1
|
Marinho Y, Villarreal ES, Aboagye SY, Williams DL, Sun J, Silva CLM, Lutz SE, Oliveira SD. Schistosomiasis-associated pulmonary hypertension unveils disrupted murine gut-lung microbiome and reduced endoprotective Caveolin-1/BMPR2 expression. Front Immunol 2023; 14:1254762. [PMID: 37908354 PMCID: PMC10613683 DOI: 10.3389/fimmu.2023.1254762] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 09/22/2023] [Indexed: 11/02/2023] Open
Abstract
Schistosomiasis-associated Pulmonary Arterial Hypertension (Sch-PAH) is a life-threatening complication of chronic S. mansoni infection that can lead to heart failure and death. During PAH, the expansion of apoptosis-resistant endothelial cells (ECs) has been extensively reported; however, therapeutic approaches to prevent the progression or reversal of this pathological phenotype remain clinically challenging. Previously, we showed that depletion of the anti-apoptotic protein Caveolin-1 (Cav-1) by shedding extracellular vesicles contributes to shifting endoprotective bone morphogenetic protein receptor 2 (BMPR2) towards transforming growth factor beta (TGF-β)-mediated survival of an abnormal EC phenotype. However, the mechanism underlying the reduced endoprotection in PAH remains unclear. Interestingly, recent findings indicate that, similar to the gut, healthy human lungs are populated by diverse microbiota, and their composition depends significantly on intrinsic and extrinsic host factors, including infection. Despite the current knowledge that the disruption of the gut microbiome contributes to the development of PAH, the role of the lung microbiome remains unclear. Thus, using a preclinical animal model of Sch-PAH, we tested whether S. mansoni infection alters the gut-lung microbiome composition and causes EC injury, initiating the expansion of an abnormal EC phenotype observed in PAH. Indeed, in vivo stimulation with S. mansoni eggs significantly altered the gut-lung microbiome profile, in addition to promoting injury to the lung vasculature, characterized by increased apoptotic markers and loss of endoprotective expression of lung Cav-1 and BMPR2. Moreover, S. mansoni egg stimulus induced severe pulmonary vascular remodeling, leading to elevated right ventricular systolic pressure and hypertrophy, characteristic of PAH. In vitro, exposure to the immunodominant S. mansoni egg antigen p40 activated TLR4/CD14-mediated transient phosphorylation of Cav-1 at Tyr14 in human lung microvascular EC (HMVEC-L), culminating in a mild reduction of Cav-1 expression, but failed to promote death and shedding of extracellular vesicles observed in vivo. Altogether, these data suggest that disruption of the host-associated gut-lung microbiota may be essential for the emergence and expansion of the abnormal lung endothelial phenotype observed in PAH, in addition to S. mansoni eggs and antigens.
Collapse
Affiliation(s)
- Ygor Marinho
- Vascular Immunobiology Lab, Department of Anesthesiology, College of Medicine, University of Illinois Chicago, Chicago, IL, United States
| | - Elizabeth S. Villarreal
- Vascular Immunobiology Lab, Department of Anesthesiology, College of Medicine, University of Illinois Chicago, Chicago, IL, United States
| | - Sammy Y. Aboagye
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, IL, United States
| | - David L. Williams
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, IL, United States
| | - Jun Sun
- Department of Medicine, College of Medicine, University of Illinois Chicago, Chicago, IL, United States
| | - Claudia L. M. Silva
- Molecular and Biochemical Pharmacology Lab, Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Sarah E. Lutz
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois Chicago, Chicago, IL, United States
| | - Suellen D. Oliveira
- Vascular Immunobiology Lab, Department of Anesthesiology, College of Medicine, University of Illinois Chicago, Chicago, IL, United States
- Vascular Immunobiology Lab, Department of Physiology and Biophysics, College of Medicine, University of Illinois Chicago, Chicago, IL, United States
| |
Collapse
|
2
|
TLR4-SIRT3 Mechanism Modulates Mitochondrial and Redox Homeostasis and Promotes EPCs Recruitment and Survival. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:1282362. [PMID: 35832490 PMCID: PMC9273456 DOI: 10.1155/2022/1282362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 06/10/2022] [Indexed: 11/30/2022]
Abstract
The low survival rate of endothelial progenitor cells (EPCs) in vivo which are susceptible to adverse microenvironments including inflammation and oxidative stress has become one primary challenge of EPCs transplantation for regenerative therapy. Recent studies reported functional expression of toll-like receptor (TLR) 4 on EPCs and dose-dependent effects of lipopolysaccharide (LPS) on cellular oxidative stress and angiogenic properties. However, the involved mechanism has not yet been elucidated well, and the influence of TLR4 signaling on EPCs survival and function in vivo is unknown. In the present study, we observed the effects of LPS and TLR4/SIRT3 on EPCs mitochondrial permeability and intracellular mitochondrial superoxide. We employed the monocrotaline-induced pulmonary arteriolar injury model to observe the effects of TLR4/SIRT3 on the recruitment and survival of transplanted EPCs. We found the destructive effects of 10 μg/mL LPS on mitochondrial homeostasis, and cellular viability was mediated by TLR4/SIRT3 signals at least partially, and the TLR4 mediates the early-stage recruitment of transplanted EPCs in pulmonary arteriolar inflammation injury; however, SIRT3 has more contribution to the survival of incorporated EPCs and ameliorated arteriolar remodeling in lung vascular tissue. The study provides insights for the critical role of TLR4/SIRT3 in LPS-induced oxidative stress and mitochondrial disorder in EPCs in vitro and in vivo. The TLR4/SIRT3 signaling is important for EPCs resistance against inflammation and oxidative stress and may represent a new manipulating target for developing efficient cell therapy strategy.
Collapse
|
3
|
Hudson J, Farkas L. Epigenetic Regulation of Endothelial Dysfunction and Inflammation in Pulmonary Arterial Hypertension. Int J Mol Sci 2021; 22:ijms222212098. [PMID: 34829978 PMCID: PMC8617605 DOI: 10.3390/ijms222212098] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/05/2021] [Accepted: 11/06/2021] [Indexed: 12/13/2022] Open
Abstract
Once perceived as a disorder treated by vasodilation, pulmonary artery hypertension (PAH) has emerged as a pulmonary vascular disease with severe endothelial cell dysfunction. In the absence of a cure, many studies seek to understand the detailed mechanisms of EC regulation to potentially create more therapeutic options for PAH. Endothelial dysfunction is characterized by complex phenotypic changes including unchecked proliferation, apoptosis-resistance, enhanced inflammatory signaling and metabolic reprogramming. Recent studies have highlighted the role of epigenetic modifications leading to pro-inflammatory response pathways, endothelial dysfunction, and the progression of PAH. This review summarizes the existing literature on epigenetic mechanisms such as DNA methylation, histone modifications, and non-coding RNAs, which can lead to aberrant endothelial function. Our goal is to develop a conceptual framework for immune dysregulation and epigenetic changes in endothelial cells in the context of PAH. These studies as well as others may lead to advances in therapeutics to treat this devastating disease.
Collapse
|
4
|
Ahmed M, Zaghloul N, Zimmerman P, Casanova NG, Sun X, Song JH, Hernon VR, Sammani S, Rischard F, Rafikova O, Rafikov R, Makino A, Kempf CL, Camp SM, Wang J, Desai AA, Lussier Y, Yuan JXJ, Garcia JG. Endothelial eNAMPT drives EndMT and preclinical PH: rescue by an eNAMPT-neutralizing mAb. Pulm Circ 2021; 11:20458940211059712. [PMID: 34790349 PMCID: PMC8591779 DOI: 10.1177/20458940211059712] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 10/25/2021] [Indexed: 12/03/2022] Open
Abstract
Pharmacologic interventions to halt/reverse the vascular remodeling and right ventricular dysfunction in pulmonary arterial hypertension (PAH) remains an unmet need. We previously demonstrated extracellular nicotinamide phosphoribosyltransferase (eNAMPT) as a DAMP (damage-associated molecular pattern protein) contributing to PAH pathobiology via TLR4 ligation. We examined the role of endothelial cell (EC)-specific eNAMPT in experimental PH and an eNAMPT-neutralizing mAb as a therapeutic strategy to reverse established PH. Hemodynamic/echocardiographic measurements and tissue analyses were performed in Sprague Dawley rats exposed to 10% hypoxia/Sugen (three weeks) followed by return to normoxia and weekly intraperitoneal delivery of the eNAMPT mAb (1 mg/kg). WT C57BL/6J mice and conditional EC-cNAMPTec-/- mice were exposed to 10% hypoxia (three weeks). Biochemical and RNA sequencing studies were performed on rat PH lung tissues and human PAH PBMCs. Hypoxia/Sugen-exposed rats exhibited multiple indices of severe PH (right ventricular systolic pressure, Fulton index), including severe vascular remodeling, compared to control rats. PH severity indices and plasma levels of eNAMPT, IL-6, and TNF-α were all significantly attenuated by eNAMPT mAb neutralization. Compared to hypoxia-exposed WT mice, cNAMPTec-/- KO mice exhibited significantly reduced PH severity and evidence of EC to mesenchymal transition (EndMT). Finally, biochemical and RNAseq analyses revealed eNAMPT mAb-mediated rectification of dysregulated inflammatory signaling pathways (TLR/NF-κB, MAP kinase, Akt/mTOR) and EndMT in rat PH lung tissues and human PAH PBMCs. These studies underscore EC-derived eNAMPT as a key contributor to PAH pathobiology and support the eNAMPT/TLR4 inflammatory pathway as a highly druggable therapeutic target to reduce PH severity and reverse PAH.
