1
|
Li Z, Bu Y, Wang C, Yu Y, Han L, Liu C, Chen G, Li C, Zhang Y, Cao H, Ma Z, Yue Z. Extracellular vesicle-packaged GBP2 from macrophages aggravates sepsis-induced acute lung injury by promoting ferroptosis in pulmonary vascular endothelial cells. Redox Biol 2025; 82:103614. [PMID: 40156957 PMCID: PMC11994402 DOI: 10.1016/j.redox.2025.103614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 03/13/2025] [Accepted: 03/24/2025] [Indexed: 04/01/2025] Open
Abstract
Macrophages play a critical role in the development of sepsis-induced acute lung injury (si-ALI), with extracellular vesicles (EVs) acting as crucial mediators. However, the effects and mechanisms of macrophage-derived EVs on si-ALI remain unclear. This study demonstrated that macrophage-derived EVs induce endothelial ferroptosis and barrier disruption during sepsis. Through proteomic sequencing and reanalysis of transcriptomic and single-cell sequencing data, guanylate-binding protein 2 (GBP2) was identified as a key EV molecule. Elevated GBP2 expression was observed in EVs and monocytes from the peripheral blood of sepsis patients, in LPS-stimulated THP-1 and RAW264.7 cells and their secreted EVs, and in macrophages within the lungs of CLP mice. Additionally, GBP2 expression in EVs showed a positive correlation with vascular barrier injury biomarkers, including ANGPT2, Syndecan-1, and sTM. Modulating GBP2 levels in macrophage-derived EVs affected EV-induced ferroptosis in endothelial cells. The mechanism by which GBP2 binds directly to OTUD5 and promotes GPX4 ubiquitination was elucidated using RNA interference, adeno-associated virus transfection, and endothelial-specific Gpx4 knockout mice. A high-throughput screening of small-molecule compounds targeting GBP2 was conducted. Molecular docking, molecular dynamics simulations, and cellular thermal shift assays further confirmed that Plantainoside D (PD) has a potent binding affinity for GBP2. PD treatment inhibited the interaction between GBP2 and OTUD5, leading to a reduction in GPX4 ubiquitination. Further research revealed that PD treatment enhanced the pulmonary protective effects of GBP2 inhibition. In conclusion, this study explored the role of EV-mediated signaling between macrophages and pulmonary vascular endothelial cells in si-ALI, highlighting the GBP2-OTUD5-GPX4 axis as a driver of endothelial ferroptosis and lung injury. Targeting this signaling axis presents a potential therapeutic strategy for si-ALI.
Collapse
Affiliation(s)
- Zhixi Li
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, PR China; The Key Laboratory of Anesthesiology and Intensive Care Research of Heilongjiang Province, Harbin, 150001, PR China; The Key Laboratory of Myocardial Ischemia Organization, Chinese Ministry of Education, Harbin, 150001, PR China; State Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin, 150001, PR China
| | - Yue Bu
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, PR China; The Key Laboratory of Anesthesiology and Intensive Care Research of Heilongjiang Province, Harbin, 150001, PR China; Department of Pain Medicine, Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, PR China
| | - Cheng Wang
- Department of Environmental Hygiene, School of Public Health, Harbin Medical University, Harbin, 150081, PR China
| | - Yongjing Yu
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, PR China; The Key Laboratory of Anesthesiology and Intensive Care Research of Heilongjiang Province, Harbin, 150001, PR China; The Key Laboratory of Myocardial Ischemia Organization, Chinese Ministry of Education, Harbin, 150001, PR China; State Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin, 150001, PR China
| | - Lei Han
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, PR China; The Key Laboratory of Anesthesiology and Intensive Care Research of Heilongjiang Province, Harbin, 150001, PR China
| | - Chang Liu
- The Key Laboratory of Anesthesiology and Intensive Care Research of Heilongjiang Province, Harbin, 150001, PR China; The Key Laboratory of Myocardial Ischemia Organization, Chinese Ministry of Education, Harbin, 150001, PR China; State Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin, 150001, PR China; Department of Anesthesiology, Harbin Medical University Cancer Hospital, Harbin, 150081, PR China
| | - Guangmin Chen
- Department of Anesthesiology, First Affiliated Hospital of Harbin Medical University, 199 Dazhi Road, Harbin, 150001, PR China
| | - Chenglong Li
- Department of Anesthesiology, Fourth Affiliated Hospital of Harbin Medical University, 37 Yiyuan Road, Harbin, 150001, PR China
| | - Yan Zhang
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, PR China; The Key Laboratory of Anesthesiology and Intensive Care Research of Heilongjiang Province, Harbin, 150001, PR China
| | - Hang Cao
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, PR China; The Key Laboratory of Anesthesiology and Intensive Care Research of Heilongjiang Province, Harbin, 150001, PR China
| | - Zhaoxue Ma
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, PR China; The Key Laboratory of Anesthesiology and Intensive Care Research of Heilongjiang Province, Harbin, 150001, PR China
| | - Ziyong Yue
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, PR China; The Key Laboratory of Anesthesiology and Intensive Care Research of Heilongjiang Province, Harbin, 150001, PR China.
| |
Collapse
|
2
|
Song XY, Sun Q, Wei SZ, Wang HR, Wang Y, Zhang WB, Ren C, Song XC, Mou YK. IL-6 mediates olfactory dysfunction in a mouse model of allergic rhinitis. Brain Res 2024; 1833:148885. [PMID: 38531465 DOI: 10.1016/j.brainres.2024.148885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 03/07/2024] [Accepted: 03/21/2024] [Indexed: 03/28/2024]
Abstract
BACKGROUND Immune-inflammatory response is a key element in the occurrence and development of olfactory dysfunction (OD) in patients with allergic rhinitis (AR). As one of the core factors in immune-inflammatory responses, interleukin (IL)-6 is closely related to the pathogenesis of allergic diseases. It may also play an important role in OD induced by diseases, such as Sjögren's syndrome and coronavirus disease 2019. However, there is no study has reported its role in OD in AR. Thus, this study aimed to investigate the role of IL-6 in AR-related OD, in an attempt to discover a new target for the prevention and treatment of OD in patients with AR. METHODS Differential expression analysis was performed using the public datasets GSE52804 and GSE140454 for AR, and differentially expressed genes (DEGs) were obtained by obtaining the intersection points between these two datasets. IL-6, a common differential factor, was obtained by intersecting the DEGs with the General Olfactory Sensitivity Database (GOSdb) again. A model of AR mice with OD was developed by sensitizing with ovalbumin (OVA) to verify the reliability of IL-6 as a key factor of OD in AR and explore the potential mechanisms. Furthermore, a supernatant and microglia co-culture model of nasal mucosa epithelial cells stimulated by the allergen house dust mite extract Derp1 was established to identify the cellular and molecular mechanisms of IL-6-mediated OD in AR. RESULTS The level of IL-6 in the nasal mucosa and olfactory bulb of AR mice with OD significantly increased and showed a positive correlation with the expression of olfactory bulb microglia marker Iba-1 and the severity of OD. In-vitro experiments showed that the level of IL-6 significantly increased in the supernatant after the nasal mucosa epithelial cells were stimulated by Derp1, along with significantly decreased barrier function of the nasal mucosa. The expression levels of neuroinflammatory markers IL-1β and INOS increased after a conditioned culture of microglia with the supernatant including IL-6. Then knockdown (KD) of IL-6R by small interfering RNA (siRNA), the expression of IL-1β and INOS significantly diminished. CONCLUSION IL-6 plays a key role in the occurrence and development of OD in AR, which may be related to its effect on olfactory bulb microglia-mediated neuroinflammation.
Collapse
Affiliation(s)
- Xiao-Yu Song
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China; Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, China; Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, China
| | - Qi Sun
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China; Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, China; Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, China
| | - Shi-Zhuang Wei
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China; Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, China; Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, China
| | - Han-Rui Wang
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China; Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, China; Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, China
| | - Yao Wang
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China; Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, China; Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, China
| | - Wen-Bin Zhang
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China; Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, China; Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, China
| | - Chao Ren
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China; Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, China; Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, China.
| | - Xi-Cheng Song
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China; Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, China; Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, China.
| | - Ya-Kui Mou
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China; Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, China; Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, China.
| |
Collapse
|
3
|
Li Z, Yu Y, Bu Y, Liu C, Jin J, Li W, Chen G, Liu E, Zhang Y, Gong W, Luo J, Yue Z. QiShenYiQi pills preserve endothelial barrier integrity to mitigate sepsis-induced acute lung injury by inhibiting ferroptosis. JOURNAL OF ETHNOPHARMACOLOGY 2024; 322:117610. [PMID: 38122915 DOI: 10.1016/j.jep.2023.117610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 11/28/2023] [Accepted: 12/14/2023] [Indexed: 12/23/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The QiShengYiQi pill (QSYQ) is a traditional Chinese medicinal formulation. The effectiveness and safety of QSYQ in treating respiratory system disorders have been confirmed. Its pharmacological actions include anti-inflammation, antioxidative stress, and improving energy metabolism. However, the mechanism of QSYQ in treating sepsis-induced acute lung injury (si-ALI) remains unclear. AIM OF THE STUDY Si-ALI presents a clinical challenge with high incidence and mortality rates. This study aims to confirm the efficacy of QSYQ in si-ALI and to explore the potential mechanisms, providing a scientific foundation for its application and insights for optimizing treatment strategies and identifying potential active components. MATERIALS AND METHODS The impact of QSYQ on si-ALI was evaluated using the cecal ligation and puncture (CLP) experimental sepsis animal model. The effects of QSYQ on endothelial cells were observed through coculturing with LPS-stimulated macrophage-conditioned medium. Inflammatory cytokine levels, HE staining, Evans blue staining, lung wet/dry ratio, and cell count and protein content in bronchoalveolar lavage fluid were used to assess the degree of lung injury. Network pharmacology was utilized to investigate the potential mechanisms of QSYQ in treating si-ALI. Western blot and immunofluorescence analyses were used to evaluate barrier integrity and validate mechanistically relevant proteins. RESULTS QSYQ reduced the inflammation and alleviated pulmonary vascular barrier damage in CLP mice (all P < 0.05). A total of 127 potential targets through which QSYQ regulates si-ALI were identified, predominantly enriched in the RAGE pathway. The results of protein-protein interaction analysis suggest that COX2, a well-established critical marker of ferroptosis, is among the key targets. In vitro and in vivo studies demonstrated that QSYQ mitigated ferroptosis and vascular barrier damage in sepsis (all P < 0.05), accompanied by a reduction in oxidative stress and the inhibition of the COX2 and RAGE (all P < 0.05). CONCLUSIONS This study demonstrated that QSYQ maintains pulmonary vascular barrier integrity by inhibiting ferroptosis in CLP mice. These findings partially elucidate the mechanism of QSYQ in si-ALI and further clarify the active components of QSYQ, thereby providing a scientific theoretical basis for treating si-ALI with QSYQ.
Collapse
Affiliation(s)
- Zhixi Li
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Harbin, 150001, PR China; Heilongjiang Province Key Laboratory of Research on Anesthesiology and Critical Care Medicine, 246 Xuefu Road, Harbin, 150001, PR China; The Key Laboratory of Myocardial Ischemia Organization, Chinese Ministry of Education, 246 Xuefu Road, Harbin, 150001, PR China
| | - Yongjing Yu
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Harbin, 150001, PR China; Heilongjiang Province Key Laboratory of Research on Anesthesiology and Critical Care Medicine, 246 Xuefu Road, Harbin, 150001, PR China; The Key Laboratory of Myocardial Ischemia Organization, Chinese Ministry of Education, 246 Xuefu Road, Harbin, 150001, PR China
| | - Yue Bu
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Harbin, 150001, PR China; Heilongjiang Province Key Laboratory of Research on Anesthesiology and Critical Care Medicine, 246 Xuefu Road, Harbin, 150001, PR China; Department of Pain Medicine, Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Harbin, 150001, PR China
| | - Chang Liu
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Harbin, 150001, PR China; Heilongjiang Province Key Laboratory of Research on Anesthesiology and Critical Care Medicine, 246 Xuefu Road, Harbin, 150001, PR China; The Key Laboratory of Myocardial Ischemia Organization, Chinese Ministry of Education, 246 Xuefu Road, Harbin, 150001, PR China
| | - Jiaqi Jin
- The Key Laboratory of Myocardial Ischemia Organization, Chinese Ministry of Education, 246 Xuefu Road, Harbin, 150001, PR China; Department of Neurology, Xuanwu Hospital, Capital Medical University, 45 Changchun Road, Beijing, 100053, PR China
| | - Wenqiang Li
- Department of Vascular Surgery, Jinshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, PR China
| | - Guangmin Chen
- Department of Anesthesiology, First Affiliated Hospital of Harbin Medical University, 199 Dazhi Road, Harbin, 150001, PR China
| | - Enran Liu
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Harbin, 150001, PR China; Heilongjiang Province Key Laboratory of Research on Anesthesiology and Critical Care Medicine, 246 Xuefu Road, Harbin, 150001, PR China
| | - Yan Zhang
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Harbin, 150001, PR China; Heilongjiang Province Key Laboratory of Research on Anesthesiology and Critical Care Medicine, 246 Xuefu Road, Harbin, 150001, PR China
| | - Weidong Gong
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Harbin, 150001, PR China; Heilongjiang Province Key Laboratory of Research on Anesthesiology and Critical Care Medicine, 246 Xuefu Road, Harbin, 150001, PR China
| | - Juan Luo
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Harbin, 150001, PR China; Heilongjiang Province Key Laboratory of Research on Anesthesiology and Critical Care Medicine, 246 Xuefu Road, Harbin, 150001, PR China
| | - Ziyong Yue
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Harbin, 150001, PR China; Heilongjiang Province Key Laboratory of Research on Anesthesiology and Critical Care Medicine, 246 Xuefu Road, Harbin, 150001, PR China.
