypertrophic heart. Unlike most signal transducers, JAKs and STATs signal in a number of different ways, both within the JAK-STAT pathway and in collaboration with other signaling pathways. In this review, we discuss how IL-6 activates cells lacking IL-6 receptors through trans-signaling and examine JAK-STAT pathway interaction with GPCR-linked pathways both within and between cells. Finally, we discuss recent studies showing how the JAK-STAT pathway can intersect with a general transcriptional regulatory mechanism to effect transcription of STAT-dependent stress response genes.
Collapse
Affiliation(s)
- Michael A Wagner
- Department of Cell Biology; Center for Cardiovascular and Muscle Research; State University of New York Downstate Medical Center; Brooklyn, NY USA
| | | |
Collapse
|
47
|
The States of Pluripotency: Pluripotent Lineage Development in the Embryo and in the Dish. ACTA ACUST UNITED AC 2014. [DOI: 10.1155/2014/208067] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The pluripotent cell lineage of the embryo comprises a series of temporally and functionally distinct intermediary cell states, the epiblast precursor cell of the newly formed blastocyst, the epiblast population of the inner cell mass, and the early and late epiblast of the postimplantation embryo, referred to here as early and late primitive ectoderm. Pluripotent cell populations representative of the embryonic populations can be formed in culture. Although multiple pluripotent cell states are now recognised, little is known about the signals and pathways that progress cells from the epiblast precursor cell to the late primitive ectoderm in the embryo or in culture. The characterisation of cell states is most advanced in mouse where conditions for culturing distinct pluripotent cell states are well established and embryonic material is accessible. This review will focus on the pluripotent cell states present during embryonic development in the mouse and what is known of the mechanisms that regulate the progression of the lineage from the epiblast precursor cell and the ground state of pluripotency to the late primitive ectoderm present immediately prior to cell differentiation.
Collapse
|
48
|
Kobayashi R, Terakawa J, Kato Y, Azimi S, Inoue N, Ohmori Y, Hondo E. The contribution of leukemia inhibitory factor (LIF) for embryo implantation differs among strains of mice. Immunobiology 2014; 219:512-21. [PMID: 24698551 DOI: 10.1016/j.imbio.2014.03.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2014] [Revised: 03/12/2014] [Accepted: 03/12/2014] [Indexed: 12/20/2022]
Abstract
Despite of the claim that maternal leukemia inhibitory factor (LIF) - a member of interleukin 6 (IL6) family of cytokines - plays indispensable roles for murine embryo implantation, these roles remain undefined in humans because the potency of LIF on implantation appears to vary among individuals. Here, we showed that the contribution of LIF for murine implantation was dependent on the strains of mice (ICR, C57BL/6J (B6), ddY, BALB/c, DBA/2Cr and MF1 strains). Inhibition of LIF during the implantation period caused severe disruption of embryo implantation in B6 and MF1 strains. Implantation was partly disrupted in other strains, but some embryos were implanted successfully. We speculated that other IL6 family members compensate for LIF actions on implantation in ICR, ddY, BALB/c, and DBA/2Cr strains. Indeed, the expression level of Ctf1 was upregulated by blockage of LIF function. CT-1 (encoded by Ctf1) treatment induced successful implantation without LIF in delayed implantation mice (ICR and B6) via phosphorylation of the signal transducer and activator of transcription 3 (STAT3) in the uterine luminal epithelium. Simultaneous inhibition of LIF and CT-1 did not block implantation completely in ICR mice, indicating that embryo implantation in this strain was robustly protected by LIF, CT-1 and other potential STAT3 activators. The present study might provide an explanation for the individual variation in the potency of LIF for embryo implantation in humans.