Collapse
Affiliation(s)
- Mohamed Ahmed
- Department of Pediatrics, University of Arizona Health Sciences, Tucson, AZ, USA
| | - Nahla Zaghloul
- Department of Pediatrics, University of Arizona Health Sciences, Tucson, AZ, USA
| | - Prisca Zimmerman
- Department of Pediatrics, University of Arizona Health Sciences, Tucson, AZ, USA
| | - Nancy G. Casanova
- Department of Medicine, University of Arizona Health Sciences, Tucson, AZ, USA
| | - Xiaoguang Sun
- Department of Medicine, University of Arizona Health Sciences, Tucson, AZ, USA
| | - Jin H. Song
- Department of Medicine, University of Arizona Health Sciences, Tucson, AZ, USA
| | - Vivian Reyes Hernon
- Department of Medicine, University of Arizona Health Sciences, Tucson, AZ, USA
| | - Saad Sammani
- Department of Medicine, University of Arizona Health Sciences, Tucson, AZ, USA
| | - Franz Rischard
- Department of Medicine, University of Arizona Health Sciences, Tucson, AZ, USA
| | - Olga Rafikova
- Department of Medicine, University of Arizona Health Sciences, Tucson, AZ, USA
| | - Ruslan Rafikov
- Department of Medicine, University of Arizona Health Sciences, Tucson, AZ, USA
| | - Ayako Makino
- Department of Medicine, University of Arizona Health Sciences, Tucson, AZ, USA
| | - Carrie L. Kempf
- Department of Medicine, University of Arizona Health Sciences, Tucson, AZ, USA
| | - Sara M. Camp
- Department of Medicine, University of Arizona Health Sciences, Tucson, AZ, USA
| | - Jian Wang
- Department of Medicine, University of Arizona Health Sciences, Tucson, AZ, USA
- State Key Laboratory of Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Ankit A. Desai
- Department of Medicine, University of Arizona Health Sciences, Tucson, AZ, USA
| | - Yves Lussier
- Department of Medicine, University of Arizona Health Sciences, Tucson, AZ, USA
| | - Jason X.-J. Yuan
- Department of Medicine, University of Arizona Health Sciences, Tucson, AZ, USA
| | - Joe G.N. Garcia
- Department of Medicine, University of Arizona Health Sciences, Tucson, AZ, USA
- State Key Laboratory of Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
5
|
Malnutrition, poor post-natal growth, intestinal dysbiosis and the developing lung. J Perinatol 2021; 41:1797-1810. [PMID: 33057133 DOI: 10.1038/s41372-020-00858-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 09/07/2020] [Accepted: 09/26/2020] [Indexed: 01/31/2023]
Abstract
In extremely preterm infants, poor post-natal growth, intestinal dysbiosis and bronchopulmonary dysplasia are common, and each is associated with long-term complications. The central hypothesis that this review will address is that these three common conditions are interrelated. Challenges to studying this hypothesis include the understanding that malnutrition and poor post-natal growth are not synonymous and that there is not agreement on what constitutes a normal intestinal microbiota in this evolutionarily new population. If this hypothesis is supported, further study of whether "correcting" intestinal dysbiosis in extremely preterm infants reduces postnatal growth restriction and/or bronchopulmonary dysplasia is indicated.
Collapse
|
6
|
Rudyk O, Aaronson PI. Redox Regulation, Oxidative Stress, and Inflammation in Group 3 Pulmonary Hypertension. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1303:209-241. [PMID: 33788196 DOI: 10.1007/978-3-030-63046-1_13] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Group 3 pulmonary hypertension (PH), which occurs secondary to hypoxia lung diseases, is one of the most common causes of PH worldwide and has a high unmet clinical need. A deeper understanding of the integrative pathological and adaptive molecular mechanisms within this group is required to inform the development of novel drug targets and effective treatments. The production of oxidants is increased in PH Group 3, and their pleiotropic roles include contributing to disease progression by promoting prolonged hypoxic pulmonary vasoconstriction and pathological pulmonary vascular remodeling, but also stimulating adaptation to pathological stress that limits the severity of this disease. Inflammation, which is increasingly being viewed as a key pathological feature of Group 3 PH, is subject to complex regulation by redox mechanisms and is exacerbated by, but also augments oxidative stress. In this review, we investigate aspects of this complex crosstalk between inflammation and oxidative stress in Group 3 PH, focusing on the redox-regulated transcription factor NF-κB and its upstream regulators toll-like receptor 4 and high mobility group box protein 1. Ultimately, we propose that the development of specific therapeutic interventions targeting redox-regulated signaling pathways related to inflammation could be explored as novel treatments for Group 3 PH.
Collapse
Affiliation(s)
- Olena Rudyk
- School of Cardiovascular Medicine & Sciences, King's College London, British Heart Foundation Centre of Research Excellence, London, UK.
| | - Philip I Aaronson
- School of Immunology and Microbial Sciences, King's College London, London, UK
| |
Collapse
|
7
|
Thomas C, Glinskii V, de Jesus Perez V, Sahay S. Portopulmonary Hypertension: From Bench to Bedside. Front Med (Lausanne) 2020; 7:569413. [PMID: 33224960 PMCID: PMC7670077 DOI: 10.3389/fmed.2020.569413] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 09/23/2020] [Indexed: 12/20/2022] Open
Abstract
Portopulmonary hypertension (PoPH) is defined as pulmonary arterial hypertension (PAH) associated with portal hypertension and is a subset of Group 1 pulmonary hypertension (PH). PoPH is a cause of significant morbidity and mortality in patients with portal hypertension with or without liver disease. Significant strides in elucidating the pathogenesis, effective screening algorithms, accurate diagnoses, and treatment options have been made in past 20 years. Survival of PoPH has remained poor compared to IPAH and other forms of PAH. Recently, the first randomized controlled trial was done in this patient population and showed promising results with PAH specific therapy. Despite positive effects on hemodynamics and functional outcomes, it is unclear whether PAH specific therapy has a beneficial effect on long term survival or transplant outcomes. In this review, we will discuss the epidemiology, pathophysiology, clinical and hemodynamic characteristics of PoPH. Additionally, this review will highlight the lacunae in our current management strategy, challenges faced and will provide direction to potentially useful futuristic management strategies.
Collapse
Affiliation(s)
- Christopher Thomas
- Division of Pulmonary, Allergy & Critical Care Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Vladimir Glinskii
- Division of Pulmonary, Allergy & Critical Care Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Vinicio de Jesus Perez
- Division of Pulmonary, Allergy & Critical Care Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Sandeep Sahay
- Houston Methodist Hospital Lung Center, Houston Methodist Hospital, Houston, TX, United States
| |
Collapse
|
8
|
Wong SK, Chin KY, Ima-Nirwana S. Toll-like Receptor as a Molecular Link between Metabolic Syndrome and Inflammation: A Review. Curr Drug Targets 2020; 20:1264-1280. [PMID: 30961493 DOI: 10.2174/1389450120666190405172524] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 03/29/2019] [Accepted: 04/01/2019] [Indexed: 02/07/2023]
Abstract
Metabolic Syndrome (MetS) involves a cluster of five conditions, i.e. obesity, hyperglycaemia, hypertension, hypertriglyceridemia and low High-Density Lipoprotein (HDL) cholesterol. All components of MetS share an underlying chronic inflammatory aetiology, manifested by increased levels of pro-inflammatory cytokines. The pathogenic role of inflammation in the development of MetS suggested that toll-like receptor (TLR) activation may trigger MetS. This review summarises the supporting evidence on the interactions between MetS and TLR activation, bridged by the elevation of TLR ligands during MetS. The regulatory circuits mediated by TLR activation, which modulates signal propagation, leading to the state of chronic inflammation, are also discussed. Taken together, TLR activation could be the molecular basis in the development of MetS-induced inflammation.
Collapse
Affiliation(s)
- Sok Kuan Wong
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, 56000 Cheras, Kuala Lumpur, Malaysia
| | - Kok-Yong Chin
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, 56000 Cheras, Kuala Lumpur, Malaysia
| | - Soelaiman Ima-Nirwana
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, 56000 Cheras, Kuala Lumpur, Malaysia
| |
Collapse
|
9
|
Xiao G, Zhuang W, Wang T, Lian G, Luo L, Ye C, Wang H, Xie L. Transcriptomic analysis identifies Toll-like and Nod-like pathways and necroptosis in pulmonary arterial hypertension. J Cell Mol Med 2020; 24:11409-11421. [PMID: 32860486 PMCID: PMC7576255 DOI: 10.1111/jcmm.15745] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 07/16/2020] [Accepted: 07/30/2020] [Indexed: 12/11/2022] Open
Abstract
Inflammation and immunity play a causal role in the pathogenesis of pulmonary vascular remodelling and pulmonary arterial hypertension (PAH). However, the pathways and mechanisms by which inflammation and immunity contribute to pulmonary vascular remodelling remain unknown. RNA sequencing was used to analyse the transcriptome in control and rats injected with monocrotaline (MCT) for various weeks. Using the transcriptional profiling of MCT‐induced PAH coupled with bioinformatics analysis, we clustered the differentially expressed genes (DEGs) and chose the increased expression patterns associated with inflammatory and immune response. We found the enrichment of Toll‐like receptor (TLR) and Nod‐like receptor (NLR) pathways and identified NF‐κB‐mediated inflammatory and immune profiling in MCT‐induced PAH. Pathway‐based data integration and visualization showed the dysregulated TLR and NLR pathways, including increased expression of TLR2 and NLRP3, and their downstream molecules. Further analysis revealed that the activation of TLR and NLR pathways was associated with up‐regulation of damage‐associated molecular patterns (DAMPs) and RIPK3‐mediated necroptosis was involved in the generation of DAMPs in MCT‐induced PAH. Collectively, we identify RIPK3‐mediated necroptosis and its triggered TLR and NLR pathways in the progression of pulmonary vascular remodelling, thus providing novel insights into the mechanisms underlying inflammation and immunity in the pathogenesis of PAH.
Collapse
Affiliation(s)
- Genfa Xiao
- Department of Geriatrics, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, People's Republic of China.,Department of General Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, People's Republic of China.,Fujian Hypertension Research Institute, The First Affiliated Hospital of Fujian Medical University, People's Republic of China
| | - Wei Zhuang
- Fujian Hypertension Research Institute, The First Affiliated Hospital of Fujian Medical University, People's Republic of China
| | - Tingjun Wang
- Department of Geriatrics, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, People's Republic of China.,Department of General Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, People's Republic of China.,Fujian Hypertension Research Institute, The First Affiliated Hospital of Fujian Medical University, People's Republic of China
| | - Guili Lian
- Fujian Hypertension Research Institute, The First Affiliated Hospital of Fujian Medical University, People's Republic of China
| | - Li Luo
- Department of Geriatrics, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, People's Republic of China.,Department of General Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, People's Republic of China.,Fujian Hypertension Research Institute, The First Affiliated Hospital of Fujian Medical University, People's Republic of China
| | - Chaoyi Ye
- Fujian Hypertension Research Institute, The First Affiliated Hospital of Fujian Medical University, People's Republic of China
| | - Huajun Wang
- Fujian Hypertension Research Institute, The First Affiliated Hospital of Fujian Medical University, People's Republic of China
| | - Liangdi Xie
- Department of Geriatrics, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, People's Republic of China.,Department of General Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, People's Republic of China.,Fujian Hypertension Research Institute, The First Affiliated Hospital of Fujian Medical University, People's Republic of China
| |
Collapse
|
10
|
Bhagwani A, Thompson AAR, Farkas L. When Innate Immunity Meets Angiogenesis-The Role of Toll-Like Receptors in Endothelial Cells and Pulmonary Hypertension. Front Med (Lausanne) 2020; 7:352. [PMID: 32850883 PMCID: PMC7410919 DOI: 10.3389/fmed.2020.00352] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 06/12/2020] [Indexed: 01/16/2023] Open
Abstract
Toll-like receptors serve a central role in innate immunity, but they can also modulate cell function in various non-immune cell types including endothelial cells. Endothelial cells are necessary for the organized function of the vascular system, and part of their fundamental role is also the regulation of immune function and inflammation. In this review, we summarize the current knowledge of how Toll-like receptors contribute to the immune and non-immune functions of the endothelial cells.