| |
Collapse
|
4
|
Mueller B. Episodic Migraine and POTS. Curr Pain Headache Rep 2023; 27:757-763. [PMID: 37804458 DOI: 10.1007/s11916-023-01173-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/15/2023] [Indexed: 10/09/2023]
Abstract
PURPOSE OF REVIEW Migraine is prevalent in patients with postural orthostatic tachycardia syndrome (POTS). The purpose of this review is to summarize and interpret studies that examine stress response systems in patients with migraine, focusing on their relevance to the pathologies associated with POTS. Important structural and functional components of the stress response network are also reviewed. RECENT FINDINGS In patients with migraine, studies examining the autonomic nervous system have demonstrated interictal sympathetic hypofunction and ictal sympathetic hyperfunction, while those focusing on the hypothalamic-pituitary-adrenal axis have demonstrated elevated responsivity. There is evidence that activation of these stress response systems during a migraine episode may exacerbate vascular dysfunction and play a role in the development of central sensitization. Activation of the stress response systems during an episode of migraine has the potential to exacerbate the pathology of POTS. Treatment approaches for the patient with comorbid episodic migraine and POTS should consider the etiology of POTS.
Collapse
Affiliation(s)
- Bridget Mueller
- Department of Neurology, Icahn School of Medicine at Mount Sinai, 5 East 98th Street, Box 1139, New York, NY, 10029, USA.
| |
Collapse
|
5
|
Chen X, Yuan S, Mi L, Long Y, He H. Pannexin1: insight into inflammatory conditions and its potential involvement in multiple organ dysfunction syndrome. Front Immunol 2023; 14:1217366. [PMID: 37711629 PMCID: PMC10498923 DOI: 10.3389/fimmu.2023.1217366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 08/10/2023] [Indexed: 09/16/2023] Open
Abstract
Sepsis represents a global health concern, and patients with severe sepsis are at risk of experiencing MODS (multiple organ dysfunction syndrome), which is associated with elevated mortality rates and a poorer prognosis. The development of sepsis involves hyperactive inflammation, immune disorder, and disrupted microcirculation. It is crucial to identify targets within these processes to develop therapeutic interventions. One such potential target is Panx1 (pannexin-1), a widely expressed transmembrane protein that facilitates the passage of molecules smaller than 1 KDa, such as ATP. Accumulating evidence has implicated the involvement of Panx1 in sepsis-associated MODS. It attracts immune cells via the purinergic signaling pathway, mediates immune responses via the Panx1-IL-33 axis, promotes immune cell apoptosis, regulates blood flow by modulating VSMCs' and vascular endothelial cells' tension, and disrupts microcirculation by elevating endothelial permeability and promoting microthrombosis. At the level of organs, Panx1 contributes to inflammatory injury in multiple organs. Panx1 primarily exacerbates injury and hinders recovery, making it a potential target for sepsis-induced MODS. While no drugs have been developed explicitly against Panx1, some compounds that inhibit Panx1 hemichannels have been used extensively in experiments. However, given that Panx1's role may vary during different phases of sepsis, more investigations are required before interventions against Panx1 can be applied in clinical. Overall, Panx1 may be a promising target for sepsis-induced MODS. Nevertheless, further research is needed to understand its complex role in different stages of sepsis fully and to develop suitable pharmaceutical interventions for clinical use.
Collapse
Affiliation(s)
| | | | | | - Yun Long
- Department of Critical Care Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Huaiwu He
- Department of Critical Care Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
6
|
Wang LJ, Feng F, Li JC, Chen TT, Liu LP. Role of heparanase in pulmonary hypertension. Front Pharmacol 2023; 14:1202676. [PMID: 37637421 PMCID: PMC10450954 DOI: 10.3389/fphar.2023.1202676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Accepted: 07/26/2023] [Indexed: 08/29/2023] Open
Abstract
Pulmonary hypertension (PH) is a pathophysiological condition of increased pulmonary circulation vascular resistance due to various reasons, which mainly leads to right heart dysfunction and even death, especially in critically ill patients. Although drug interventions have shown some efficacy in improving the hemodynamics of PH patients, the mortality rate remains high. Hence, the identification of new targets and treatment strategies for PH is imperative. Heparanase (HPA) is an enzyme that specifically cleaves the heparan sulfate (HS) side chains in the extracellular matrix, playing critical roles in inflammation and tumorigenesis. Recent studies have indicated a close association between HPA and PH, suggesting HPA as a potential therapeutic target. This review examines the involvement of HPA in PH pathogenesis, including its effects on endothelial cells, inflammation, and coagulation. Furthermore, HPA may serve as a biomarker for diagnosing PH, and the development of HPA inhibitors holds promise as a targeted therapy for PH treatment.
Collapse
Affiliation(s)
- Lin-Jun Wang
- The First Clinical Medical School of Lanzhou University, Lanzhou, Gansu, China
| | - Fei Feng
- The First Clinical Medical School of Lanzhou University, Lanzhou, Gansu, China
| | - Jian-Chun Li
- The First Clinical Medical School of Lanzhou University, Lanzhou, Gansu, China
| | - Ting-Ting Chen
- The First Clinical Medical School of Lanzhou University, Lanzhou, Gansu, China
| | - Li-Ping Liu
- The First Clinical Medical School of Lanzhou University, Lanzhou, Gansu, China
- Departments of Emergency Critical Care Medicine, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| |
Collapse
|
7
|
Villegas-Esguevillas M, Cho S, Vera-Zambrano A, Kwon JW, Barreira B, Telli G, Navarro-Dorado J, Morales-Cano D, de Olaiz B, Moreno L, Greenwood I, Pérez-Vizcaíno F, Kim SJ, Climent B, Cogolludo A. The novel K V7 channel activator URO-K10 exerts enhanced pulmonary vascular effects independent of the KCNE4 regulatory subunit. Biomed Pharmacother 2023; 164:114952. [PMID: 37295249 DOI: 10.1016/j.biopha.2023.114952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 05/23/2023] [Accepted: 05/27/2023] [Indexed: 06/12/2023] Open
Abstract
KV7 channels exert a pivotal role regulating vascular tone in several vascular beds. In this context, KV7 channel agonists represent an attractive strategy for the treatment of pulmonary arterial hypertension (PAH). Therefore, in this study, we have explored the pulmonary vascular effects of the novel KV7 channel agonist URO-K10. Consequently, the vasodilator and electrophysiological effects of URO-K10 were tested in rat and human pulmonary arteries (PA) and PA smooth muscle cells (PASMC) using myography and patch-clamp techniques. Protein expression was also determined by Western blot. Morpholino-induced knockdown of KCNE4 was assessed in isolated PA. PASMC proliferation was measured by BrdU incorporation assay. In summary, our data show that URO-K10 is a more effective relaxant of PA than the classical KV7 activators retigabine and flupirtine. URO-K10 enhanced KV currents in PASMC and its electrophysiological and relaxant effects were inhibited by the KV7 channel blocker XE991. The effects of URO-K10 were confirmed in human PA. URO-K10 also exhibited antiproliferative effects in human PASMC. Unlike retigabine and flupirtine, URO-K10-induced pulmonary vasodilation was not affected by morpholino-induced knockdown of the KCNE4 regulatory subunit. Noteworthy, the pulmonary vasodilator efficacy of this compound was considerably increased under conditions mimicking the ionic remodelling (as an in vitro model of PAH) and in PA from monocrotaline-induced pulmonary hypertensive rats. Taking all together, URO-K10 behaves as a KCNE4-independent KV7 channel activator with much increased pulmonary vascular effects compared to classical KV7 channel activators. Our study identifies a promising new drug in the context of PAH.
Collapse
Affiliation(s)
- Marta Villegas-Esguevillas
- Departamento de Farmacología, Facultad de Medicina, Universidad Complutense, Madrid, Spain; Institute of Health Research Gregorio Marañón (IiSGM), Madrid, Spain; CIBER Enfermedades Respiratorias (Ciberes), Madrid, Spain
| | - Suhan Cho
- Department of Physiology, College of Medicine, Seoul National University, Seoul, South Korea
| | - Alba Vera-Zambrano
- Departamento de Farmacología, Facultad de Medicina, Universidad Complutense, Madrid, Spain; Institute of Health Research Gregorio Marañón (IiSGM), Madrid, Spain; CIBER Enfermedades Respiratorias (Ciberes), Madrid, Spain
| | - Jae Won Kwon
- Institute of Health Research Gregorio Marañón (IiSGM), Madrid, Spain
| | - Bianca Barreira
- Departamento de Farmacología, Facultad de Medicina, Universidad Complutense, Madrid, Spain; Institute of Health Research Gregorio Marañón (IiSGM), Madrid, Spain; CIBER Enfermedades Respiratorias (Ciberes), Madrid, Spain
| | - Göcken Telli
- Department of Pharmacology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Jorge Navarro-Dorado
- Departamento de Fisiología, Facultad de Farmacia, Universidad Complutense, Madrid, Spain
| | - Daniel Morales-Cano
- Departamento de Farmacología, Facultad de Medicina, Universidad Complutense, Madrid, Spain; Institute of Health Research Gregorio Marañón (IiSGM), Madrid, Spain; CIBER Enfermedades Respiratorias (Ciberes), Madrid, Spain
| | - Beatriz de Olaiz
- Department of Thoracic Surgery, Hospital Universitario de Getafe, Getafe, Spain
| | - Laura Moreno
- Departamento de Farmacología, Facultad de Medicina, Universidad Complutense, Madrid, Spain; Institute of Health Research Gregorio Marañón (IiSGM), Madrid, Spain; CIBER Enfermedades Respiratorias (Ciberes), Madrid, Spain
| | - Iain Greenwood
- Vascular Biology Research Centre, Institute of Molecular and Clinical Sciences, St George's University of London, United Kingdom
| | - Francisco Pérez-Vizcaíno
- Departamento de Farmacología, Facultad de Medicina, Universidad Complutense, Madrid, Spain; Institute of Health Research Gregorio Marañón (IiSGM), Madrid, Spain; CIBER Enfermedades Respiratorias (Ciberes), Madrid, Spain
| | - Sung Joon Kim
- Department of Physiology, College of Medicine, Seoul National University, Seoul, South Korea
| | - Belén Climent
- Departamento de Fisiología, Facultad de Farmacia, Universidad Complutense, Madrid, Spain.
| | - Angel Cogolludo
- Departamento de Farmacología, Facultad de Medicina, Universidad Complutense, Madrid, Spain; Institute of Health Research Gregorio Marañón (IiSGM), Madrid, Spain; CIBER Enfermedades Respiratorias (Ciberes), Madrid, Spain
| |
Collapse
|
8
|
Xu WJ, Wu Q, He WN, Wang S, Zhao YL, Huang JX, Yan XS, Jiang R. Interleukin-6 and pulmonary hypertension: from physiopathology to therapy. Front Immunol 2023; 14:1181987. [PMID: 37449201 PMCID: PMC10337993 DOI: 10.3389/fimmu.2023.1181987] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 06/12/2023] [Indexed: 07/18/2023] Open
Abstract
Pulmonary hypertension (PH) is a progressive, pulmonary vascular disease with high morbidity and mortality. Unfortunately, the pathogenesis of PH is complex and remains unclear. Existing studies have suggested that inflammatory factors are key factors in PH. Interleukin-6 (IL-6) is a multifunctional cytokine that plays a crucial role in the regulation of the immune system. Current studies reveal that IL-6 is elevated in the serum of patients with PH and it is negatively correlated with lung function in those patients. Since IL-6 is one of the most important mediators in the pathogenesis of inflammation in PH, signaling mechanisms targeting IL-6 may become therapeutic targets for this disease. In this review, we detailed the potential role of IL-6 in accelerating PH process and the specific mechanisms and signaling pathways. We also summarized the current drugs targeting these inflammatory pathways to treat PH. We hope that this study will provide a more theoretical basis for targeted treatment in patients with PH in the future.