Collapse
Affiliation(s)
- Ryosuke Kobayashi
- Laboratory of Animal Morphology, Division of Biofunctional Development, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan
| | - Jumpei Terakawa
- Laboratory of Animal Morphology, Division of Biofunctional Development, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan
| | - Yasumasa Kato
- Department of Oral Function and Molecular Biology, Ohu University School of Dentistry, 963-8611 Koriyama, Japan
| | - Shafiqullah Azimi
- Laboratory of Animal Morphology, Division of Biofunctional Development, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan
| | - Naoko Inoue
- Laboratory of Animal Morphology, Division of Biofunctional Development, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan
| | - Yasushige Ohmori
- Laboratory of Animal Morphology, Division of Biofunctional Development, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan
| | - Eiichi Hondo
- Laboratory of Animal Morphology, Division of Biofunctional Development, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan.
| |
Collapse
|
49
|
O'Reilly S, Ciechomska M, Cant R, van Laar JM. Interleukin-6 (IL-6) trans signaling drives a STAT3-dependent pathway that leads to hyperactive transforming growth factor-β (TGF-β) signaling promoting SMAD3 activation and fibrosis via Gremlin protein. J Biol Chem 2014; 289:9952-60. [PMID: 24550394 DOI: 10.1074/jbc.m113.545822] [Citation(s) in RCA: 188] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Fibrosis is a common and intractable condition associated with various pathologies. It is characterized by accumulation of an excessive amount of extracellular matrix molecules that primarily include collagen type I. IL-6 is a profibrotic cytokine that is elevated in the prototypic fibrotic autoimmune condition systemic sclerosis and is known to induce collagen I expression, but the mechanism(s) behind this induction are currently unknown. Using healthy dermal fibroblasts in vitro, we analyzed the signaling pathways that underscore the IL-6-mediated induction of collagen. We show that IL-6 trans signaling is important and that the effect is dependent on STAT3; however, the effect is indirect and mediated through enhanced TGF-β signaling and the classic downstream cellular mediator Smad3. This is due to induction of the bone morphogenetic protein (BMP) antagonist Gremlin-1, and we show that Gremlin-1 is profibrotic and is mediated through canonical TGF-β signaling.
Collapse
Affiliation(s)
- Steven O'Reilly
- From the Musculoskeletal Research Group, Institute of Cellular Medicine, 4th Floor Cookson Building, Framlington Place, Newcastle upon Tyne NE2 4HH, United Kingdom
| | | | | | | |
Collapse
|
50
|
Moreno MU, San José G, Pejenaute Á, Landecho MF, Díez J, Beloqui Ó, Fortuño A, Zalba G. Association of phagocytic NADPH oxidase activity with hypertensive heart disease: a role for cardiotrophin-1? Hypertension 2013; 63:468-74. [PMID: 24324051 DOI: 10.1161/hypertensionaha.113.01470] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Left ventricular hypertrophy (LVH) is an independent marker of mortality in hypertension. Although the mechanisms contributing to LVH are complex, inflammation and oxidative stress may favor its development. We analyzed the association of the phagocytic NADPH oxidase-mediated superoxide anion release and LVH in patients with essential hypertension and the role of cardiotrophin-1 (CT-1) and interleukin-6 (IL-6), cytokines implicated in cardiac growth. Blood pressure, echocardiography data, and serum CT-1 and IL-6 levels were obtained in 140 subjects: 18 normotensives without LVH, 42 hypertensives without LVH, and 80 hypertensives with LVH. The NADPH oxidase-dependent superoxide production was assessed by chemiluminescence in peripheral blood mononuclear cells. Peripheral blood mononuclear cells were stimulated with CT-1 in vitro. Superoxide anion production by peripheral blood mononuclear cells associated with LVH and correlated with the left ventricular mass index. Serum CT-1 and IL-6 levels, which associated with the left ventricular mass index, correlated with superoxide production. Serum CT-1 and IL-6 levels were correlated. CT-1 stimulated NADPH oxidase superoxide production in peripheral blood mononuclear cells, which resulted in an increased release of IL-6. Our results show that superoxide anion production by the phagocytic NADPH oxidase associates with hypertensive heart disease, being significantly enhanced in hypertensive patients with LVH. This may be attributable to the activation of the NADPH oxidase by CT-1 and the subsequent release of IL-6. The phagocytic NADPH oxidase may be a therapeutic target in hypertensive heart disease.
Collapse
Affiliation(s)
- María U Moreno
- Department of Biochemistry and Genetics, Irunlarrea 1, 31008-Pamplona, Spain.
| | | | | | | | | | | | | | | |
Collapse
|