Collapse
Affiliation(s)
- Aneel Bhagwani
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Davis Heart & Lung Research Institute, The Ohio State University, Columbus, OH, United States
- Department of Physiology and Biophysics, Virginia Commonwealth University, Richmond, VA, United States
| | - A. A. Roger Thompson
- Department of Infection, Immunity & Cardiovascular Disease, Faculty of Medicine, Dentistry & Health, University of Sheffield, Sheffield, United Kingdom
| | - Laszlo Farkas
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Davis Heart & Lung Research Institute, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
11
|
Sun X, Sun BL, Babicheva A, Vanderpool R, Oita RC, Casanova N, Tang H, Gupta A, Lynn H, Gupta G, Rischard F, Sammani S, Kempf CL, Moreno-Vinasco L, Ahmed M, Camp SM, Wang J, Desai AA, Yuan JXJ, Garcia JGN. Direct Extracellular NAMPT Involvement in Pulmonary Hypertension and Vascular Remodeling. Transcriptional Regulation by SOX and HIF-2α. Am J Respir Cell Mol Biol 2020; 63:92-103. [PMID: 32142369 PMCID: PMC7328254 DOI: 10.1165/rcmb.2019-0164oc] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 03/05/2020] [Indexed: 12/21/2022] Open
Abstract
We previously demonstrated involvement of NAMPT (nicotinamide phosphoribosyltransferase) in pulmonary arterial hypertension (PAH) and now examine NAMPT regulation and extracellular NAMPT's (eNAMPT's) role in PAH vascular remodeling. NAMPT transcription and protein expression in human lung endothelial cells were assessed in response to PAH-relevant stimuli (PDGF [platelet-derived growth factor], VEGF [vascular endothelial growth factor], TGF-β1 [transforming growth factor-β1], and hypoxia). Endothelial-to-mesenchymal transition was detected by SNAI1 (snail family transcriptional repressor 1) and PECAM1 (platelet endothelial cell adhesion molecule 1) immunofluorescence. An eNAMPT-neutralizing polyclonal antibody was tested in a PAH model of monocrotaline challenge in rats. Plasma eNAMPT concentrations, significantly increased in patients with idiopathic pulmonary arterial hypertension, were highly correlated with indices of PAH severity. eNAMPT increased endothelial-to-mesenchymal transition, and each PAH stimulus significantly increased endothelial cell NAMPT promoter activity involving transcription factors STAT5 (signal transducer and activator of transcription 5), SOX18 (SRY-box transcription factor 18), and SOX17 (SRY-box transcription factor 17), a PAH candidate gene newly defined by genome-wide association study. The hypoxia-induced transcription factor HIF-2α (hypoxia-inducible factor-2α) also potently regulated NAMPT promoter activity, and HIF-2α binding sites were identified between -628 bp and -328 bp. The PHD2 (prolyl hydroxylase domain-containing protein 2) inhibitor FG-4592 significantly increased NAMPT promoter activity and protein expression in an HIF-2α-dependent manner. Finally, the eNAMPT-neutralizing polyclonal antibody significantly reduced monocrotaline-induced vascular remodeling, PAH hemodynamic alterations, and NF-κB activation. eNAMPT is a novel and attractive therapeutic target essential to PAH vascular remodeling.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Mohamed Ahmed
- Department of Pediatrics, University of Arizona Health Sciences, Tucson, Arizona
| | | | | | | | | | | |
Collapse
|
12
|
RNA Signaling in Pulmonary Arterial Hypertension-A Double-Stranded Sword. Int J Mol Sci 2020; 21:ijms21093124. [PMID: 32354189 PMCID: PMC7247700 DOI: 10.3390/ijms21093124] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 04/26/2020] [Accepted: 04/27/2020] [Indexed: 12/17/2022] Open
Abstract
Recognition of and response to pathogens and tissue injury is driven by the innate immune system via activation of pattern recognition receptors. One of the many patterns recognized is RNA and, while several receptors bind RNA, Toll-like receptor 3 (TLR3) is well placed for initial recognition of RNA molecules due to its localization within the endosome. There is a growing body of work describing a role for TLR3 in maintenance of vascular homeostasis. For example, TLR3 deficiency has been shown to play repair and remodeling roles in the systemic vasculature and in lung parenchyma. A hallmark of pulmonary arterial hypertension (PAH) is pulmonary vascular remodeling, yet drivers and triggers of this remodeling remain incompletely understood. Based on its role in the systemic vasculature, our group discovered reduced endothelial TLR3 expression in PAH and revealed a protective role for a TLR3 agonist in rodent models of pulmonary hypertension. This review will provide an overview of RNA signaling in the vasculature and how it relates to PAH pathobiology, including whether targeting double-stranded RNA signaling is a potential treatment option for PAH.
Collapse
|
13
|
Wedgwood S, Gerard K, Halloran K, Hanhauser A, Monacelli S, Warford C, Thai PN, Chiamvimonvat N, Lakshminrusimha S, Steinhorn RH, Underwood MA. Intestinal Dysbiosis and the Developing Lung: The Role of Toll-Like Receptor 4 in the Gut-Lung Axis. Front Immunol 2020; 11:357. [PMID: 32194566 PMCID: PMC7066082 DOI: 10.3389/fimmu.2020.00357] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Accepted: 02/14/2020] [Indexed: 01/19/2023] Open
Abstract
Background In extremely premature infants, postnatal growth restriction (PNGR) is common and increases the risk of developing bronchopulmonary dysplasia (BPD) and pulmonary hypertension (PH). Mechanisms by which poor nutrition impacts lung development are unknown, but alterations in the gut microbiota appear to play a role. In a rodent model, PNGR plus hyperoxia causes BPD and PH and increases intestinal Enterobacteriaceae, Gram-negative organisms that stimulate Toll-like receptor 4 (TLR4). We hypothesized that intestinal dysbiosis activates intestinal TLR4 triggering systemic inflammation which impacts lung development. Methods Rat pups were assigned to litters of 17 (PNGR) or 10 (normal growth) at birth and exposed to room air or 75% oxygen for 14 days. Half of the pups were treated with the TLR4 inhibitor TAK-242 from birth or beginning at day 3. After 14 days, pulmonary arterial pressure was evaluated by echocardiography and hearts were examined for right ventricular hypertrophy (RVH). Lungs and serum samples were analyzed by western blotting and immunohistochemistry. Results Postnatal growth restriction + hyperoxia increased pulmonary arterial pressure and RVH with trends toward increased plasma IL1β and decreased IκBα, the inhibitor of NFκB, in lung tissue. Treatment with the TLR4 inhibitor attenuated PH and inflammation. Conclusion Postnatal growth restriction induces an increase in intestinal Enterobacteriaceae leading to PH. Activation of the TLR4 pathway is a promising mechanism by which intestinal dysbiosis impacts the developing lung.
Collapse
Affiliation(s)
- Stephen Wedgwood
- Department of Pediatrics, UC Davis School of Medicine, Sacramento, CA, United States
| | - Kimberly Gerard
- Department of Pediatrics, UC Davis School of Medicine, Sacramento, CA, United States
| | - Katrina Halloran
- Department of Pediatrics, UC Davis School of Medicine, Sacramento, CA, United States
| | - Ashley Hanhauser
- Department of Pediatrics, UC Davis School of Medicine, Sacramento, CA, United States
| | - Sveva Monacelli
- Department of Pediatrics, UC Davis School of Medicine, Sacramento, CA, United States
| | - Cris Warford
- Department of Pediatrics, UC Davis School of Medicine, Sacramento, CA, United States
| | - Phung N Thai
- Division of Cardiovascular Medicine, Department of Internal Medicine, UC Davis Health System, Sacramento, CA, United States
| | - Nipavan Chiamvimonvat
- Division of Cardiovascular Medicine, Department of Internal Medicine, UC Davis Health System, Sacramento, CA, United States.,Department of Veterans Affairs, Northern California Health Care System, Mather, CA, United States
| | | | - Robin H Steinhorn
- Department of Hospital Medicine, Children's National Health System, Washington, DC, United States
| | - Mark A Underwood
- Department of Pediatrics, UC Davis School of Medicine, Sacramento, CA, United States
| |
Collapse
|
14
|
Wedgwood S, Warford C, Agvatisiri SR, Thai PN, Chiamvimonvat N, Kalanetra KM, Lakshminrusimha S, Steinhorn RH, Mills DA, Underwood MA. The developing gut-lung axis: postnatal growth restriction, intestinal dysbiosis, and pulmonary hypertension in a rodent model. Pediatr Res 2020; 87:472-479. [PMID: 31537010 PMCID: PMC7035999 DOI: 10.1038/s41390-019-0578-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 07/13/2019] [Accepted: 08/29/2019] [Indexed: 01/08/2023]
Abstract
BACKGROUND Postnatal growth restriction (PNGR) in premature infants increases risk of pulmonary hypertension (PH). In a rodent model, PNGR causes PH, while combining PNGR and hyperoxia increases PH severity. We hypothesized that PNGR causes intestinal dysbiosis and that treatment with a probiotic attenuates PNGR-associated PH. METHOD Pups were randomized at birth to room air or 75% oxygen (hyperoxia), to normal milk intake (10 pups/dam) or PNGR (17 pups/dam), and to probiotic Lactobacillus reuteri DSM 17938 or phosphate-buffered saline. After 14 days, PH was assessed by echocardiography and right ventricular hypertrophy (RVH) was assessed by Fulton's index (right ventricular weight/left ventricle + septal weight). The small bowel and cecum were analyzed by high-throughput 16S ribosomal RNA gene sequencing. RESULTS PNGR with or without hyperoxia (but not hyperoxia alone) altered the microbiota of the distal small bowel and cecum. Treatment with DSM 17938 attenuated PH and RVH in pups with PNGR, but not hyperoxia alone. DSM 17938 treatment decreased α-diversity. The intestinal microbiota differed based on oxygen exposure, litter size, and probiotic treatment. CONCLUSION PNGR causes intestinal dysbiosis and PH. Treatment with DSM 17938 prevents PNGR-associated RVH and PH. Changes in the developing intestine and intestinal microbiota impact the developing lung vasculature and RV.