Collapse
Affiliation(s)
- Wei-Jie Xu
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Qiong Wu
- Department of Pulmonary and Critical Care Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wen-Ni He
- Department of Cardiopulmonary Circulation, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Shang Wang
- Department of Cardiopulmonary Circulation, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Ya-Lin Zhao
- Department of Respiratory Critical Care Medicine, The First Hospital of Kunming, Kunming, China
| | - Jun-Xia Huang
- Department of Hematology, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Xue-Shen Yan
- Department of Hematology, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Rong Jiang
- Department of Cardiopulmonary Circulation, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
9
|
Chen P, Wu M, He Y, Jiang B, He ML. Metabolic alterations upon SARS-CoV-2 infection and potential therapeutic targets against coronavirus infection. Signal Transduct Target Ther 2023; 8:237. [PMID: 37286535 DOI: 10.1038/s41392-023-01510-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 04/18/2023] [Accepted: 05/19/2023] [Indexed: 06/09/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) caused by coronavirus SARS-CoV-2 infection has become a global pandemic due to the high viral transmissibility and pathogenesis, bringing enormous burden to our society. Most patients infected by SARS-CoV-2 are asymptomatic or have mild symptoms. Although only a small proportion of patients progressed to severe COVID-19 with symptoms including acute respiratory distress syndrome (ARDS), disseminated coagulopathy, and cardiovascular disorders, severe COVID-19 is accompanied by high mortality rates with near 7 million deaths. Nowadays, effective therapeutic patterns for severe COVID-19 are still lacking. It has been extensively reported that host metabolism plays essential roles in various physiological processes during virus infection. Many viruses manipulate host metabolism to avoid immunity, facilitate their own replication, or to initiate pathological response. Targeting the interaction between SARS-CoV-2 and host metabolism holds promise for developing therapeutic strategies. In this review, we summarize and discuss recent studies dedicated to uncovering the role of host metabolism during the life cycle of SARS-CoV-2 in aspects of entry, replication, assembly, and pathogenesis with an emphasis on glucose metabolism and lipid metabolism. Microbiota and long COVID-19 are also discussed. Ultimately, we recapitulate metabolism-modulating drugs repurposed for COVID-19 including statins, ASM inhibitors, NSAIDs, Montelukast, omega-3 fatty acids, 2-DG, and metformin.
Collapse
Affiliation(s)
- Peiran Chen
- Department of Biomedical Sciences, City University of Hong Kong, HKSAR, Hong Kong, China
| | - Mandi Wu
- Department of Biomedical Sciences, City University of Hong Kong, HKSAR, Hong Kong, China
| | - Yaqing He
- Shenzhen Center for Disease Control and Prevention, Shenzhen, 518055, Guangdong, China
| | - Binghua Jiang
- Cell Signaling and Proteomic Center, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Ming-Liang He
- Department of Biomedical Sciences, City University of Hong Kong, HKSAR, Hong Kong, China.
| |
Collapse
|
10
|
Morales-Cano D, Izquierdo-García JL, Barreira B, Esquivel-Ruiz S, Callejo M, Pandolfi R, Villa-Valverde P, Rodríguez I, Cogolludo A, Ruiz-Cabello J, Perez-Vizcaino F, Moreno L. Impact of a TAK-1 inhibitor as a single or as an add-on therapy to riociguat on the metabolic reprograming and pulmonary hypertension in the SUGEN5416/hypoxia rat model. Front Pharmacol 2023; 14:1021535. [PMID: 37063275 PMCID: PMC10090662 DOI: 10.3389/fphar.2023.1021535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 03/13/2023] [Indexed: 03/31/2023] Open
Abstract
Background: Despite increasing evidence suggesting that pulmonary arterial hypertension (PAH) is a complex disease involving vasoconstriction, thrombosis, inflammation, metabolic dysregulation and vascular proliferation, all the drugs approved for PAH mainly act as vasodilating agents. Since excessive TGF-β signaling is believed to be a critical factor in pulmonary vascular remodeling, we hypothesized that blocking TGFβ-activated kinase 1 (TAK-1), alone or in combination with a vasodilator therapy (i.e., riociguat) could achieve a greater therapeutic benefit.Methods: PAH was induced in male Wistar rats by a single injection of the VEGF receptor antagonist SU5416 (20 mg/kg) followed by exposure to hypoxia (10%O2) for 21 days. Two weeks after SU5416 administration, vehicle, riociguat (3 mg/kg/day), the TAK-1 inhibitor 5Z-7-oxozeaenol (OXO, 3 mg/kg/day), or both drugs combined were administered for 7 days. Metabolic profiling of right ventricle (RV), lung tissues and PA smooth muscle cells (PASMCs) extracts were performed by magnetic resonance spectroscopy, and the differences between groups analyzed by multivariate statistical methods.Results:In vitro, riociguat induced potent vasodilator effects in isolated pulmonary arteries (PA) with negligible antiproliferative effects and metabolic changes in PASMCs. In contrast, 5Z-7-oxozeaenol effectively inhibited the proliferation of PASMCs characterized by a broad metabolic reprogramming but had no acute vasodilator effects. In vivo, treatment with riociguat partially reduced the increase in pulmonary arterial pressure (PAP), RV hypertrophy (RVH), and pulmonary vascular remodeling, attenuated the dysregulation of inosine, glucose, creatine and phosphocholine (PC) in RV and fully abolished the increase in lung IL-1β expression. By contrast, 5Z-7-oxozeaenol significantly reduced pulmonary vascular remodeling and attenuated the metabolic shifts of glucose and PC in RV but had no effects on PAP or RVH. Importantly, combined therapy had an additive effect on pulmonary vascular remodeling and induced a significant metabolic effect over taurine, amino acids, glycolysis, and TCA cycle metabolism via glycine-serine-threonine metabolism. However, it did not improve the effects induced by riociguat alone on pulmonary pressure or RV remodeling. None of the treatments attenuated pulmonary endothelial dysfunction and hyperresponsiveness to serotonin in isolated PA.Conclusion: Our results suggest that inhibition of TAK-1 induces antiproliferative effects and its addition to short-term vasodilator therapy enhances the beneficial effects on pulmonary vascular remodeling and RV metabolic reprogramming in experimental PAH.
Collapse
Affiliation(s)
- Daniel Morales-Cano
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
- Ciber Enfermedades Respiratorias (Ciberes), Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón (IISGM), Madrid, Spain
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Jose Luis Izquierdo-García
- Ciber Enfermedades Respiratorias (Ciberes), Madrid, Spain
- Department of Chemistry in Pharmaceutical Sciences, School of Pharmacy, Universidad Complutense de Madrid, Madrid, Spain
- Instituto Pluridisciplinar, Universidad Complutense de Madrid, Madrid, Spain
| | - Bianca Barreira
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
- Ciber Enfermedades Respiratorias (Ciberes), Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón (IISGM), Madrid, Spain
| | - Sergio Esquivel-Ruiz
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
- Ciber Enfermedades Respiratorias (Ciberes), Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón (IISGM), Madrid, Spain
| | - Maria Callejo
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
- Ciber Enfermedades Respiratorias (Ciberes), Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón (IISGM), Madrid, Spain
| | - Rachele Pandolfi
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
- Ciber Enfermedades Respiratorias (Ciberes), Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón (IISGM), Madrid, Spain
| | - Palmira Villa-Valverde
- Ciber Enfermedades Respiratorias (Ciberes), Madrid, Spain
- ICTS Bioimagen Complutense, Universidad Complutense de Madrid, Madrid, Spain
| | - Ignacio Rodríguez
- Ciber Enfermedades Respiratorias (Ciberes), Madrid, Spain
- Department of Chemistry in Pharmaceutical Sciences, School of Pharmacy, Universidad Complutense de Madrid, Madrid, Spain
| | - Angel Cogolludo
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
- Ciber Enfermedades Respiratorias (Ciberes), Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón (IISGM), Madrid, Spain
| | - Jesus Ruiz-Cabello
- Ciber Enfermedades Respiratorias (Ciberes), Madrid, Spain
- Department of Chemistry in Pharmaceutical Sciences, School of Pharmacy, Universidad Complutense de Madrid, Madrid, Spain
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Donostia San Sebastián, Spain
| | - Francisco Perez-Vizcaino
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
- Ciber Enfermedades Respiratorias (Ciberes), Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón (IISGM), Madrid, Spain
| | - Laura Moreno
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
- Ciber Enfermedades Respiratorias (Ciberes), Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón (IISGM), Madrid, Spain
- *Correspondence: Laura Moreno,
| |
Collapse
|
11
|
Lai C, Monnet X, Teboul JL. Hemodynamic Implications of Prone Positioning in Patients with ARDS. Crit Care 2023; 27:98. [PMID: 36941694 PMCID: PMC10027593 DOI: 10.1186/s13054-023-04369-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2023] Open
Abstract
This article is one of ten reviews selected from the Annual Update in Intensive Care and Emergency Medicine 2023. Other selected articles can be found online at https://www.biomedcentral.com/collections/annualupdate2023 . Further information about the Annual Update in Intensive Care and Emergency Medicine is available from https://link.springer.com/bookseries/8901 .
Collapse
Affiliation(s)
- Christopher Lai
- AP-HP, Service de Médecine Intensive-Réanimation, Hôpital de Bicêtre, DMU CORREVE, Inserm UMR S_999, FHU SEPSIS, Groupe de Recherche Clinique CARMAS, Université Paris-Saclay, Le Kremlin-Bicêtre, France.
| | - Xavier Monnet
- AP-HP, Service de Médecine Intensive-Réanimation, Hôpital de Bicêtre, DMU CORREVE, Inserm UMR S_999, FHU SEPSIS, Groupe de Recherche Clinique CARMAS, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Jean-Louis Teboul
- AP-HP, Service de Médecine Intensive-Réanimation, Hôpital de Bicêtre, DMU CORREVE, Inserm UMR S_999, FHU SEPSIS, Groupe de Recherche Clinique CARMAS, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| |
Collapse
|
12
|
Bernardelli C, Caretti A, Lesma E. Dysregulated lipid metabolism in lymphangioleiomyomatosis pathogenesis as a paradigm of chronic lung diseases. Front Med (Lausanne) 2023; 10:1124008. [PMID: 36744130 PMCID: PMC9894443 DOI: 10.3389/fmed.2023.1124008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 01/06/2023] [Indexed: 01/20/2023] Open
Abstract
A chronic inflammatory condition characterizes various lung diseases. Interestingly, a great contribution to inflammation is made by altered lipids metabolism, that can be caused by the deregulation of the mammalian target of rapamycin complex-1 (mTORC1) activity. There is evidence that one of mTOR downstream effectors, the sterol regulatory element-binding protein (SREBP), regulates the transcription of enzymes involved in the de novo fatty acid synthesis. Given its central role in cell metabolism, mTOR is involved in several biological processes. Among those, mTOR is a driver of senescence, a process that might contribute to the establishment of chronic lung disease because the characteristic irreversible inhibition of cell proliferation, associated to the acquisition of a pro-inflammatory senescence-associated secretory phenotype (SASP) supports the loss of lung parenchyma. The deregulation of mTORC1 is a hallmark of lymphangioleiomyomatosis (LAM), a rare pulmonary disease predominantly affecting women which causes cystic remodeling of the lung and progressive loss of lung function. LAM cells have senescent features and secrete SASP components, such as growth factors and pro-inflammatory molecules, like cancer cells. Using LAM as a paradigm of chronic and metastatic lung disease, here we review the published data that point out the role of dysregulated lipid metabolism in LAM pathogenesis. We will discuss lipids' role in the development and progression of the disease, to hypothesize novel LAM biomarkers and to propose the pharmacological regulation of lipids metabolism as an innovative approach for the treatment of the disease.
Collapse
Affiliation(s)
- Clara Bernardelli
- Laboratory of Pharmacology, Department of Health Sciences, Università degli Studi di Milano, Milan, Italy
| | - Anna Caretti
- Laboratory of Biochemistry and Molecular Biology, Department of Health Sciences, Università degli Studi di Milano, Milan, Italy
| | - Elena Lesma
- Laboratory of Pharmacology, Department of Health Sciences, Università degli Studi di Milano, Milan, Italy,*Correspondence: Elena Lesma,
| |
Collapse
|
13
|
Soliman YMA, Elkorashy RIM, Aziz AA, Abdelnaby A, Magdy S. Impact and predictors of outcome of COVID-19 in pulmonary hypertension patients. THE EGYPTIAN JOURNAL OF BRONCHOLOGY 2022. [DOI: 10.1186/s43168-022-00158-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Abstract
Background
The pandemic had a significant impact on those with underlying chronic health conditions being at risk of developing a more severe disease with rapid progression, significant complications, and with increased risk of mortality.