Collapse
MESH Headings
- Animal Nutritional Physiological Phenomena
- Animals
- Animals, Newborn
- Caloric Restriction/adverse effects
- Cecum/microbiology
- Disease Models, Animal
- Dysbiosis
- Female
- Gastrointestinal Microbiome
- Hyperoxia/complications
- Hypertension, Pulmonary/etiology
- Hypertension, Pulmonary/microbiology
- Hypertension, Pulmonary/physiopathology
- Hypertension, Pulmonary/prevention & control
- Hypertrophy, Right Ventricular/etiology
- Hypertrophy, Right Ventricular/microbiology
- Hypertrophy, Right Ventricular/physiopathology
- Hypertrophy, Right Ventricular/prevention & control
- Intestine, Small/microbiology
- Limosilactobacillus reuteri/physiology
- Litter Size
- Lung/blood supply
- Nutritional Status
- Pregnancy
- Probiotics/administration & dosage
- Rats, Sprague-Dawley
Collapse
Affiliation(s)
- Stephen Wedgwood
- Department of Pediatrics, UC Davis School of Medicine, Sacramento, CA, USA
| | - Cris Warford
- Department of Pediatrics, UC Davis School of Medicine, Sacramento, CA, USA
| | | | - Phung N Thai
- Department of Internal Medicine, Division of Cardiovascular Medicine, UC Davis Health System, Sacramento, CA, USA
| | - Nipavan Chiamvimonvat
- Department of Internal Medicine, Division of Cardiovascular Medicine, UC Davis Health System, Sacramento, CA, USA
- Department of Veterans Affairs, Northern California Health Care System, Mather, CA, USA
| | | | | | - Robin H Steinhorn
- Department of Hospitalist Medicine, Children's National Health System, Washington, DC, USA
| | - David A Mills
- Department of Food Science and Technology, UC Davis, Davis, CA, USA
| | - Mark A Underwood
- Department of Pediatrics, UC Davis School of Medicine, Sacramento, CA, USA.
| |
Collapse
|
15
|
Goldenberg NM, Hu Y, Hu X, Volchuk A, Zhao YD, Kucherenko MM, Knosalla C, de Perrot M, Tracey KJ, Al-Abed Y, Steinberg BE, Kuebler WM. Therapeutic Targeting of High-Mobility Group Box-1 in Pulmonary Arterial Hypertension. Am J Respir Crit Care Med 2019; 199:1566-1569. [PMID: 30939030 PMCID: PMC7125426 DOI: 10.1164/rccm.201808-1597le] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Affiliation(s)
- Neil M. Goldenberg
- Hospital for Sick ChildrenToronto, Ontario, Canada
- University of TorontoToronto, Ontario, Canada
| | - Yijie Hu
- St. Michael’s HospitalToronto, Ontario, Canada
- Third Military Medical UniversityChongqing, China
| | - Xudong Hu
- St. Michael’s HospitalToronto, Ontario, Canada
| | | | | | - Mariya M. Kucherenko
- Charité Universitätsmedizin BerlinBerlin, Germany
- German Heart InstituteBerlin, Germany
- DZHK (German Center for Cardiovascular Research)Berlin, Germany
| | - Christoph Knosalla
- German Heart InstituteBerlin, Germany
- DZHK (German Center for Cardiovascular Research)Berlin, Germany
| | | | - Kevin J. Tracey
- Feinstein Institute for Medical ResearchManhasset, New Yorkand
| | - Yousef Al-Abed
- Feinstein Institute for Medical ResearchManhasset, New Yorkand
| | - Benjamin E. Steinberg
- Hospital for Sick ChildrenToronto, Ontario, Canada
- University of TorontoToronto, Ontario, Canada
| | - Wolfgang M. Kuebler
- University of TorontoToronto, Ontario, Canada
- Charité Universitätsmedizin BerlinBerlin, Germany
- DZHK (German Center for Cardiovascular Research)Berlin, Germany
- St. Michael’s HospitalToronto, Canada
| |
Collapse
|
16
|
Wang J, Tian XT, Peng Z, Li WQ, Cao YY, Li Y, Li XH. HMGB1/TLR4 promotes hypoxic pulmonary hypertension via suppressing BMPR2 signaling. Vascul Pharmacol 2019; 117:35-44. [DOI: 10.1016/j.vph.2018.12.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 12/17/2018] [Accepted: 12/28/2018] [Indexed: 11/25/2022]
|
17
|
Nunes KP, de Oliveira AA, Mowry FE, Biancardi VC. Targeting toll-like receptor 4 signalling pathways: can therapeutics pay the toll for hypertension? Br J Pharmacol 2018; 176:1864-1879. [PMID: 29981161 DOI: 10.1111/bph.14438] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 06/09/2018] [Accepted: 06/24/2018] [Indexed: 02/06/2023] Open
Abstract
The immune system plays a prominent role in the initiation and maintenance of hypertension. The innate immune system, via toll-like receptors (TLRs), identifies distinct signatures of invading microbes and damage-associated molecular patterns and triggers a chain of downstream signalling cascades, leading to secretion of pro-inflammatory cytokines and shaping the adaptive immune response. Over the past decade, a dysfunctional TLR-mediated response, particularly via TLR4, has been suggested to support a chronic inflammatory state in hypertension, inducing deleterious local and systemic effects in host cells and tissues and contributing to disease progression. While the underlying mechanisms triggering TLR4 need further research, evidence suggests that sustained elevations in BP disrupt homeostasis, releasing endogenous TLR4 ligands in hypertension. In this review, we discuss the emerging role of TLR4 in the pathogenesis of hypertension and whether targeting this receptor and its signalling pathways could offer a therapeutic strategy for management of this multifaceted disease. LINKED ARTICLES: This article is part of a themed section on Immune Targets in Hypertension. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.12/issuetoc.
Collapse
Affiliation(s)
- Kenia Pedrosa Nunes
- Department of Biological Sciences, Florida Institute of Technology, Melbourne, FL, USA
| | | | | | | |
Collapse
|
18
|
Ranchoux B, Bigorgne A, Hautefort A, Girerd B, Sitbon O, Montani D, Humbert M, Tcherakian C, Perros F. Gut-Lung Connection in Pulmonary Arterial Hypertension. Am J Respir Cell Mol Biol 2018; 56:402-405. [PMID: 28248132 DOI: 10.1165/rcmb.2015-0404le] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Benoît Ranchoux
- 1 University Paris-Sud Le Kremlin-Bicêtre, France.,2 Hôpital Bicêtre Le Kremlin-Bicêtre, France.,3 Inserm U999 Le Plessis-Robinson, France
| | - Amélie Bigorgne
- 6 Inserm U1163 - Imagine Institute Paris, France and.,7 University Paris Descartes Paris, France
| | - Aurélie Hautefort
- 1 University Paris-Sud Le Kremlin-Bicêtre, France.,2 Hôpital Bicêtre Le Kremlin-Bicêtre, France.,3 Inserm U999 Le Plessis-Robinson, France
| | - Barbara Girerd
- 1 University Paris-Sud Le Kremlin-Bicêtre, France.,2 Hôpital Bicêtre Le Kremlin-Bicêtre, France.,3 Inserm U999 Le Plessis-Robinson, France
| | - Olivier Sitbon
- 1 University Paris-Sud Le Kremlin-Bicêtre, France.,2 Hôpital Bicêtre Le Kremlin-Bicêtre, France.,3 Inserm U999 Le Plessis-Robinson, France
| | - David Montani
- 1 University Paris-Sud Le Kremlin-Bicêtre, France.,2 Hôpital Bicêtre Le Kremlin-Bicêtre, France.,3 Inserm U999 Le Plessis-Robinson, France
| | - Marc Humbert
- 1 University Paris-Sud Le Kremlin-Bicêtre, France.,2 Hôpital Bicêtre Le Kremlin-Bicêtre, France.,3 Inserm U999 Le Plessis-Robinson, France
| | - Colas Tcherakian
- 5 Hôpital Foch Suresnes, France and.,8 Université de Versailles-Saint-Quentin-en-Yvelines Versailles, France
| | - Frédéric Perros
- 1 University Paris-Sud Le Kremlin-Bicêtre, France.,2 Hôpital Bicêtre Le Kremlin-Bicêtre, France.,3 Inserm U999 Le Plessis-Robinson, France
| |
Collapse
|
19
|
Chou HC, Lin W, Chen CM. Human mesenchymal stem cells attenuate pulmonary hypertension induced by prenatal lipopolysaccharide treatment in rats. Clin Exp Pharmacol Physiol 2017; 43:906-14. [PMID: 27273502 DOI: 10.1111/1440-1681.12604] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2016] [Revised: 05/17/2016] [Accepted: 06/04/2016] [Indexed: 11/26/2022]
Abstract
Intra-amniotic injection of lipopolysaccharide (LPS) induces pulmonary hypertension in newborn rats. This study was designed to test whether human mesenchymal stem cells (MSCs) reduce pulmonary hypertension and alleviate cardiac hypertrophy in prenatal LPS-treated rats. Pregnant Sprague-Dawley rats were injected intraperitoneally with LPS (0.5 mg/kg per day) or untreated on gestational days 20 and 21. Human MSCs (3×10(5) cells and 1×10(6) cells) in 0.03 mL of normal saline (NS) were transplanted intratracheally on postnatal day 5. Four study groups were considered: normal, LPS+NS, LPS+MSCs (3×10(5) cells), and LPS+MSCs (1×10(6) cells). On postnatal day 14, lung and heart tissues were collected for measuring the arterial medial wall thickness (MWT) and β-myosin heavy chain (β-MHC) level as markers of pulmonary hypertension and cardiac hypertrophy, respectively. The LPS+NS group exhibited a significantly higher right ventricle (RV)/[left ventricle (LV)+ interventricular septum (IVS)] thickness ratio and MWT, a greater cardiomyocyte width, a greater number of cardiomyocyte nuclei per squared millimeter, and higher β-MHC expression than those observed in the normal group. Human MSC transplantation (3×10(5) cells and 1×10(6) cells) in LPS-treated rats reduced MWT and the RV/(LV+IVS) thickness ratio to normal levels. This improvement in right ventricular hypertrophy was accompanied by a decrease in toll-like receptor 4 (TLR4), nuclear factor-κB, and tumor necrosis factor-α expression in the heart. Intratracheal human MSCs transplantation can attenuate pulmonary hypertension and right ventricular hypertrophy in prenatal LPS-treated rats; this attenuation may be associated with suppression of TLR4 expression via paracrine pathways.
Collapse
Affiliation(s)
- Hsiu-Chu Chou
- Department of Anatomy and Cellular Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Willie Lin
- Meridigen Biotech Co., Ltd., Taipei, Taiwan
| | - Chung-Ming Chen
- Department of Pediatrics, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Pediatrics, Taipei Medical University Hospital, Taipei, Taiwan
| |
Collapse
|
20
|
Zhang Y, Huang Z, Li H. Insights into innate immune signalling in controlling cardiac remodelling. Cardiovasc Res 2017; 113:1538-1550. [DOI: 10.1093/cvr/cvx130] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 06/29/2017] [Indexed: 01/22/2023] Open
Affiliation(s)
- Yaxing Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuchang District, Wuhan 430060, People’s Republic of China
- Institute of Model Animal of Wuhan University, Donghu Road 115, Wuchang District, Wuhan 430071, People’s Republic of China
- Medical Research Institute, School of Medicine, Wuhan University, Donghu Road 115, Wuchang District, Wuhan 430071, People’s Republic of China
| | - Zan Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuchang District, Wuhan 430060, People’s Republic of China
- Institute of Model Animal of Wuhan University, Donghu Road 115, Wuchang District, Wuhan 430071, People’s Republic of China
- Medical Research Institute, School of Medicine, Wuhan University, Donghu Road 115, Wuchang District, Wuhan 430071, People’s Republic of China
- College of Life Sciences, Wuhan University, Wuhan 430072, People’s Republic of China
| | - Hongliang Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuchang District, Wuhan 430060, People’s Republic of China
- Institute of Model Animal of Wuhan University, Donghu Road 115, Wuchang District, Wuhan 430071, People’s Republic of China
- Medical Research Institute, School of Medicine, Wuhan University, Donghu Road 115, Wuchang District, Wuhan 430071, People’s Republic of China
| |
Collapse
|
21
|
Rodrigues-Diez R, González-Guerrero C, Ocaña-Salceda C, Rodrigues-Diez RR, Egido J, Ortiz A, Ruiz-Ortega M, Ramos AM. Calcineurin inhibitors cyclosporine A and tacrolimus induce vascular inflammation and endothelial activation through TLR4 signaling. Sci Rep 2016; 6:27915. [PMID: 27295076 PMCID: PMC4904742 DOI: 10.1038/srep27915] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 05/26/2016] [Indexed: 12/20/2022] Open
Abstract
The introduction of the calcineurin inhibitors (CNIs) cyclosporine and tacrolimus greatly reduced the rate of allograft rejection, although their chronic use is marred by a range of side effects, among them vascular toxicity. In transplant patients, it is proved that innate immunity promotes vascular injury triggered by ischemia-reperfusion damage, atherosclerosis and hypertension. We hypothesized that activation of the innate immunity and inflammation may contribute to CNI toxicity, therefore we investigated whether TLR4 mediates toxic responses of CNIs in the vasculature. Cyclosporine and tacrolimus increased the production of proinflammatory cytokines and endothelial activation markers in cultured murine endothelial and vascular smooth muscle cells as well as in ex vivo cultures of murine aortas. CNI-induced proinflammatory events were prevented by pharmacological inhibition of TLR4. Moreover, CNIs were unable to induce inflammation and endothelial activation in aortas from TLR4−/− mice. CNI-induced cytokine and adhesion molecules synthesis in endothelial cells occurred even in the absence of calcineurin, although its expression was required for maximal effect through upregulation of TLR4 signaling. CNI-induced TLR4 activity increased O2−/ROS production and NF-κB-regulated synthesis of proinflammatory factors in cultured as well as aortic endothelial and VSMCs. These data provide new insight into the mechanisms associated with CNI vascular inflammation.
Collapse
Affiliation(s)
- Raquel Rodrigues-Diez
- Laboratory of Nephrology and Vascular Pathology, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz (IIS-FJD), Madrid, Spain
| | - Cristian González-Guerrero
- Laboratory of Nephrology and Vascular Pathology, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz (IIS-FJD), Madrid, Spain
| | - Carlos Ocaña-Salceda
- Laboratory of Nephrology and Vascular Pathology, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz (IIS-FJD), Madrid, Spain
| | - Raúl R Rodrigues-Diez
- Laboratory of Nephrology and Vascular Pathology, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz (IIS-FJD), Madrid, Spain
| | - Jesús Egido
- Laboratory of Nephrology and Vascular Pathology, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz (IIS-FJD), Madrid, Spain.,Fundación Renal Íñigo Álvarez de Toledo (FRIAT), Madrid, Spain
| | - Alberto Ortiz
- Laboratory of Nephrology and Vascular Pathology, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz (IIS-FJD), Madrid, Spain.,Fundación Renal Íñigo Álvarez de Toledo (FRIAT), Madrid, Spain
| | - Marta Ruiz-Ortega
- Cellular Biology in Renal Diseases Laboratory, Universidad Autónoma de Madrid, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz (IIS-FJD), Madrid, Spain
| | - Adrián M Ramos
- Laboratory of Nephrology and Vascular Pathology, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz (IIS-FJD), Madrid, Spain
| |
Collapse
|
22
|
Voelkel NF, Tamosiuniene R, Nicolls MR. Challenges and opportunities in treating inflammation associated with pulmonary hypertension. Expert Rev Cardiovasc Ther 2016; 14:939-51. [PMID: 27096622 DOI: 10.1080/14779072.2016.1180976] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Inflammatory cells are present in the lungs from patients with many, if not all, forms of severe pulmonary hypertension. AREAS COVERED Historically the first inflammatory cell identified in the pulmonary vascular lesions was the mast cell. T and B lymphocytes, as well as macrophages, are present in and around the pulmonary arterioles and many patients have elevated blood levels of interleukin 1 and 6; some patients show elevated levels of leukotriene B4. An overlap between collagen-vascular disease-associated pulmonary arterial hypertension (PAH) and idiopathic PAH exists, yet only a few studies have been designed that evaluate the effect of anti-inflammatory treatments. Here we review the pertinent data that connect PAH and inflammation/autoimmune dysregulation and evaluate experimental models of severe PAH with an emphasis on the Sugen/athymic rat model of severe PAH. Expert commentary: We postulate that there are several inflammatory phenotypes and predict that there will be several anti-inflammatory treatment strategies for severe PAH.
Collapse
Affiliation(s)
- Norbert F Voelkel
- a School of Pharmacy , Virginia Commonwealth University , Richmond , VA , USA
| | - Rasa Tamosiuniene
- b Pulmonary and Critical Care Medicine Division , Stanford University , Palo Alto , CA , USA
| | - Mark R Nicolls
- b Pulmonary and Critical Care Medicine Division , Stanford University , Palo Alto , CA , USA
| |
Collapse
|
23
|
Rafikova O, Meadows ML, Kinchen JM, Mohney RP, Maltepe E, Desai AA, Yuan JXJ, Garcia JGN, Fineman JR, Rafikov R, Black SM. Metabolic Changes Precede the Development of Pulmonary Hypertension in the Monocrotaline Exposed Rat Lung. PLoS One 2016; 11:e0150480. [PMID: 26937637 PMCID: PMC4777490 DOI: 10.1371/journal.pone.0150480] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 02/14/2016] [Indexed: 12/13/2022] Open
Abstract
There is increasing interest in the potential for metabolic profiling to evaluate the progression of pulmonary hypertension (PH). However, a detailed analysis of the metabolic changes in lungs at the early stage of PH, characterized by increased pulmonary artery pressure but prior to the development of right ventricle hypertrophy and failure, is lacking in a preclinical animal model of PH. Thus, we undertook a study using rats 14 days after exposure to monocrotaline (MCT), to determine whether we could identify early stage metabolic changes prior to the manifestation of developed PH. We observed changes in multiple pathways associated with the development of PH, including activated glycolysis, increased markers of proliferation, disruptions in carnitine homeostasis, increased inflammatory and fibrosis biomarkers, and a reduction in glutathione biosynthesis. Further, our global metabolic profile data compare favorably with prior work carried out in humans with PH. We conclude that despite the MCT-model not recapitulating all the structural changes associated with humans with advanced PH, including endothelial cell proliferation and the formation of plexiform lesions, it is very similar at a metabolic level. Thus, we suggest that despite its limitations it can still serve as a useful preclinical model for the study of PH.