This was also expected in the pulmonary vascular community owing to the vulnerable nature of this population, who are characterized by an increase in the pulmonary vascular resistance leading to right heart failure.
This study is aiming to identify the incidence of COVID-19 infection among pulmonary hypertension patients receiving specific therapy as well as the predictors of the COVID-19 disease severity and outcome in those patients.
Results
Data analysis of 197 PAH and CTEPH patients, showed that the incidence of SARS-CoV-2 infection is 10.66% (n = 21). Seven patients (33.3%) required hospitalization. Mortality rate is 14.3% (3/21).
Severity of COVID19 disease in those patients has statistically significant moderate to strong correlation with higher values of d-dimer (r = 0.821, P = 0.000), ferritin (r = 0.718, p = 0.000), CRP (r = 0.613, p = 0.04), acute renal failure (r = 0.557, p = 0.009), and hypoxemia (r = 0.825, p = 0.000).
Mortality from COVID-19 show moderate to strong statistically significant correlations with acute renal failure (r = 0.795, p = 0.000), hypoxemia (r = 0.645, p = 0.002), higher values of ferritin (r = 0.689, p = 0.001) and d-dimer (r = 0.603, P = 0.004).
Conclusions
COVID-19 in PAH and CTEPH patients is challenging, higher COVID-19 infection rate is present in those patients and is associated with increased disease severity and higher mortality.
Collapse
|
14
|
Hou H, Guo C, Que C, Nie L, Zhang Q, Zhao H, Nong L, Ma W, Wang Q, Liang Z, Wang B, Ma J, Wang G. Diffuse large B-cell lymphoma presenting as reversible intrapulmonary arteriovenous shunts with hypoxia, fever and progressive jaundice: a case report and literature review. BMC Pulm Med 2022; 22:89. [PMID: 35292006 PMCID: PMC8922084 DOI: 10.1186/s12890-022-01881-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 03/07/2022] [Indexed: 11/16/2022] Open
Abstract
Background Intrapulmonary arteriovenous shunts is rare seen in a patient without lung involvement. Case presentation This is the first report of reversible intrapulmonary arteriovenous shunts secondary to extrapulmonary lymphoma as one initial symptom. The patient presented as fever of unknown origin and dyspnea, and examinations of infection were negative. Diagnosis of DLBCL was finally confirmed through bone marrow and splenic biopsies. Intrapulmonary arteriovenous shunts were diagnosed through 100% oxygen inhalation test and transthoracic contrast echocardiography (TTCE). After the treatment of lymphoma, his respiratory failure was relieved. We rechecked the 100% oxygen inhalation test and TTCE, which both indicated that his intrapulmonary arteriovenous shunts had resolved. Conclusions We speculated the prominent inflammation from active DLBCL was the most possible mechanism associated with the reversible intrapulmonary shunt in this patient. These findings will assist us to better understand the mechanism of intrapulmonary shunts.
Collapse
Affiliation(s)
- Huan Hou
- Department of Respiratory and Critical Care Medicine, Peking University First Hospital, 8 Xishiku Street, Xicheng District, Beijing, 100034, China
| | - Cuiyan Guo
- Department of Respiratory and Critical Care Medicine, Peking University First Hospital, 8 Xishiku Street, Xicheng District, Beijing, 100034, China
| | - Chengli Que
- Department of Respiratory and Critical Care Medicine, Peking University First Hospital, 8 Xishiku Street, Xicheng District, Beijing, 100034, China
| | - Ligong Nie
- Department of Respiratory and Critical Care Medicine, Peking University First Hospital, 8 Xishiku Street, Xicheng District, Beijing, 100034, China
| | - Qi Zhang
- Department of Respiratory and Critical Care Medicine, Peking University First Hospital, 8 Xishiku Street, Xicheng District, Beijing, 100034, China
| | - Hong Zhao
- Department of Infectious Diseases, Center for Liver Disease, Peking University First Hospital, Beijing, China
| | - Lin Nong
- Department of Pathology, Peking University First Hospital, Beijing, China
| | - Wei Ma
- Department of Cardiology, Peking University First Hospital, Beijing, China
| | - Qian Wang
- Department of Hematology, Peking University First Hospital, Beijing, China
| | - Zeyin Liang
- Department of Hematology, Peking University First Hospital, Beijing, China
| | - Bingjie Wang
- Department of Hematology, Peking University First Hospital, Beijing, China
| | - Jing Ma
- Department of Respiratory and Critical Care Medicine, Peking University First Hospital, 8 Xishiku Street, Xicheng District, Beijing, 100034, China.
| | - Guangfa Wang
- Department of Respiratory and Critical Care Medicine, Peking University First Hospital, 8 Xishiku Street, Xicheng District, Beijing, 100034, China
| |
Collapse
|
15
|
Administration of an Acidic Sphingomyelinase (ASMase) Inhibitor, Imipramine, Reduces Hypoglycemia-Induced Hippocampal Neuronal Death. Cells 2022; 11:cells11040667. [PMID: 35203316 PMCID: PMC8869983 DOI: 10.3390/cells11040667] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/31/2022] [Accepted: 02/12/2022] [Indexed: 01/27/2023] Open
Abstract
Severe hypoglycemia (below 35 mg/dL) appears most often in diabetes patients who continuously inject insulin. To rapidly cease the hypoglycemic state in this study, glucose reperfusion was conducted, which can induce a secondary neuronal death cascade following hypoglycemia. Acid sphingomyelinase (ASMase) hydrolyzes sphingomyelin into ceramide and phosphorylcholine. ASMase activity can be influenced by cations, pH, redox, lipids, and other proteins in the cells, and there are many changes in these factors in hypoglycemia. Thus, we expect that ASMase is activated excessively after hypoglycemia. Ceramide is known to cause free radical production, excessive inflammation, calcium dysregulation, and lysosomal injury, resulting in apoptosis and the necrosis of neurons. Imipramine is mainly used in the treatment of depression and certain anxiety disorders, and it is particularly known as an ASMase inhibitor. We hypothesized that imipramine could decrease hippocampal neuronal death by reducing ceramide via the inhibition of ASMase after hypoglycemia. In the present study, we confirmed that the administration of imipramine significantly reduced hypoglycemia-induced neuronal death and improved cognitive function. Therefore, we suggest that imipramine may be a promising therapeutic tool for preventing hypoglycemia-induced neuronal death.
Collapse
|
16
|
Zeng C, Lagier D, Lee JW, Melo MFV. Perioperative Pulmonary Atelectasis: Part I. Biology and Mechanisms. Anesthesiology 2022; 136:181-205. [PMID: 34499087 PMCID: PMC9869183 DOI: 10.1097/aln.0000000000003943] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Pulmonary atelectasis is common in the perioperative period. Physiologically, it is produced when collapsing forces derived from positive pleural pressure and surface tension overcome expanding forces from alveolar pressure and parenchymal tethering. Atelectasis impairs blood oxygenation and reduces lung compliance. It is increasingly recognized that it can also induce local tissue biologic responses, such as inflammation, local immune dysfunction, and damage of the alveolar-capillary barrier, with potential loss of lung fluid clearance, increased lung protein permeability, and susceptibility to infection, factors that can initiate or exaggerate lung injury. Mechanical ventilation of a heterogeneously aerated lung (e.g., in the presence of atelectatic lung tissue) involves biomechanical processes that may precipitate further lung damage: concentration of mechanical forces, propagation of gas-liquid interfaces, and remote overdistension. Knowledge of such pathophysiologic mechanisms of atelectasis and their consequences in the healthy and diseased lung should guide optimal clinical management.
Collapse
Affiliation(s)
- Congli Zeng
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - David Lagier
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jae-Woo Lee
- Department of Anesthesia, University of California San Francisco, San Francisco, CA, USA
| | - Marcos F. Vidal Melo
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
17
|
Dei Cas M, Ottolenghi S, Morano C, Rinaldo R, Roda G, Chiumello D, Centanni S, Samaja M, Paroni R. Link between serum lipid signature and prognostic factors in COVID-19 patients. Sci Rep 2021; 11:21633. [PMID: 34737330 PMCID: PMC8568966 DOI: 10.1038/s41598-021-00755-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 10/14/2021] [Indexed: 01/08/2023] Open
Abstract
Although the serum lipidome is markedly affected by COVID-19, two unresolved issues remain: how the severity of the disease affects the level and the composition of serum lipids and whether serum lipidome analysis may identify specific lipids impairment linked to the patients' outcome. Sera from 49 COVID-19 patients were analyzed by untargeted lipidomics. Patients were clustered according to: inflammation (C-reactive protein), hypoxia (Horowitz Index), coagulation state (D-dimer), kidney function (creatinine) and age. COVID-19 patients exhibited remarkable and distinctive dyslipidemia for each prognostic factor associated with reduced defense against oxidative stress. When patients were clustered by outcome (7 days), a peculiar lipidome signature was detected with an overall increase of 29 lipid species, including-among others-four ceramide and three sulfatide species, univocally related to this analysis. Considering the lipids that were affected by all the prognostic factors, we found one sphingomyelin related to inflammation and viral infection of the respiratory tract and two sphingomyelins, that are independently related to patients' age, and they appear as candidate biomarkers to monitor disease progression and severity. Although preliminary and needing validation, this report pioneers the translation of lipidome signatures to link the effects of five critical clinical prognostic factors with the patients' outcomes.
Collapse
Grants
- This research was funded by Dipartimento di Scienze della Salute, Università degli Studi di Milano (Piano di Sostegno alla Ricerca LINEA 2: Dotazione annuale per attività istituzionali within a project entitled “FeOx. Iron handling in patients exposed to acute and chronic hypoxia", by Ministero dell'Istruzione, dell'Università e della Ricerca (Programma Nazionale di Ricerca in Antartide, PNRA18_00071-F within a project entitled “Concorde. Impact of the Antarctic environments on human homeostasis, psychology, physiology and immunity”), by Ministero dell'Istruzione, dell'Università e della Ricerca (FISR-COVID-19 Project FISR2020IP_01583, within a project entitled “HITCoA. Impact of Hypoxia, Iron Toxicity and oxidative stress on COvid19 Anemia”).
Collapse
Affiliation(s)
- Michele Dei Cas
- Department of Health Sciences, Università degli Studi di Milano, via A. di Rudinì 8, Milan, Italy
| | - Sara Ottolenghi
- Department of Health Sciences, Università degli Studi di Milano, via A. di Rudinì 8, Milan, Italy
| | - Camillo Morano
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, Milan, Italy
| | - Rocco Rinaldo
- Department of Health Sciences, Università degli Studi di Milano, via A. di Rudinì 8, Milan, Italy
- Respiratory Unit, San Paolo University Hospital, ASST Santi Paolo e Carlo, Milan, Italy
| | - Gabriella Roda
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, Milan, Italy
| | - Davide Chiumello
- Department of Health Sciences, Università degli Studi di Milano, via A. di Rudinì 8, Milan, Italy
- Department of Anesthesia and Intensive Care, San Paolo University Hospital, ASST Santi Paolo e Carlo, Milan, Italy
| | - Stefano Centanni
- Department of Health Sciences, Università degli Studi di Milano, via A. di Rudinì 8, Milan, Italy
- Respiratory Unit, San Paolo University Hospital, ASST Santi Paolo e Carlo, Milan, Italy
| | - Michele Samaja
- Department of Health Sciences, Università degli Studi di Milano, via A. di Rudinì 8, Milan, Italy
| | - Rita Paroni
- Department of Health Sciences, Università degli Studi di Milano, via A. di Rudinì 8, Milan, Italy.
| |
Collapse
|
18
|
Gierhardt M, Pak O, Walmrath D, Seeger W, Grimminger F, Ghofrani HA, Weissmann N, Hecker M, Sommer N. Impairment of hypoxic pulmonary vasoconstriction in acute respiratory distress syndrome. Eur Respir Rev 2021; 30:30/161/210059. [PMID: 34526314 DOI: 10.1183/16000617.0059-2021] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 06/05/2021] [Indexed: 12/29/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a serious complication of severe systemic or local pulmonary inflammation, such as caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. ARDS is characterised by diffuse alveolar damage that leads to protein-rich pulmonary oedema, local alveolar hypoventilation and atelectasis. Inadequate perfusion of these areas is the main cause of hypoxaemia in ARDS. High perfusion in relation to ventilation (V/Q<1) and shunting (V/Q=0) is not only caused by impaired hypoxic pulmonary vasoconstriction but also redistribution of perfusion from obstructed lung vessels. Rebalancing the pulmonary vascular tone is a therapeutic challenge. Previous clinical trials on inhaled vasodilators (nitric oxide and prostacyclin) to enhance perfusion to high V/Q areas showed beneficial effects on hypoxaemia but not on mortality. However, specific patient populations with pulmonary hypertension may profit from treatment with inhaled vasodilators. Novel treatment targets to decrease perfusion in low V/Q areas include epoxyeicosatrienoic acids and specific leukotriene receptors. Still, lung protective ventilation and prone positioning are the best available standard of care. This review focuses on disturbed perfusion in ARDS and aims to provide basic scientists and clinicians with an overview of the vascular alterations and mechanisms of V/Q mismatch, current therapeutic strategies, and experimental approaches.