Collapse
Affiliation(s)
- Olga Rafikova
- Division of Translational and Regenerative Medicine, The University of Arizona, Tucson, Arizona, United States of America
- Department of Medicine, The University of Arizona, Tucson, Arizona, United States of America
| | - Mary L. Meadows
- Vascular Biology Center, Georgia Regents University, Augusta, Georgia, United States of America
| | | | | | - Emin Maltepe
- Division of Neonatology, University of California San Francisco, San Francisco, California, United States of America
| | - Ankit A. Desai
- Department of Medicine, The University of Arizona, Tucson, Arizona, United States of America
| | - Jason X.-J. Yuan
- Division of Translational and Regenerative Medicine, The University of Arizona, Tucson, Arizona, United States of America
- Department of Medicine, The University of Arizona, Tucson, Arizona, United States of America
| | - Joe G. N. Garcia
- Department of Medicine, The University of Arizona, Tucson, Arizona, United States of America
| | - Jeffrey R. Fineman
- Department of Pediatrics, University of California San Francisco, San Francisco, California, United States of America
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, California, United States of America
| | - Ruslan Rafikov
- Division of Translational and Regenerative Medicine, The University of Arizona, Tucson, Arizona, United States of America
- Department of Medicine, The University of Arizona, Tucson, Arizona, United States of America
- * E-mail:
| | - Stephen M. Black
- Division of Translational and Regenerative Medicine, The University of Arizona, Tucson, Arizona, United States of America
- Department of Medicine, The University of Arizona, Tucson, Arizona, United States of America
| |
Collapse
|
24
|
Sasaki H, Hirai K, Martins CM, Furusho H, Battaglino R, Hashimoto K. Interrelationship Between Periapical Lesion and Systemic Metabolic Disorders. Curr Pharm Des 2016; 22:2204-15. [PMID: 26881444 PMCID: PMC4856634 DOI: 10.2174/1381612822666160216145107] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 02/15/2016] [Indexed: 12/25/2022]
Abstract
BACKGROUND Periapical periodontitis, also known as periapical lesion, is a common dental disease, along with periodontitis (gum disease). Periapical periodontitis is a chronic inflammatory disease, caused by endodontic infection, and its development is regulated by the host immune/inflammatory response. Metabolic disorders, which are largely dependent on life style such as eating habits, have been interpreted as a "metabolically-triggered" low-grade systemic inflammation and may interact with periapical periodontitis by triggering immune modulation. The host immune system is therefore considered the common fundamental mechanism of both disease conditions. METHOD We have reviewed >200 articles to discuss the interrelationship between periapical lesions and metabolic disorders including type 2 diabetes mellitus, hypertension, and non-alcoholic fatty liver diseases (NAFLD), and their common pathological background in immunology/osteoimmunology and cytokine biology. RESULTS An elevated inflammatory state caused by metabolic disorders can impact the clinical outcome of periapical lesions and interfere with wound healing after endodontic treatment. Although additional well-designed clinical studies are needed, periapical lesions appear to affect insulin sensitivity and exacerbate non-alcoholic steatohepatitis. CONCLUSION Immune regulatory cytokines produced by various cell types, including immune cells and adipose tissue, play an important role in this interrelationship.
Collapse
Affiliation(s)
- Hajime Sasaki
- Department of Immunology & Infectious Diseases, The Forsyth Institute, 245 First Street, Cambridge, MA 02494, U.S.A.
| | | | | | | | | | | |
Collapse
|
25
|
Ma L, Ambalavanan N, Liu H, Sun Y, Jhala N, Bradley WE, Dell'Italia LJ, Michalek S, Wu H, Steele C, Benza RL, Chen Y. TLR4 regulates pulmonary vascular homeostasis and remodeling via redox signaling. Front Biosci (Landmark Ed) 2016; 21:397-409. [PMID: 26709781 DOI: 10.2741/4396] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Pulmonary arterial hypertension (PAH) contributes to morbidity and mortality of patients with lung and heart diseases. We demonstrated that hypoxia induced PAH and increased pulmonary arterial wall thickness in wild-type mice. Mice deficient in toll-like receptor 4 (TLR4-/-) spontaneously developed PAH, which was not further enhanced by hypoxia. Echocardiography determined right ventricular hypertrophy and decreased pulmonary arterial acceleration time were associated with the development of PAH in TLR4(-/-) mice. In pulmonary arterial smooth muscle cells (PASMC), hypoxia decreased TLR4 expression and induced reactive oxygen species (ROS) and Nox1/Nox4. Inhibition of NADPH oxidase decreased hypoxia-induced proliferation of wild-type PASMC. PASMC derived from TLR4(-/-) mice exhibited increased ROS and Nox4/Nox1 expression. Our studies demonstrate an important role of TLR4 in maintaining normal pulmonary vasculature and in hypoxia-induced PAH. Inhibition of TLR4, by genetic ablation or hypoxia, increases the expression of Nox1/Nox4 and induces PASMC proliferation and vascular remodeling. These results support a novel function of TLR4 in regulating the development of PAH and reveal a new regulatory axis contributing to TLR4 deficiency-induced vascular hypertrophy and remodeling.
Collapse
Affiliation(s)
- Liping Ma
- Department of Pathology, University of Alabama at Birmingham, Birmingham AL 35294, *current address: Sun Yat-Sen Memorial Hospital ,Sun Yat-Sen University, Guangzhou 510120, China
| | | | - Hui Liu
- Department of Medicine, University of Alabama at Birmingham, Birmingham AL 35294
| | - Yong Sun
- Department of Pathology, University of Alabama at Birmingham, Birmingham AL 35294
| | - Nirag Jhala
- Department of Pathology, University of Alabama at Birmingham, Birmingham AL 35294
| | - Wayne E Bradley
- Department of Medicine, University of Alabama at Birmingham, Birmingham AL 35294
| | - Louis J Dell'Italia
- Department of Medicine, University of Alabama at Birmingham, Birmingham AL 35294; VA Medical Center, Birmingham AL 35294
| | - Sue Michalek
- Department of Microbiology, University of Alabama at Birmingham, Birmingham AL 35294
| | - Hui Wu
- Department of Microbiology, University of Alabama at Birmingham, Birmingham AL 35294; Department of Pediatric Dentistry, University of Alabama at Birmingham, Birmingham AL 35294
| | - Chad Steele
- Department of Medicine, University of Alabama at Birmingham, Birmingham AL 35294
| | - Raymond L Benza
- Department of Medicine, University of Alabama at Birmingham, Birmingham AL 35294
| | - Yabing Chen
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294; VA Medical Center, Birmingham AL 35294,
| |
Collapse
|
26
|
Beneficial Effects of Qili Qiangxin Capsule on Lung Structural Remodeling in Ischemic Heart Failure via TGF-β1/Smad3 Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2015; 2015:298631. [PMID: 26604970 PMCID: PMC4641193 DOI: 10.1155/2015/298631] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Revised: 09/21/2015] [Accepted: 09/27/2015] [Indexed: 01/22/2023]
Abstract
Qili qiangxin (QL) capsule is a traditional Chinese medicine that is widely used for the treatment of patients with chronic heart failure (CHF) of all etiologies, although the exact mechanisms of action remain unclear. CHF leads to pulmonary vascular remodelling and thickening of the alveolar-capillary barrier that may be important mechanisms in the poor clinical outcome in patients with end-stage heart failure. We examined whether QL could improve lung injury in ischemic CHF by reducing lung remodeling. Rats with myocardial infarct received QL (1.0 g/kg/day) for 4 weeks. Echocardiographic and morphometric measurements were obtained followed by echocardiography, histological staining, and immunohistochemical analysis of lung sections. CHF caused significant lung structural remodeling evidenced by collagen deposition and thickening of the alveolar septa after myocardial infarct that were greatly improved by QL. Lung weight increased after infarct with no evidence of pulmonary edema and was normalized by QL. QL also reduced lung transforming growth factor-β1 (TGF-β1), p-Smad3, tumor necrosis factor-α (TNF-α), and Toll-like receptor-4 (TLR4) expression. Thus, QL reduces lung remodeling associated with CHF, mainly by suppressing the TGF-β1/Smad3 signaling pathway. The mechanism may also involve inhibition of TLR4 intracellular signaling.
Collapse
|
27
|
Bobek G, Surmon L, Mirabito KM, Makris A, Hennessy A. Placental Regulation of Inflammation and Hypoxia after TNF-α Infusion in Mice. Am J Reprod Immunol 2015. [PMID: 26220019 DOI: 10.1111/aji.12417] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
PROBLEM Increased levels of inflammatory cytokines are demonstrated in the serum of women with pre-eclampsia. TNF-α infusion in animal models induces proteinuric hypertension similar to human pre-eclampsia. The effect of TNF-α on regulation of the immune and hypoxic pathways in the developing placenta and their relationship with experimental pre-eclampsia remains unexamined. METHOD OF STUDY TNF-α was infused into pregnant mice, and the effects on maternal hypertension, proteinuria, circulating levels of sFlt-1 and corresponding placental changes in molecules responding to inflammation (TLR-3 and TLR-4) and hypoxia (HIF-1α) were examined. RESULTS TNF-α infusion resulted in maternal hypertension and proteinuria. Molecular changes in the placenta involved upregulation of TLR-3, TLR-4 and HIF-1α. Serum levels of sFlt-1 were high in pregnant animals, but not further upregulated by TNF-α infusion. CONCLUSION A role for maladaptive regulation of TLR and HIF-1α induced by an imbalance in inflammatory cytokines is implicated in the pathogenesis of pre-eclampsia.
Collapse
Affiliation(s)
- Gabriele Bobek
- School of Medicine, University of Western Sydney, Campbelltown, NSW, Australia
| | - Laura Surmon
- School of Medicine, University of Western Sydney, Campbelltown, NSW, Australia
| | | | - Angela Makris
- School of Medicine, University of Western Sydney, Campbelltown, NSW, Australia.,The Heart Research Institute, University of Sydney, Sydney, NSW, Australia.,Liverpool Hospital, Renal Unit Liverpool, Liverpool, NSW, Australia
| | - Annemarie Hennessy
- School of Medicine, University of Western Sydney, Campbelltown, NSW, Australia.,The Heart Research Institute, University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
28
|
Abstract
Pulmonary hypertension (PH) is a progressive lung disease characterized by elevated pressure in the lung vasculature, resulting in right-sided heart failure and premature death. The pathogenesis of PH is complex and multifactorial, involving a dysregulated autonomic nervous system and immune response. Inflammatory mechanisms have been linked to the development and progression of PH; however, these are usually restricted to systemic and/or local lung tissue. Inflammation within the CNS, often referred to as neuroinflammation involves activation of the microglia, the innate immune cells that are found specifically in the brain and spinal cord. Microglial activation results in the release of several cytokines and chemokines that trigger neuroinflammation, and has been implicated in the pathogenesis of several disease conditions such as Alzheimer's, Parkinson's, hypertension, atherosclerosis, and metabolic disorders. In this review, we introduce the concept of neuroinflammation in the context of PH, and discuss possible strategies that could be developed for PH therapy based on this concept.