Collapse
Affiliation(s)
- Mareike Gierhardt
- Dept of Internal Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig University, Giessen, Germany.,Excellence Cluster Cardio-Pulmonary Institute (CPI), Giessen, Germany.,Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA) - CONICET - Partner Institute of the Max Planck Society, Buenos Aires, Argentina.,Department of Lung Development and Remodeling, Max Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Member of the Cardio-Pulmonary Institute (CPI) Bad Nauheim, Germany
| | - Oleg Pak
- Dept of Internal Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig University, Giessen, Germany.,Excellence Cluster Cardio-Pulmonary Institute (CPI), Giessen, Germany
| | - Dieter Walmrath
- Dept of Internal Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig University, Giessen, Germany
| | - Werner Seeger
- Dept of Internal Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig University, Giessen, Germany.,Excellence Cluster Cardio-Pulmonary Institute (CPI), Giessen, Germany.,Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA) - CONICET - Partner Institute of the Max Planck Society, Buenos Aires, Argentina.,Institute for Lung Health (ILH), Giessen, Germany
| | - Friedrich Grimminger
- Dept of Internal Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig University, Giessen, Germany.,Excellence Cluster Cardio-Pulmonary Institute (CPI), Giessen, Germany
| | - Hossein A Ghofrani
- Dept of Internal Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig University, Giessen, Germany.,Excellence Cluster Cardio-Pulmonary Institute (CPI), Giessen, Germany.,Dept of Medicine, Imperial College London, London, UK
| | - Norbert Weissmann
- Dept of Internal Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig University, Giessen, Germany.,Excellence Cluster Cardio-Pulmonary Institute (CPI), Giessen, Germany
| | - Matthias Hecker
- Dept of Internal Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig University, Giessen, Germany.,Both authors contributed equally
| | - Natascha Sommer
- Dept of Internal Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig University, Giessen, Germany.,Excellence Cluster Cardio-Pulmonary Institute (CPI), Giessen, Germany.,Both authors contributed equally
| |
Collapse
|
19
|
Shen SY, Ren LQ, Chen HD, Zhu HF, Zhou DF, Zhang B, Tan XQ, Xie YH. Geniposide protects pulmonary arterial smooth muscle cells from lipopolysaccharide-induced injury via α7nAchR-mediated TLR-4/MyD88 signaling. Exp Ther Med 2021; 22:1234. [PMID: 34539830 PMCID: PMC8438699 DOI: 10.3892/etm.2021.10668] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Accepted: 02/10/2021] [Indexed: 12/18/2022] Open
Abstract
Geniposide is a bioactive iridoid glucoside derived from Gardenia jasminoides that has proven anti-inflammatory effects against acute lung injury. The aim of this study was to determine whether geniposide could protect pulmonary arterial smooth muscle cells (PASMCs) from lipopolysaccharide (LPS)-induced injury and to explore the participation of α7 nicotinic acetylcholine receptor (α7nAChR), which was previously reported to suppress pro-inflammatory cytokine production in LPS-stimulated macrophages. In the present study, rat PASMCs were isolated and stimulated using LPS. The effect of geniposide on LPS-induced PASMC injury was then explored. Geniposide exerted anti-apoptotic and anti-inflammatory effects on LPS-treated PASMCs, as demonstrated by the downregulation of pro-apoptotic proteins and pro-inflammatory cytokines, respectively. Furthermore, the α7nAChR agonist PNU282987 accentuated the protective effect of geniposide against LPS-induced injury in PASMCs by inhibiting toll-like receptor-4/myeloid differentiation primary response 88 (TLR-4/MyD88) signaling and downregulating nuclear factor (NF)-κB expression. Conversely, methyllycaconitine, an inhibitor of α7nAChR, attenuated the effects of geniposide. These findings collectively suggested that in conjunction with geniposide, the activation of α7nAChR may contribute to further mitigating LPS-induced PASMC apoptosis and inflammation. In addition, the underlying mechanisms critically involve the NF-κB/MyD88 signaling axis. These results may provide novel insights into the treatment and management of lung diseases via geniposide administration.
Collapse
Affiliation(s)
- San-Ying Shen
- Department of Respiratory Medicine, Wuhan Fourth Hospital, Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430033, P.R. China
| | - Li-Quan Ren
- Department of Medical Services, Wuhan Fourth Hospital, Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430033, P.R. China
| | - Hui-Dong Chen
- Department of Respiratory Medicine, Wuhan Jinyintan Hospital, Wuhan, Hubei 430023, P.R. China
| | - Hong-Fei Zhu
- Hubei Research Institute of Traditional Chinese Medicine, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, Hubei 430072, P.R. China
| | - Deng-Feng Zhou
- Department of Respiratory Medicine, Wuhan Fourth Hospital, Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430033, P.R. China
| | - Bo Zhang
- Department of Respiratory Medicine, Wuhan Fourth Hospital, Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430033, P.R. China
| | - Xiao-Qin Tan
- Department of Respiratory Medicine, Wuhan Fourth Hospital, Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430033, P.R. China
| | - Yong-Hua Xie
- Department of Traditional Chinese Medicine, Wuhan Fourth Hospital, Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430033, P.R. China
| |
Collapse
|
20
|
Effect of Dihydropyridine Calcium Channel Blocker on Mortality of Hypertension Patients With Moderate-Severe Pulmonary Acute Respiratory Distress Syndrome: A Multicenter Retrospective Observational Cohort Study. Crit Care Explor 2021; 3:e0506. [PMID: 34514419 PMCID: PMC8425825 DOI: 10.1097/cce.0000000000000506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
The aim was to evaluate the effect of dihydropyridine calcium channel blocker on the prognosis for moderate-severe pulmonary acute respiratory distress syndrome in hypertension patients. DESIGN A retrospective, observational, multicenter cohort study. SETTING A total of 307 patients without propensity score matching and 186 adult inpatients with propensity score matching diagnosed with hypertension and moderate-severe pulmonary acute respiratory distress syndrome in five teaching hospitals in Jiangsu province, China, from December 2015 to December 2020 were enrolled. PATIENTS A total of 307 patients without propensity score matching and 186 patients with propensity score matching diagnosed with hypertension and moderate-severe pulmonary acute respiratory distress syndrome were included in the final analysis. INTERVENTIONS None. MEASUREMENTS AND MAIN RESULTS Demographic characteristics and clinical characteristics were recorded. The propensity score matching method was used to eliminate the difference between group with dihydropyridine calcium channel blocker and group without dihydropyridine calcium channel blocker. The primary outcome was in-hospital mortality. We used univariate and multivariate regression analyses for both patients with or without propensity score matching to assess the effect of these variables on mortality. In the subset of 186 patients with propensity score matching, in-hospital mortality was 53.2%. Inpatient mortality was significantly higher in patients treated with dihydropyridine calcium channel blocker than in those not treated with dihydropyridine calcium channel blocker of patients without propensity score matching (65.4% vs 40.4%; p < 0.01). Multivariate analysis for patients without propensity score matching showed that dihydropyridine calcium channel blocker (hazard ratio, 1.954; 95% CI, 1.415-2.699), lactate dehydrogenase greater than or equal to 600 U/L (hazard ratio, 3.809; 95% CI, 2.106-4.531), and lactate greater than or equal to 2 mmol/L (hazard ratio, 1.454; 95% CI, 1.041-2.029) were independently associated with in-hospital mortality. Based on univariate analysis for patients with propensity score matching, dihydropyridine calcium channel blocker (hazard ratio, 2.021; 95% CI, 1.333-3.064), lactate dehydrogenase greater than or equal to 600 U/L (hazard ratio, 4.379; 95% CI, 2.642-7.257), and lactate greater than or equal to 2 mmol/L (hazard ratio, 2.461; 95% CI, 1.534-3.951) were independently associated with in-hospital mortality. In contrast, patients not treated with dihydropyridine calcium channel blocker had a significant survival advantage over those treated with dihydropyridine calcium channel blocker in both patients without or with propensity score matching (p < 0.001; p = 0.001 by Kaplan-Meier analysis). CONCLUSIONS Dihydropyridine calcium channel blocker, lactate dehydrogenase greater than or equal to 600 U/L, and lactate greater than or equal to 2 mmol/L at admission were independent risk factors for patients with hypertension and moderate-severe pulmonary acute respiratory distress syndrome.
Collapse
|
21
|
Perez-Vizcaino F, Moreno L, Lorente JA. Interleukin-6 and intrapulmonary shunt. Eur Respir J 2021; 58:13993003.01292-2021. [PMID: 34210792 PMCID: PMC8358234 DOI: 10.1183/13993003.01292-2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 06/06/2021] [Indexed: 11/13/2022]
Abstract
We read with interest the article by Kotwicaet al. [1] showing the utility of clinical pulse oximetry measurements to quantify shunt and ventilation–perfusion mismatch and their predictive value in severe coronavirus disease 2019 (COVID-19). The authors found that shunt correlated with markers of activated inflammatory response (i.e. C-reactive protein) but not those of activated coagulation (such as D-dimer). Their results reinforce the growing evidence for the role of impaired hypoxic pulmonary vasoconstriction (HPV) as a primary cause for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-induced hypoxaemia [2]. Based on animal studies and indirect clinical evidence, it may be speculated that IL-6 has a pathophysiological role in intrapulmonary shunt associated to COVID-19https://bit.ly/3whQVqd
Collapse
Affiliation(s)
- Francisco Perez-Vizcaino
- Dept of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain.,CIBER de Enfermedades Respiratorias (CIBERES), Madrid, Spain.,Instituto de Investigación Sanitaria Gregorio Marañón (IISGM), Madrid, Spain
| | - Laura Moreno
- Dept of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain.,CIBER de Enfermedades Respiratorias (CIBERES), Madrid, Spain.,Instituto de Investigación Sanitaria Gregorio Marañón (IISGM), Madrid, Spain
| | - José A Lorente
- CIBER de Enfermedades Respiratorias (CIBERES), Madrid, Spain.,Critical Care Service, Hospital Universitario de Getafe, Madrid, Spain.,Universidad Europea, Madrid, Spain
| |
Collapse
|
22
|
Huang W, Liu H, Pan Y, Yang H, Lin J, Zhang H. Mechanical stretching of the pulmonary vein mediates pulmonary hypertension due to left heart disease by regulating SAC/MAPK pathway and the expression of IL-6 and TNF-α. J Cardiothorac Surg 2021; 16:127. [PMID: 33971931 PMCID: PMC8107413 DOI: 10.1186/s13019-021-01471-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Accepted: 04/05/2021] [Indexed: 01/09/2023] Open
Abstract
Background This study aimed to explore whether the mechanical stretching-induced expression of interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α) in pulmonary veins occurred through the stretch-activated channel (SAC)/ mitogen-activated protein kinases (MAPKs) pathway. Methods Sixty male Sprague-Dawley rats were divided into three sham groups and seven model groups. A metal clip was placed on the ascending aorta in the model group to establish PH-LHD rat model. The sham group received a similar operation without ascending aorta clamped. On day 25, pulmonary vein was given mechanical stretching with 0 g, 2.0 g tension in two model groups and two sham groups. Another four model groups were given 2.0 g tension after MAPKs pathway inhibitors soaked. The last sham group and model group rats’ pulmonary veins, pulmonary artery and lung tissues were obtained on day 35. Pulmonary vein, pulmonary artery and lung tissue were evaluated by echocardiography, HE staining, immunohistochemistry and western blotting respectively. Results On day 25, left heart weight, right ventricular pressure (35.339 cmH2O) and left atrial pressure (13.657 cmH2O) were increased in model group than those in sham group. Echocardiography showed left heart failure in the PH-LHD group (Interventrieular septum dimension 1.716 mm, left ventricular internal end diastolic dimension 4.888 mm, left ventricular posterior wall thickness in diastole 1.749 mm, ejection fraction 76.917%). But there was no difference in lung tissue between the sham group and PH-LHD group as showed by HE staining. Our results showed that the expression of IL-6 and TNF-α was highly expressed in PH-LHD rats’ serum and pulmonary vein, which were further increased after 2.0 g tension was given and were decreased after SAC/MAPKs inhibitors treatment. Meanwhile, on day 25, immunohistochemistry analysis showed the expression of IL-6 and TNF-α was higher in the PH-LHD rats’ pulmonary vein than that in pulmonary artery and lung tissue, and these expressions in pulmonary vein of PH-LHD group were also higher than that in sham group. However, on day 35, IL-6 and TNF-α were all increased in the pulmonary veins, arteries and lung tissues. Besides, our results uncovered that SAC/MAPKs pathway were upregulating in PH-LHD rats’ pulmonary vein. Conclusion In conclusion, pulmonary vein mechanical stretching exacerbated PH-LHD possibly through the SAC/MAPKs pathway and upregulating expression of IL-6 and TNF-α.