Collapse
|
29
|
Irwin DC, Baek JH, Hassell K, Nuss R, Eigenberger P, Lisk C, Loomis Z, Maltzahn J, Stenmark KR, Nozik-Grayck E, Buehler PW. Hemoglobin-induced lung vascular oxidation, inflammation, and remodeling contribute to the progression of hypoxic pulmonary hypertension and is attenuated in rats with repeated-dose haptoglobin administration. Free Radic Biol Med 2015; 82:50-62. [PMID: 25656991 PMCID: PMC4387123 DOI: 10.1016/j.freeradbiomed.2015.01.012] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Revised: 12/11/2014] [Accepted: 01/20/2015] [Indexed: 12/28/2022]
Abstract
Haptoglobin (Hp) is an approved treatment in Japan for trauma, burns, and massive transfusion-related hemolysis. Additional case reports suggest uses in other acute hemolytic events that lead to acute kidney injury. However, Hp's protective effects on the pulmonary vasculature have not been evaluated within the context of mitigating the consequences of chronic hemoglobin (Hb) exposure in the progression of pulmonary hypertension (PH) secondary to hemolytic diseases. This study was performed to assess the utility of chronic Hp therapy in a preclinical model of Hb and hypoxia-mediated PH. Rats were simultaneously exposed to chronic Hb infusion (35 mg per day) and hypobaric hypoxia for 5 weeks in the presence or absence of Hp treatment (90 mg/kg twice a week). Hp inhibited the Hb plus hypoxia-mediated nonheme iron accumulation in lung and heart tissue, pulmonary vascular inflammation and resistance, and right-ventricular hypertrophy, which suggests a positive impact on impeding the progression of PH. In addition, Hp therapy was associated with a reduction in critical mediators of PH, including lung adventitial macrophage population and endothelial ICAM-1 expression. By preventing Hb-mediated pathology, Hp infusions: (1) demonstrate a critical role for Hb in vascular remodeling associated with hypoxia and (2) suggest a novel therapy for chronic hemolysis-associated PH.
Collapse
Affiliation(s)
- David C. Irwin
- Cardiovascular Pulmonary Research Group, Division of Cardiology, School of Medicine, University of Colorado Denver | Anschutz Medical Campus, Aurora, Colorado
- Corresponding Author: David C. Irwin, Ph.D., Assistant Professor, 12700 East 19th Avenue, Research Building 2, Room 8121, Aurora, CO 80045, Phone: 303 724-3684, Fax: 303 724-3693,
| | - Jin Hyen Baek
- Laboratory of Biochemistry and Vascular Biology, Office of Blood Research and Review, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Bethesda, Maryland
| | - Kathryn Hassell
- Colorado Sickle Cell Treatment and Research Center, University of Colorado Denver | Anschutz Medical Campus, Aurora, Colorado
| | - Rachelle Nuss
- Colorado Sickle Cell Treatment and Research Center, University of Colorado Denver | Anschutz Medical Campus, Aurora, Colorado
| | - Paul Eigenberger
- Cardiovascular Pulmonary Research Group, Division of Cardiology, School of Medicine, University of Colorado Denver | Anschutz Medical Campus, Aurora, Colorado
| | - Christina Lisk
- Cardiovascular Pulmonary Research Group, Division of Cardiology, School of Medicine, University of Colorado Denver | Anschutz Medical Campus, Aurora, Colorado
| | - Zoe Loomis
- Colorado Sickle Cell Treatment and Research Center, University of Colorado Denver | Anschutz Medical Campus, Aurora, Colorado
| | - Joanne Maltzahn
- Cardiovascular Pulmonary Research Group, Division of Cardiology, School of Medicine, University of Colorado Denver | Anschutz Medical Campus, Aurora, Colorado
| | - Kurt R Stenmark
- Cardiovascular Pulmonary Research Group, Pediatrics, School of Medicine, University of Colorado Denver | Anschutz Medical Campus, Aurora Colorado
| | - Eva Nozik-Grayck
- Cardiovascular Pulmonary Research Group, Pediatrics, School of Medicine, University of Colorado Denver | Anschutz Medical Campus, Aurora Colorado
| | - Paul W. Buehler
- Cardiovascular Pulmonary Research Group, Division of Cardiology, School of Medicine, University of Colorado Denver | Anschutz Medical Campus, Aurora, Colorado
- Laboratory of Biochemistry and Vascular Biology, Office of Blood Research and Review, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Bethesda, Maryland
| |
Collapse
|
30
|
Wang LM, Zhong NZ, Liu SJ, Zhu XY, Liu YJ. Hypoxia-induced acute lung injury is aggravated in Streptozotocin diabetic mice. Exp Lung Res 2014; 41:146-54. [DOI: 10.3109/01902148.2014.983280] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
31
|
Mehta JL, Ding Z, Liu S, Wang X, Khaidakov M. Hypertension, TLR4 activation in brain and cardiac hypertrophy. Cardiovasc Res 2014; 103:3-4. [PMID: 24835276 DOI: 10.1093/cvr/cvu128] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Jawahar L Mehta
- Central Arkansas Veterans Healthcare System, Little Rock, AR, USA Division of Cardiovascular Medicine, The University of Arkansas for Medical Sciences, Little Rock, AR 72212, USA
| | - Zufeng Ding
- Central Arkansas Veterans Healthcare System, Little Rock, AR, USA Division of Cardiovascular Medicine, The University of Arkansas for Medical Sciences, Little Rock, AR 72212, USA Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Shijie Liu
- Central Arkansas Veterans Healthcare System, Little Rock, AR, USA Division of Cardiovascular Medicine, The University of Arkansas for Medical Sciences, Little Rock, AR 72212, USA Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Xianwei Wang
- Central Arkansas Veterans Healthcare System, Little Rock, AR, USA Division of Cardiovascular Medicine, The University of Arkansas for Medical Sciences, Little Rock, AR 72212, USA
| | - Magomed Khaidakov
- Central Arkansas Veterans Healthcare System, Little Rock, AR, USA Division of Cardiovascular Medicine, The University of Arkansas for Medical Sciences, Little Rock, AR 72212, USA
| |
Collapse
|
32
|
Sen P, Dharmadhikari AV, Majewski T, Mohammad MA, Kalin TV, Zabielska J, Ren X, Bray M, Brown HM, Welty S, Thevananther S, Langston C, Szafranski P, Justice MJ, Kalinichenko VV, Gambin A, Belmont J, Stankiewicz P. Comparative analyses of lung transcriptomes in patients with alveolar capillary dysplasia with misalignment of pulmonary veins and in foxf1 heterozygous knockout mice. PLoS One 2014; 9:e94390. [PMID: 24722050 PMCID: PMC3983164 DOI: 10.1371/journal.pone.0094390] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2013] [Accepted: 03/14/2014] [Indexed: 12/24/2022] Open
Abstract
Alveolar Capillary Dysplasia with Misalignment of Pulmonary Veins (ACDMPV) is a developmental disorder of the lungs, primarily affecting their vasculature. FOXF1 haploinsufficiency due to heterozygous genomic deletions and point mutations have been reported in most patients with ACDMPV. The majority of mice with heterozygous loss-of-function of Foxf1 exhibit neonatal lethality with evidence of pulmonary hemorrhage in some of them. By comparing transcriptomes of human ACDMPV lungs with control lungs using expression arrays, we found that several genes and pathways involved in lung development, angiogenesis, and in pulmonary hypertension development, were deregulated. Similar transcriptional changes were found in lungs of the postnatal day 0.5 Foxf1+/− mice when compared to their wildtype littermate controls; 14 genes, COL15A1, COL18A1, COL6A2, ESM1, FSCN1, GRINA, IGFBP3, IL1B, MALL, NOS3, RASL11B, MATN2, PRKCDBP, and SIRPA, were found common to both ACDMPV and Foxf1 heterozygous lungs. Our results advance knowledge toward understanding of the molecular mechanism of ACDMPV, lung development, and its vasculature pathology. These data may also be useful for understanding etiologies of other lung disorders, e.g. pulmonary hypertension, bronchopulmonary dysplasia, or cancer.
Collapse
Affiliation(s)
- Partha Sen
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, United States of America
- * E-mail: (P. Sen); (P. Stankiewicz)
| | - Avinash V. Dharmadhikari
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
- Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, Texas, United States of America
| | - Tadeusz Majewski
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Mahmoud A. Mohammad
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Tanya V. Kalin
- Division of Pulmonary Biology, Cincinnati Children's Hospital Research Foundation, Cincinnati, Ohio, United States of America
| | | | - Xiaomeng Ren
- Division of Pulmonary Biology, Cincinnati Children's Hospital Research Foundation, Cincinnati, Ohio, United States of America
| | - Molly Bray
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Hannah M. Brown
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
- Robinson Research Institute, School of Pediatrics and Reproductive Health, University of Adelaide, Adelaide, Australia
| | - Stephen Welty
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Sundararajah Thevananther
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, United States of America
- Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, Texas, United States of America
| | - Claire Langston
- Department of Pathology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Przemyslaw Szafranski
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Monica J. Justice
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
- Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, Texas, United States of America
| | - Vladimir V. Kalinichenko
- Division of Pulmonary Biology, Cincinnati Children's Hospital Research Foundation, Cincinnati, Ohio, United States of America
| | - Anna Gambin
- Institute of Informatics, University of Warsaw, Warsaw, Poland
- Mossakowski Medical Research Center, Polish Academy of Sciences, Warsaw, Poland
| | - John Belmont
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Pawel Stankiewicz
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
- Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, Texas, United States of America
- * E-mail: (P. Sen); (P. Stankiewicz)
| |
Collapse
|
33
|
Ball MK, Waypa GB, Mungai PT, Nielsen JM, Czech L, Dudley VJ, Beussink L, Dettman RW, Berkelhamer SK, Steinhorn RH, Shah SJ, Schumacker PT. Regulation of hypoxia-induced pulmonary hypertension by vascular smooth muscle hypoxia-inducible factor-1α. Am J Respir Crit Care Med 2014. [PMID: 24251580 DOI: 10.1164/rccm.201302-03020c] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
RATIONALE Chronic hypoxia induces pulmonary vascular remodeling, pulmonary hypertension, and right ventricular hypertrophy. At present, little is known about mechanisms driving these responses. Hypoxia-inducible factor-1α (HIF-1α) is a master regulator of transcription in hypoxic cells, up-regulating genes involved in energy metabolism, proliferation, and extracellular matrix reorganization. Systemic loss of a single HIF-1α allele has been shown to attenuate hypoxic pulmonary hypertension, but the cells contributing to this response have not been identified. OBJECTIVES We sought to determine the contribution of HIF-1α in smooth muscle on pulmonary vascular and right heart responses to chronic hypoxia. METHODS We used mice with homozygous conditional deletion of HIF-1α combined with tamoxifen-inducible smooth muscle-specific Cre recombinase expression. Mice received either tamoxifen or vehicle followed by exposure to either normoxia or chronic hypoxia (10% O2) for 30 days before measurement of cardiopulmonary responses. MEASUREMENTS AND MAIN RESULTS Tamoxifen-induced smooth muscle-specific deletion of HIF-1α attenuated pulmonary vascular remodeling and pulmonary hypertension in chronic hypoxia. However, right ventricular hypertrophy was unchanged despite attenuated pulmonary pressures. CONCLUSIONS These results indicate that HIF-1α in smooth muscle contributes to pulmonary vascular remodeling and pulmonary hypertension in chronic hypoxia. However, loss of HIF-1 function in smooth muscle does not affect hypoxic cardiac remodeling, suggesting that the cardiac hypertrophy response is not directly coupled to the increase in pulmonary artery pressure.