Collapse
Affiliation(s)
- Wenhui Huang
- Department of Cardiovascular Surgery, Union Hospital, Fujian Medical University, Fuzhou, 350001, Fujian Province, People's Republic of China.,Anesthesiology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350004, Fujian Province, People's Republic of China
| | - Hongjin Liu
- Department of Cardiovascular Surgery, Union Hospital, Fujian Medical University, Fuzhou, 350001, Fujian Province, People's Republic of China
| | - Yichao Pan
- Department of Cardiovascular Surgery, Union Hospital, Fujian Medical University, Fuzhou, 350001, Fujian Province, People's Republic of China
| | - Hongwei Yang
- Department of Cardiovascular Surgery, Union Hospital, Fujian Medical University, Fuzhou, 350001, Fujian Province, People's Republic of China
| | - Jing Lin
- Department of Cardiovascular Surgery, Union Hospital, Fujian Medical University, Fuzhou, 350001, Fujian Province, People's Republic of China
| | - Hui Zhang
- Department of Intensive Care Unit, Union Hospital, Fujian Medical University, Fuzhou, 350004, Fujian Province, People's Republic of China.
| |
Collapse
|
23
|
Zhang L, Jiang Y, Deng S, Mo Y, Huang Y, Li W, Ge C, Ren X, Zhang H, Zhang X, Peng Q, Liu Z, Huang L, Zhou F, Ai Y. S100B/RAGE/Ceramide signaling pathway is involved in sepsis-associated encephalopathy. Life Sci 2021; 277:119490. [PMID: 33862114 DOI: 10.1016/j.lfs.2021.119490] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 03/26/2021] [Accepted: 03/30/2021] [Indexed: 01/22/2023]
Abstract
AIMS Sepsis-associated encephalopathy (SAE) is one of the most common complications of sepsis, and it might lead to long-term cognitive dysfunction and disability. This study aimed to explore the role of S100 calcium binding protein B (S100B)/RAGE/ceramide signaling pathway in SAE. MAIN METHODS FPS-ZM1 (an inhibitor of RAGE), myriocin and GW4869 (an inhibitor of ceramide) were used to explore the role of S100B/RAGE/ceramide in acute brain injury and long-term cognitive impairment in sepsis. In addition, Mdivi-1 (inhibitor of Drp1) and Drp1 siRNA were utilized to assess the effects of C2-ceramide on neuronal mitochondria, and to explore the specific underlying mechanism in C2 ceramide-induced death of HT22 mouse hippocampal neuronal cells. KEY FINDINGS Western blot analysis showed that sepsis significantly up-regulated S100B and RAGE. Nissl staining and Morris water maze (MWM) test revealed that inhibition of RAGE with FPS-ZM1 markedly attenuated cecal ligation and puncture (CLP)-induced brain damage and cognitive dysfunction. Furthermore, FPS-ZM1 relieved sepsis-induced C2-ceramide accumulation and abnormal mitochondrial dynamics. Moreover, inhibition of ceramide also showed similar protective effects both in vivo and in vitro. Furthermore, Mdivi-1 and Drp1 siRNA significantly reduced C2-ceramide-induced neuronal mitochondrial fragmentation and cell apoptosis in vitro. SIGNIFICANCE This study confirmed that S100B regulates mitochondrial dynamics through RAGE/ceramide pathway, in addition to the role of this pathway in acute brain injury and long-term cognitive impairment during sepsis.
Collapse
Affiliation(s)
- Lina Zhang
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China.
| | - Yuan Jiang
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Songyun Deng
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, China.
| | - Yunan Mo
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, China.
| | - Yan Huang
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, China.
| | - Wenchao Li
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, China.
| | - Chenglong Ge
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, China.
| | - Xinshu Ren
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, China.
| | - Haisong Zhang
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, China.
| | - Xiaolei Zhang
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, China.
| | - Qianyi Peng
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, China.
| | - Zhiyong Liu
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, China.
| | - Li Huang
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, China.
| | - Fan Zhou
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha 410011, China.
| | - Yuhang Ai
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China.
| |
Collapse
|
24
|
Ghrelin Protects Lipopolysaccharide-Induced Acute Lung Injury Rats against Pulmonary Vascular Dysfunction by Inhibiting Inflammation. Can Respir J 2021. [DOI: 10.1155/2021/6643398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Objective. To determine the effect and mechanism of the anti-inflammatory agent ghrelin on pulmonary vascular dysfunction (PVD) in lipopolysaccharide- (LPS-) induced acute lung injury (ALI) rat models. Methods. Thirty-two adult male Sprague Dawley rats (n = 16/group) were randomly divided into ghrelin and saline groups, wherein ghrelin (10 nmol/kg) or saline was subcutaneously administered. After 30 min, eight rats from each group were randomly selected, and LPS (5 mg/kg) or saline was administered by intratracheal instillation to induce ALI. Four hours after establishing the ALI rat model, the mean pulmonary arterial pressure (mPAP), mean right ventricular systolic pressure (RVSP), levels of proinflammatory cytokines tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) in the bronchoalveolar lavage fluid (BALF), BALF cell count, wet-to-dry (W/D) lung weight ratios, and myeloperoxidase (MPO) activity in lung tissue for all four groups (ghrelin, ghrelin + ALI, saline, and saline + ALI) were measured. Immunohistochemical staining to detect alpha-smooth muscle actin (α-SMA) and proliferating cell nuclear antigen (PCNA) expression was performed to assess the intrapulmonary arterial wall thickness and the proliferation of smooth muscle cells, respectively. Results. The ghrelin-pretreated ALI rats showed lower mPAP, RVSP, PCNA expression, MPO activity, W/D lung weight ratio, TNF-α and IL-6 levels, and BALF cell count than the saline-pretreated ALI rats, but ghrelin had no effect on the intrapulmonary arterial wall thickness of ALI rats. Conclusion. Our results confirmed the association between inflammation and PVD in ALI and suggested that the suppression of inflammation by ghrelin pretreatment could protect LPS-induced ALI rats against PVD.
Collapse
|
25
|
Jiang J, Shi Y, Cao J, Lu Y, Sun G, Yang J. Role of ASM/Cer/TXNIP signaling module in the NLRP3 inflammasome activation. Lipids Health Dis 2021; 20:19. [PMID: 33612104 PMCID: PMC7897379 DOI: 10.1186/s12944-021-01446-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Accepted: 02/08/2021] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND This study aimed to explore the effects of ceramide (Cer) on NLRP3 inflammasome activation and their underlying mechanisms. METHODS Lipopolysaccharide (LPS)/adenosine triphosphate (ATP)-induced NLRP3 inflammasome activation in J774A.1 cells and THP-1 macrophages was used as an in vitro model of inflammation. Western blotting and real-time PCR (RT-PCR) were used to detect the protein and mRNA levels, respectively. IL-1β and IL-18 levels were measured by ELISA. ASM assay kit and immunofluorescence were used to detect ASM activity and Cer content. RESULTS Imipramine, a well-known inhibitor of ASM, significantly inhibited LPS/ATP-induced activity of ASM and the consequent accumulation of Cer. Additionally, imipramine suppressed the LPS/ATP-induced expression of thioredoxin interacting protein (TXNIP), NLRP3, caspase-1, IL-1β, and IL-18 at the protein and mRNA level. Interestingly verapamil, a TXNIP inhibitor, suppressed LPS/ATP-induced activation of TXNIP/NLRP3 inflammasome but did not affect LPS/ATP-induced ASM activation and Cer formation. TXNIP siRNA and verapamil inhibited C2-Cer-induced upregulation of TXNIP and activation of the NLRP3 inflammasome. In addition, the pretreatment of cells with sulfo-N-succinimidyl oleate (SSO), an irreversible inhibitor of the scavenger receptor CD36, blocked Cer-induced upregulation of nuclear factor-κB (NF-κB) activity, TXNIP expression, and NLRP3 inflammasome activation. Inhibition of NF-κB activation by SN50 prevented Cer-induced upregulation of TXNIP and activation of the NLRP3 inflammasome but did not affect CD36 expression. CONCLUSION This study demonstrated that the ASM/Cer/TXNIP signaling pathway is involved in NLRP3 inflammasome activation. The results documented that the CD36-dependent NF-κB-TXNIP signaling pathway plays an essential role in the Cer-induced activation of NLRP3 inflammasomes in macrophages.
Collapse
Affiliation(s)
- Jianjun Jiang
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, 230022, Anhui, China
| | - Yining Shi
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Hefei, 230601, Anhui, China
| | - Jiyu Cao
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Youjin Lu
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Hefei, 230601, Anhui, China
| | - Gengyun Sun
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, 230022, Anhui, China.
| | - Jin Yang
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Hefei, 230601, Anhui, China.
| |
Collapse
|
26
|
Guo C, Sun L, Zhang L, Dong F, Zhang X, Yao L, Chang C. Serum sphingolipid profile in asthma. J Leukoc Biol 2021; 110:53-59. [PMID: 33600023 DOI: 10.1002/jlb.3ma1120-719r] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 01/01/2021] [Accepted: 01/28/2021] [Indexed: 12/17/2022] Open
Abstract
Sphingolipids metabolism is an important cell process and plays critical roles in asthma. However, the involvement of sphingolipids in the pathogenesis of asthma and its subtypes is unknown. The present study aimed to determine the role of sphingolipids in asthma and its subtypes. Clinical data from 51 asthma patients and 9 healthy individuals were collected and serum samples were performed to analyze the levels of serum sphingolipids by liquid chromatography-mass spectrometry-based targeted metabolomics. Results showed that the levels of sphingomyelin (SM) including SM34:2, SM38:1, and SM40:1 were significantly decreased in asthmatic patients compared to healthy controls. Moreover, serum SM levels were obviously decreased in the blood noneosinophilic asthma (bNEA) group compared with blood eosinophilic asthma group. Similar tendencies of serum SM level changes were observed in the early-onset group compared with late-onset group. Correlation analysis revealed that SM 40:1 was negatively related to sputum IL-17A (r = -0.621, P = 0.042). The present study presented that the SM may be a protective factor of asthma and contributes to the mechanism of asthma, especially bNEA. SM may be a potential biomarker and therapeutic target in asthma.
Collapse
Affiliation(s)
- Chenglin Guo
- Department of Pulmonary and Critical Care Medicine, Peking University Third Hospital, Beijing, China
| | - Lina Sun
- Department of Pulmonary and Critical Care Medicine, Peking University Third Hospital, Beijing, China
| | - Linlin Zhang
- Department of Pulmonary and Critical Care Medicine, Peking University Third Hospital, Beijing, China
| | - Fawu Dong
- Department of Pulmonary and Critical Care Medicine, Peking University Third Hospital, Beijing, China
| | - Xu Zhang
- Tianjin Key Laboratory of Metabolic Diseases, Key Laboratory of Immune Microenvironment and Disease-Ministry of Education, Department of Physiology and Pathophysiology, Collaborative Innovation Center of Tianjin for Medical Epigenetics, Tianjin Medical University, Tianjin, China
| | - Liu Yao
- Tianjin Key Laboratory of Metabolic Diseases, Key Laboratory of Immune Microenvironment and Disease-Ministry of Education, Department of Physiology and Pathophysiology, Collaborative Innovation Center of Tianjin for Medical Epigenetics, Tianjin Medical University, Tianjin, China
| | - Chun Chang
- Department of Pulmonary and Critical Care Medicine, Peking University Third Hospital, Beijing, China
| |
Collapse
|
27
|
Nicholson RJ, Pezzolesi MG, Summers SA. Rotten to the Cortex: Ceramide-Mediated Lipotoxicity in Diabetic Kidney Disease. Front Endocrinol (Lausanne) 2021; 11:622692. [PMID: 33584550 PMCID: PMC7876379 DOI: 10.3389/fendo.2020.622692] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 12/11/2020] [Indexed: 12/11/2022] Open
Abstract
Diabetic kidney disease (DKD) is a prevalent and progressive comorbidity of diabetes mellitus that increases one's risk of developing renal failure. Progress toward development of better DKD therapeutics is limited by an incomplete understanding of forces driving and connecting the various features of DKD, which include renal steatosis, fibrosis, and microvascular dysfunction. Herein we review the literature supporting roles for bioactive ceramides as inducers of local and systemic DKD pathology. In rodent models of DKD, renal ceramides are elevated, and genetic and pharmacological ceramide-lowering interventions improve kidney function and ameliorate DKD histopathology. In humans, circulating sphingolipid profiles distinguish human DKD patients from diabetic controls. These studies highlight the potential for ceramide to serve as a central and therapeutically tractable lipid mediator of DKD.