Collapse
|
34
|
Ball MK, Waypa GB, Mungai PT, Nielsen JM, Czech L, Dudley VJ, Beussink L, Dettman RW, Berkelhamer SK, Steinhorn RH, Shah SJ, Schumacker PT. Regulation of hypoxia-induced pulmonary hypertension by vascular smooth muscle hypoxia-inducible factor-1α. Am J Respir Crit Care Med 2014; 189:314-24. [PMID: 24251580 DOI: 10.1164/rccm.201302-0302oc] [Citation(s) in RCA: 198] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
RATIONALE Chronic hypoxia induces pulmonary vascular remodeling, pulmonary hypertension, and right ventricular hypertrophy. At present, little is known about mechanisms driving these responses. Hypoxia-inducible factor-1α (HIF-1α) is a master regulator of transcription in hypoxic cells, up-regulating genes involved in energy metabolism, proliferation, and extracellular matrix reorganization. Systemic loss of a single HIF-1α allele has been shown to attenuate hypoxic pulmonary hypertension, but the cells contributing to this response have not been identified. OBJECTIVES We sought to determine the contribution of HIF-1α in smooth muscle on pulmonary vascular and right heart responses to chronic hypoxia. METHODS We used mice with homozygous conditional deletion of HIF-1α combined with tamoxifen-inducible smooth muscle-specific Cre recombinase expression. Mice received either tamoxifen or vehicle followed by exposure to either normoxia or chronic hypoxia (10% O2) for 30 days before measurement of cardiopulmonary responses. MEASUREMENTS AND MAIN RESULTS Tamoxifen-induced smooth muscle-specific deletion of HIF-1α attenuated pulmonary vascular remodeling and pulmonary hypertension in chronic hypoxia. However, right ventricular hypertrophy was unchanged despite attenuated pulmonary pressures. CONCLUSIONS These results indicate that HIF-1α in smooth muscle contributes to pulmonary vascular remodeling and pulmonary hypertension in chronic hypoxia. However, loss of HIF-1 function in smooth muscle does not affect hypoxic cardiac remodeling, suggesting that the cardiac hypertrophy response is not directly coupled to the increase in pulmonary artery pressure.
Collapse
|
35
|
Bauer EM, Chanthaphavong RS, Sodhi CP, Hackam DJ, Billiar TR, Bauer PM. Genetic deletion of toll-like receptor 4 on platelets attenuates experimental pulmonary hypertension. Circ Res 2014; 114:1596-600. [PMID: 24637196 DOI: 10.1161/circresaha.114.303662] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
RATIONALE Recent studies demonstrate a role for toll-like receptor 4 (TLR4) in the pathogenesis of pulmonary hypertension (PH); however, the cell types involved in mediating the effects of TLR4 remain unknown. OBJECTIVES The objective of this study was to determine the contribution of TLR4 expressed on nonparenchymal cells to the pathogenesis of PH. METHODS AND RESULTS TLR4 bone marrow chimeric mice revealed an equal contribution of TLR4 on nonparenchymal and parenchymal cells in the pathogenesis of PH as determined by measuring right ventricular (RV) systolic pressure and RV hypertrophy. However, the deletion of TLR4 from myeloid lineage cells had no effect on the development of PH because we found no difference in RV systolic pressure or RV hypertrophy in wild-type versus LysM-TLR4(-/-) mice. To explore the potential role of platelet TLR4 in the pathogenesis of PH, platelet-specific TLR4(-/-) mice were generated (PF4-TLR4(-/-) mice). TLR4(-/-) platelets from either global TLR4(-/-) or PF4-TLR4(-/-) mice were functional but failed to respond to lipopolysaccharide, demonstrating a lack of TLR4. PF4-TLR4(-/-) mice demonstrated significant protection from hypoxia-induced PH, including attenuated increases in RV systolic pressure and RV hypertrophy, decreased platelet activation, and less pulmonary vascular remodeling. The deletion of TLR4 from platelets attenuated serotonin release after chronic hypoxia, and lipopolysaccharide-stimulated platelets released serotonin and promoted pulmonary artery smooth muscle cell proliferation in a serotonin-dependent manner. CONCLUSIONS Our data demonstrate that TLR4 on platelets contributes to the pathogenesis of PH and further highlights the role of platelets in PH.
Collapse
Affiliation(s)
- Eileen M Bauer
- From the Department of Surgery (E.M.B., R.S.C., C.P.S., D.J.H., T.R.B., P.M.B.), University of Pittsburgh Cancer Institute (E.M.B.), Division of Pediatric Surgery, Children's Hospital Pittsburgh (C.P.S., D.J.H.), Department of Pharmacology and Chemical Biology (P.M.B.), and Vascular Medicine Institute (P.M.B.), University of Pittsburgh School of Medicine, PA
| | | | | | | | | | | |
Collapse
|
36
|
Bauer EM, Shapiro R, Billiar TR, Bauer PM. High mobility group Box 1 inhibits human pulmonary artery endothelial cell migration via a Toll-like receptor 4- and interferon response factor 3-dependent mechanism(s). J Biol Chem 2012; 288:1365-73. [PMID: 23148224 DOI: 10.1074/jbc.m112.434142] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
In pulmonary hypertension the loss of precapillary arterioles results from vascular injury causing endothelial dysfunction. Endothelial cell migration and proliferation are critical for vascular regeneration. This study focused on the effect of high mobility group box 1 protein (HMGB1) on these critical processes. HMGB1 had no effect on human pulmonary artery endothelial cell (HPAEC) proliferation. In contrast, treatment of HPAECs with HMGB1 dose-dependently inhibited VEGF-stimulated HPAEC migration. The effect of HMGB1 on HPAEC migration was TLR4-dependent because it was reversed by TLR4 siRNA or TLR4-neutralizing antibody. Exposure of HPAECs to hypoxia caused translocation and release of HMGB1 and inhibition of HPAEC migration. The effect of hypoxia on HPAEC migration was mediated by HMGB1 because HMGB1-neutralizing antibody but not control IgG restored HPAEC migration. Likewise, TLR4 siRNA but not control siRNA reversed the inhibitory effect of hypoxia in HPAECs. The canonical TLR4 signaling pathway requires the adaptor protein MyD88 and leads to downstream NFκB activation. Interestingly, HMGB1 failed to stimulate NFκB translocation to the nucleus, but instead activated an alternative pathway characterized by activation of interferon response factor 3 (IRF3). This was in contrast to human umbilical vein endothelial cells in which HMGB1 stimulated nuclear translocation of NFκB but not IRF3. IRF3 siRNA, but not MyD88 siRNA, reversed the inhibitory effect of HMGB1 on HPAEC migration. These data demonstrate that HMGB1 inhibits HPAEC migration, a critical process for vascular regeneration, via TLR4- and IRF3-dependent mechanisms.
Collapse
Affiliation(s)
- Eileen M Bauer
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213, USA
| | | | | | | |
Collapse
|
37
|
Porter KM, Sutliff RL. HIV-1, reactive oxygen species, and vascular complications. Free Radic Biol Med 2012; 53:143-59. [PMID: 22564529 PMCID: PMC3377788 DOI: 10.1016/j.freeradbiomed.2012.03.019] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2011] [Revised: 03/16/2012] [Accepted: 03/18/2012] [Indexed: 02/07/2023]
Abstract
Over 1 million people in the United States and 33 million individuals worldwide suffer from HIV/AIDS. Since its discovery, HIV/AIDS has been associated with an increased susceptibility to opportunistic infection due to immune dysfunction. Highly active antiretroviral therapies restore immune function and, as a result, people infected with HIV-1 are living longer. This improved survival of HIV-1 patients has revealed a previously unrecognized risk of developing vascular complications, such as atherosclerosis and pulmonary hypertension. The mechanisms underlying these HIV-associated vascular disorders are poorly understood. However, HIV-induced elevations in reactive oxygen species (ROS), including superoxide and hydrogen peroxide, may contribute to vascular disease development and progression by altering cell function and redox-sensitive signaling pathways. In this review, we summarize the clinical and experimental evidence demonstrating HIV- and HIV antiretroviral therapy-induced alterations in reactive oxygen species and how these effects are likely to contribute to vascular dysfunction and disease.
Collapse
Affiliation(s)
- Kristi M Porter
- Pulmonary, Allergy and Critical Care Division, Emory University School of Medicine/Atlanta VA Medical Center, 1670 Clairmont Road, Mailstop 151P, Decatur, GA 30033, USA.
| | | |
Collapse
|
38
|
The essential roles of Toll-like receptor signaling pathways in sterile inflammatory diseases. Int Immunopharmacol 2011; 11:1422-32. [PMID: 21600309 DOI: 10.1016/j.intimp.2011.04.026] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2011] [Revised: 04/29/2011] [Accepted: 04/30/2011] [Indexed: 02/06/2023]
Abstract
Toll-like receptors (TLRs) form a family of pattern recognition receptors with at least 11 members in human and 13 in mouse. TLRs recognize a wide variety of putative host-derived agonists that have emerged as key mediators of innate immunity. TLR signaling also plays an important role in the activation of the adaptive immune system by inducing pro-inflammatory cytokines and upregulating costimulatory molecules of antigen presenting cells. Inappropriate activation of TLRs by self-components generated by damaged tissues may result in sterile inflammation. This review discusses the contribution of TLR signaling to the initiation and progression of non-infectious inflammatory processes, such as ischemia and reperfusion (I/R) injury, tissue repair and regeneration and autoimmune diseases. The involvement of TLR signaling in the pathogenesis of sterile inflammation-related diseases may provide novel targets for the development of therapeutics.
Collapse
|