Collapse
Affiliation(s)
- Rebekah J. Nicholson
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, United States
- Diabetes and Metabolism Research Center, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Marcus G. Pezzolesi
- Diabetes and Metabolism Research Center, University of Utah School of Medicine, Salt Lake City, UT, United States
- Division of Nephrology and Hypertension, Department of Internal Medicine, University of Utah, Salt Lake City, UT, United States
| | - Scott A. Summers
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, United States
- Diabetes and Metabolism Research Center, University of Utah School of Medicine, Salt Lake City, UT, United States
| |
Collapse
|
28
|
Revercomb L, Hanmandlu A, Wareing N, Akkanti B, Karmouty-Quintana H. Mechanisms of Pulmonary Hypertension in Acute Respiratory Distress Syndrome (ARDS). Front Mol Biosci 2021; 7:624093. [PMID: 33537342 PMCID: PMC7848216 DOI: 10.3389/fmolb.2020.624093] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 12/09/2020] [Indexed: 12/19/2022] Open
Abstract
Background: Acute respiratory distress syndrome (ARDS) is a severe and often fatal disease. The causes that lead to ARDS are multiple and include inhalation of salt water, smoke particles, or as a result of damage caused by respiratory viruses. ARDS can also arise due to systemic complications such as blood transfusions, sepsis, or pancreatitis. Unfortunately, despite a high mortality rate of 40%, there are limited treatment options available for ARDS outside of last resort options such as mechanical ventilation and extracorporeal support strategies. Aim of review: A complication of ARDS is the development of pulmonary hypertension (PH); however, the mechanisms that lead to PH in ARDS are not fully understood. In this review, we summarize the known mechanisms that promote PH in ARDS. Key scientific concepts of review: (1) Provide an overview of acute respiratory distress syndrome; (2) delineate the mechanisms that contribute to the development of PH in ARDS; (3) address the implications of PH in the setting of coronavirus disease 2019 (COVID-19).
Collapse
Affiliation(s)
- Lucy Revercomb
- Department of BioSciences, Rice University, Houston, TX, United States
- Department of Biochemistry and Molecular Biology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Ankit Hanmandlu
- Department of Biochemistry and Molecular Biology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Nancy Wareing
- Department of Biochemistry and Molecular Biology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Bindu Akkanti
- Divisions of Critical Care, Pulmonary and Sleep Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Harry Karmouty-Quintana
- Department of Biochemistry and Molecular Biology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
- Divisions of Critical Care, Pulmonary and Sleep Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| |
Collapse
|
29
|
Hoevenaar M, Goossens D, Roorda J. Angiotensin-converting enzyme 2, the complement system, the kallikrein-kinin system, type-2 diabetes, interleukin-6, and their interactions regarding the complex COVID-19 pathophysiological crossroads. J Renin Angiotensin Aldosterone Syst 2020; 21:1470320320979097. [PMID: 33283602 PMCID: PMC7724427 DOI: 10.1177/1470320320979097] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 11/11/2020] [Indexed: 12/15/2022] Open
Abstract
Because of the current COVID-19-pandemic, the world is currently being held hostage in various lockdowns. ACE2 facilitates SARS-CoV-2 cell-entry, and is at the very center of several pathophysiological pathways regarding the RAAS, CS, KKS, T2DM, and IL-6. Their interactions with severe COVID-19 complications (e.g. ARDS and thrombosis), and potential therapeutic targets for pharmacological intervention, will be reviewed.
Collapse
Affiliation(s)
| | | | - Janne Roorda
- Medical Doctor, General Practice
van Dijk, Oisterwijk, The Netherlands
| |
Collapse
|
30
|
Ryan JJ, Melendres-Groves L, Zamanian RT, Oudiz RJ, Chakinala M, Rosenzweig EB, Gomberg-Maitland M. Care of patients with pulmonary arterial hypertension during the coronavirus (COVID-19) pandemic. Pulm Circ 2020; 10:2045894020920153. [PMID: 32426111 PMCID: PMC7222260 DOI: 10.1177/2045894020920153] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 03/30/2020] [Indexed: 01/08/2023] Open
Abstract
The COVID-19 pandemic presents many unique challenges when caring for patients with pulmonary hypertension. The COVID-19 pandemic has altered routine standard of care practice and the acute management particularly for those patients with pulmonary arterial hypertension, where pulmonary arterial hypertension-specific treatments are used. It is important to balance the ongoing care and evaluation of pulmonary arterial hypertension patients with "exposure risk" to COVID-19 for patients coming to clinic or the hospital. If there is a morbidity and mortality benefit from starting pulmonary arterial hypertension therapies, for example in a patient with high-likelihood of pulmonary arterial hypertension, then it remains important to complete the thorough evaluation. However, the COVID-19 outbreak may also represent a unique time when pulmonary hypertension experts have to weigh the risks and benefits of the diagnostic work-up including potential exposure to COVID-19 versus initiating targeted pulmonary arterial hypertension therapy in a select high-risk, high likelihood World Symposium Pulmonary Hypertension Group 1 pulmonary arterial hypertension patients. This document will highlight some of the issues facing providers, patients, and the pulmonary arterial hypertension community in real-time as the COVID-19 pandemic is evolving and is intended to share expected common clinical scenarios and best clinical practices to help the community at-large.
Collapse
Affiliation(s)
- John J. Ryan
- Division of Cardiovascular Medicine, University of Utah, Salt Lake City, UT, USA
| | - Lana Melendres-Groves
- Division of Pulmonary and Critical Care Medicine, University of New Mexico, Albuquerque, NM, USA
| | - Roham T. Zamanian
- Division of Pulmonary and Critical Care Medicine, Stanford University, Stanford, CA, USA
| | - Ronald J. Oudiz
- Division of Cardiology, Lundquist Institute for Biomedical Innovation & Research at Harbor–UCLA Medical Center, Torrance, CA, USA
| | - Murali Chakinala
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Erika B. Rosenzweig
- Division of Pediatric Cardiology, Columbia University College of Physicians & Surgeons, New York, NY, USA
| | - Mardi Gomberg-Maitland
- Division of Cardiovascular Medicine, George Washington University Medicine and Health Sciences, Washington, DC, USA
| |
Collapse
|
31
|
Iron and Sphingolipids as Common Players of (Mal)Adaptation to Hypoxia in Pulmonary Diseases. Int J Mol Sci 2020; 21:ijms21010307. [PMID: 31906427 PMCID: PMC6981703 DOI: 10.3390/ijms21010307] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 12/24/2019] [Accepted: 12/31/2019] [Indexed: 12/11/2022] Open
Abstract
Hypoxia, or lack of oxygen, can occur in both physiological (high altitude) and pathological conditions (respiratory diseases). In this narrative review, we introduce high altitude pulmonary edema (HAPE), acute respiratory distress syndrome (ARDS), Chronic Obstructive Pulmonary Disease (COPD), and Cystic Fibrosis (CF) as examples of maladaptation to hypoxia, and highlight some of the potential mechanisms influencing the prognosis of the affected patients. Among the specific pathways modulated in response to hypoxia, iron metabolism has been widely explored in recent years. Recent evidence emphasizes hepcidin as highly involved in the compensatory response to hypoxia in healthy subjects. A less investigated field in the adaptation to hypoxia is the sphingolipid (SPL) metabolism, especially through Ceramide and sphingosine 1 phosphate. Both individually and in concert, iron and SPL are active players of the (mal)adaptation to physiological hypoxia, which can result in the pathological HAPE. Our aim is to identify some pathways and/or markers involved in the physiological adaptation to low atmospheric pressures (high altitudes) that could be involved in pathological adaptation to hypoxia as it occurs in pulmonary inflammatory diseases. Hepcidin, Cer, S1P, and their interplay in hypoxia are raising growing interest both as prognostic factors and therapeutical targets.
Collapse
|
32
|
Li Y, Lu Z, Zhang L, Kirkwood KL, Lopes-Virella MF, Huang Y. Acid sphingomyelinase deficiency exacerbates LPS-induced experimental periodontitis. Oral Dis 2019; 26:637-646. [PMID: 31883406 DOI: 10.1111/odi.13268] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 11/11/2019] [Accepted: 12/06/2019] [Indexed: 12/29/2022]
Abstract
BACKGROUND Mutation of the gene for acid sphingomyelinase (ASMase) causes Niemann-Pick disease. However, the effect of ASMase deficiency on periodontal health is unknown. Periodontal disease is a disease resulting from infection and inflammation of periodontal tissue and alveolar bone that support the teeth. The goal of this study was to determine the role of ASMase deficiency in periodontal inflammation and alveolar bone loss. METHODS We induced periodontitis in wild-type and ASMase-deficient (ASMase-/- ) mice with periodontal lipopolysaccharide (LPS) injection and compared the alveolar bone loss and periodontal inflammation between these mice. RESULTS Results showed that ASMase deficiency did not significantly change metabolic parameters, but exacerbated LPS-induced alveolar bone loss, osteoclastogenesis, and periodontal tissue inflammation. To understand the mechanisms by which ASMase deficiency aggravates LPS-induced periodontitis, we analyzed sphingolipids in periodontal tissues. Results showed that ASMase deficiency led to increases in not only sphingomyelin, but also ceramide (CER), a bioactive sphingolipid known to promote inflammation. Results further showed that ASMase deficiency increased CER de novo synthesis. CONCLUSION ASMase deficiency exacerbated LPS-induced alveolar bone loss and periodontal inflammation. ASMase deficiency leads to an unexpected CER increase by stimulating de novo synthesis CER, which is likely to be involved in the ASMase deficiency-exacerbated periodontitis.
Collapse
Affiliation(s)
- Yanchun Li
- Division of Endocrinology, Diabetes and Medical Genetics, Department of Medicine, College of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Zhongyang Lu
- Division of Endocrinology, Diabetes and Medical Genetics, Department of Medicine, College of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Lixia Zhang
- Departments of Oral Biology, School of Dental Medicine, University at Buffalo, Buffalo, New York
| | - Keith L Kirkwood
- Departments of Oral Biology, School of Dental Medicine, University at Buffalo, Buffalo, New York.,Department of Oral Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Maria F Lopes-Virella
- Division of Endocrinology, Diabetes and Medical Genetics, Department of Medicine, College of Medicine, Medical University of South Carolina, Charleston, South Carolina.,Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina
| | - Yan Huang
- Division of Endocrinology, Diabetes and Medical Genetics, Department of Medicine, College of Medicine, Medical University of South Carolina, Charleston, South Carolina.,Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina
| |
Collapse
|
33
|
IL-38 is a biomarker for acute respiratory distress syndrome in humans and down-regulates Th17 differentiation in vivo. Clin Immunol 2019; 210:108315. [PMID: 31756565 DOI: 10.1016/j.clim.2019.108315] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 06/21/2019] [Accepted: 11/19/2019] [Indexed: 11/20/2022]
|
34
|
Hollmann C, Wiese T, Dennstädt F, Fink J, Schneider-Schaulies J, Beyersdorf N. Translational Approaches Targeting Ceramide Generation From Sphingomyelin in T Cells to Modulate Immunity in Humans. Front Immunol 2019; 10:2363. [PMID: 31681273 PMCID: PMC6798155 DOI: 10.3389/fimmu.2019.02363] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 09/19/2019] [Indexed: 12/12/2022] Open
Abstract
In T cells, as in all other cells of the body, sphingolipids form important structural components of membranes. Due to metabolic modifications, sphingolipids additionally play an active part in the signaling of cell surface receptors of T cells like the T cell receptor or the co-stimulatory molecule CD28. Moreover, the sphingolipid composition of their membranes crucially affects the integrity and function of subcellular compartments such as the lysosome. Previously, studying sphingolipid metabolism has been severely hampered by the limited number of analytical methods/model systems available. Besides well-established high resolution mass spectrometry new tools are now available like novel minimally modified sphingolipid subspecies for click chemistry as well as recently generated mouse mutants with deficiencies/overexpression of sphingolipid-modifying enzymes. Making use of these tools we and others discovered that the sphingolipid sphingomyelin is metabolized to ceramide to different degrees in distinct T cell subpopulations of mice and humans. This knowledge has already been translated into novel immunomodulatory approaches in mice and will in the future hopefully also be applicable to humans. In this paper we are, thus, summarizing the most recent findings on the impact of sphingolipid metabolism on T cell activation, differentiation, and effector functions. Moreover, we are discussing the therapeutic concepts arising from these insights and drugs or drug candidates which are already in clinical use or could be developed for clinical use in patients with diseases as distant as major depression and chronic viral infection.
Collapse
Affiliation(s)
- Claudia Hollmann
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Teresa Wiese
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Fabio Dennstädt
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Julian Fink
- Institute of Organic Chemistry, University of Würzburg, Würzburg, Germany
| | | | - Niklas Beyersdorf
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| |
Collapse
|
35
|
Xiao Q, Wang Y, Sun L, Lu S, Li J, Chen Y, Wu Y. Immediate Prone Positioning After Massive Gastric Aspiration Reduces Lung Injury Possibly by Attenuating Interleukin-6-Mediated Lung-Tissue Inflammation in Pigs. Biol Res Nurs 2019; 22:64-74. [PMID: 31529988 DOI: 10.1177/1099800419875521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Gastric aspiration, which can cause acute, diffuse, inflammatory lung injury, is of particular concern in critically ill patients. This study aimed to determine the effects of immediate prone positioning on the degree of lung injury and inflammatory response induced by gastric aspiration. Following induction of gastric aspiration by injection of gastric fluid, 16 healthy pigs were randomized to one of two groups: supine position (SP) or prone position (PP). After ventilation and monitoring for 6 hr, all pigs were euthanized. The ratio of the partial pressure of arterial oxygen/fraction of inspired oxygen (PaO2/FIO2) and the partial pressure of arterial carbon dioxide (PaCO2) were recorded during the 6-hr study period. Serum levels of interleukin (IL)-6 were measured every 2 hr, and the mean optical density (MOD) of IL-6 in lung tissues and lung-injury scores were measured at the end of the experiment. The PP group showed a significantly higher PaO2/FIO2 ratio, lower serum IL-6 concentration (p = .015), lower lung-injury scores (p = .012), and lower IL-6 concentration and MOD of IL-6 in lung tissue, especially in dorsal (p = .001, p = .021, respectively) and nondependent regions (p = .005, p = .035, respectively) than the SP group. There were no statistically significant differences in PaCO2 between the groups. Lung-injury severity was positively correlated with the IL-6 concentration and MOD of IL-6 in lung tissues (p < .05). These results suggest that immediate prone positioning alleviated the degree of aspiration-induced lung injury, possibly through mitigating IL-6-mediated lung inflammation.
Collapse
Affiliation(s)
- Qian Xiao
- School of Nursing, Capital Medical University, Beijing, China
| | - Yanling Wang
- School of Nursing, Capital Medical University, Beijing, China
| | - Liu Sun
- School of Nursing, Capital Medical University, Beijing, China
| | - Sai Lu
- College of Health and Biomedicine, Victoria University, Melbourne, Australia
| | - Jia Li
- School of Nursing, Capital Medical University, Beijing, China
| | - Yun Chen
- School of Nursing, Capital Medical University, Beijing, China
| | - Ying Wu
- School of Nursing, Capital Medical University, Beijing, China
| |
Collapse
|
36
|
Alvarez-Fuente M, Moreno L, Lopez-Ortego P, Arruza L, Avila-Alvarez A, Muro M, Gutierrez E, Zozaya C, Sanchez-Helguera G, Elorza D, Martinez-Ramas A, Villar G, Labrandero C, Martinez L, Casado T, Cuadrado I, Del Cerro MJ. Exploring clinical, echocardiographic and molecular biomarkers to predict bronchopulmonary dysplasia. PLoS One 2019; 14:e0213210. [PMID: 30840669 PMCID: PMC6402695 DOI: 10.1371/journal.pone.0213210] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 02/15/2019] [Indexed: 12/04/2022] Open
Abstract
Introduction Bronchopulmonary dysplasia (BPD) is the most common chronic lung disease in childhood, related to prematurity, and the most common cause of pulmonary hypertension (PH) secondary to pulmonary disease in children. Moderate and severe BPD have a worse outcome and relate more frequently with PH. The prediction of moderate or severe BPD development in extremely premature newborns is vital to implement preventive strategies. Starting with the hypothesis that molecular biomarkers were better than clinical and echocardiographic factors, this study aims to explore the ability of clinical, echocardiographic and analytical variables to predict moderate or severe BPD in a cohort of extremely preterm infants. Patients and methods We designed a prospective longitudinal study, in which we followed a cohort of preterm newborns (gestational age <28 weeks and weight ≤ 1250 grams). In these newborns we recorded weekly clinical and echocardiographic variables as well as blood and tracheal aspirate samples, to analyze molecular biomarkers (IL-6, IL-1, IP10, uric acid, HGF, endothelin-1, VEGF, CCL5). Variables and samples were collected since birth up to week 36 (postmenstrual age), time-point at which the diagnosis of BPD is established. Results We included 50 patients with a median gestational age of 26 weeks (IQR 25–27) and weight of 871 g (SD 161,0) (range 590-1200g). Three patients were excluded due to an early death. Thirty-five patients (74.5%) developed BPD (mild n = 14, moderate n = 15, severe n = 6). We performed a logistic regression in order to identify risk factors for moderate or severe BPD. We compared two predictive models, one with two variables (mechanical ventilation and inter-ventricular septum flattening), and another-one with an additional molecular biomarker (ET-1). Conclusions The combination of clinical and echocardiographic variables is a valuable tool for determining the risk of BPD. We find the two variable model (mechanical ventilation and echocardiographic signs of PH) more practical for clinical and research purposes. Future research on BPD prediction should be oriented to explore the potential role of ET-1.
Collapse
Affiliation(s)
- Maria Alvarez-Fuente
- Pediatric Cardiology Department, Ramón y Cajal University Hospital, Madrid, Spain
| | - Laura Moreno
- Department of Pharmacology, School of Medicine, University Complutense of Madrid, Instituto de Investigación Sanitaria Gregorio Marañon (IiSGM), Ciber Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | | | - Luis Arruza
- Neonatology Department, Institute of the Child and Adolescent, Clínico San Carlos University Hospital-IdISSC, Madrid, Spain
| | | | - Marta Muro
- Neonatology Department, Puerta de Hierro University Hospital, Majadahonda, Madrid, Spain
| | - Enrique Gutierrez
- Public Health and Preventive Medicine Unit, School of Public Health- Instituto de Salud Carlos III, Madrid, Spain
| | - Carlos Zozaya
- Neonatology Department, La Paz University Hospital, Madrid, Spain
| | | | - Dolores Elorza
- Neonatology Department, La Paz University Hospital, Madrid, Spain
| | - Andrea Martinez-Ramas
- Department of Pharmacology, School of Medicine, University Complutense of Madrid, Instituto de Investigación Sanitaria Gregorio Marañon (IiSGM), Ciber Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Gema Villar
- Neonatology Department, Getafe University Hospital, Getafe, Madrid, Spain
| | - Carlos Labrandero
- Pediatric Cardiology Department, La Paz University Hospital, Madrid, Spain
| | - Lucia Martinez
- Neonatology Department, Getafe University Hospital, Getafe, Madrid, Spain
| | - Teresa Casado
- Pediatric Cardiology Department, Ramón y Cajal University Hospital, Madrid, Spain
| | - Irene Cuadrado
- Neonatology Department, Getafe University Hospital, Getafe, Madrid, Spain
| | | |
Collapse
|
37
|
Cogolludo A, Villamor E, Perez-Vizcaino F, Moreno L. Ceramide and Regulation of Vascular Tone. Int J Mol Sci 2019; 20:ijms20020411. [PMID: 30669371 PMCID: PMC6359388 DOI: 10.3390/ijms20020411] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 01/02/2019] [Accepted: 01/16/2019] [Indexed: 02/07/2023] Open
Abstract
In addition to playing a role as a structural component of cellular membranes, ceramide is now clearly recognized as a bioactive lipid implicated in a variety of physiological functions. This review aims to provide updated information on the role of ceramide in the regulation of vascular tone. Ceramide may induce vasodilator or vasoconstrictor effects by interacting with several signaling pathways in endothelial and smooth muscle cells. There is a clear, albeit complex, interaction between ceramide and redox signaling. In fact, reactive oxygen species (ROS) activate different ceramide generating pathways and, conversely, ceramide is known to increase ROS production. In recent years, ceramide has emerged as a novel key player in oxygen sensing in vascular cells and mediating vascular responses of crucial physiological relevance such as hypoxic pulmonary vasoconstriction (HPV) or normoxic ductus arteriosus constriction. Likewise, a growing body of evidence over the last years suggests that exaggerated production of vascular ceramide may have detrimental effects in a number of pathological processes including cardiovascular and lung diseases.
Collapse
Affiliation(s)
- Angel Cogolludo
- Department of Pharmacology and Toxicology, School of Medicine, University Complutense of Madrid, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Ciudad Universitaria S/N, 28040 Madrid, Spain.
- Ciber Enfermedades Respiratorias (CIBERES), 28029 Madrid, Spain.
| | - Eduardo Villamor
- Department of Pediatrics, Maastricht University Medical Center (MUMC+), School for Oncology and Developmental Biology (GROW), 6202 AZ Maastricht, The Netherlands.
| | - Francisco Perez-Vizcaino
- Department of Pharmacology and Toxicology, School of Medicine, University Complutense of Madrid, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Ciudad Universitaria S/N, 28040 Madrid, Spain.
- Ciber Enfermedades Respiratorias (CIBERES), 28029 Madrid, Spain.
| | - Laura Moreno
- Department of Pharmacology and Toxicology, School of Medicine, University Complutense of Madrid, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Ciudad Universitaria S/N, 28040 Madrid, Spain.
- Ciber Enfermedades Respiratorias (CIBERES), 28029 Madrid, Spain.
| |
Collapse
|
38
|
Off-label mesenchymal stromal cell treatment in two infants with severe bronchopulmonary dysplasia: clinical course and biomarkers profile. Cytotherapy 2018; 20:1337-1344. [PMID: 30327248 DOI: 10.1016/j.jcyt.2018.09.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 09/12/2018] [Accepted: 09/13/2018] [Indexed: 12/19/2022]
Abstract
BACKGROUND Bronchopulmonary dysplasia (BPD) is the most prevalent sequelae of premature birth, for which therapeutic options are currently limited. Mesenchymal stromal cells (MSCs) are a potential therapy for prevention or reversal of BPD. SERIES OF CASES We report on two infants with severe BPD in whom off-label treatment with repeated intravenous doses of allogeneic bone marrow-derived MSCs were administered. We analyzed the temporal profile of serum and tracheal cytokines and growth factors as well as safety, tolerability and clinical response. The administration of repeated intravenous doses of MSCs in two human babies with severe and advanced BPD was feasible and safe and was associated with a decrease of pro-inflammatory molecules and lung injury biomarkers. Both patients were at very advanced stages of BPD with very severe lung fibrosis and did not survive the disease. CONCLUSIONS MSCs are a promising therapy for BPD, but they should be administered in early stages of the disease.
Collapse
|
39
|
Rico JE, Giesy SL, Haughey NJ, Boisclair YR, McFadden JW. Intravenous Triacylglycerol Infusion Promotes Ceramide Accumulation and Hepatic Steatosis in Dairy Cows. J Nutr 2018; 148:1529-1535. [PMID: 30281114 DOI: 10.1093/jn/nxy155] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 06/27/2018] [Indexed: 11/12/2022] Open
Abstract
Background Increased plasma free fatty acids (FFAs) impair insulin sensitivity in dairy cows via unknown mechanisms. In nonruminants, saturated FFAs upregulate the hepatic synthesis and secretion of ceramide, which inhibits insulin action. Objective We aimed to determine whether an increase in plasma FFAs promotes hepatic and plasma ceramide accumulation in dairy cows. Methods Six nonpregnant, nonlactating Holstein cows were used in a study with a crossover design and treatments consisting of intravenous infusion of either saline (control) or triacylglycerol emulsion (TG; 20 g/h) for 16 h. The feeding level was set at 120% of energy requirements. Blood was collected at regular intervals and liver was biopsied at 16 h. Ceramides, monohexosylceramides (Glc/Gal-Cer), lactosylceramides (LacCer), and sphingomyelins (SMs) in plasma and liver were profiled. Hepatic expression of ceramide synthases was determined. Data were analyzed with the use of mixed models, regressions, and Spearman rank correlations. Results After 16 h of infusion, plasma FFA concentrations were >5-fold and liver triacylglycerol concentrations were 4-fold greater in TG cows, relative to control. Plasma total and very long-chain ceramide (e.g., C24:0-ceramide) concentrations increased ∼4-fold in TG over control by hour 16 of infusion, while C16:0-ceramide were not modified by TG. Infusion of TG increased plasma Glc/Gal-Cer (e.g., C16:0-Glc/Gal-Cer, 4-fold by hour 16) relative to control, but did not alter LacCer or SM concentrations. Hepatic ceramide concentrations increased with TG relative to control (e.g., C24:0-ceramide by 1.7-fold). Hepatic expression of ceramide synthase 2 was 60% greater after TG infusion compared with the control. Circulating ceramides were related to circulating FFA and hepatic triacylglycerol concentrations (e.g., C24:0-ceramide, ρ = 0.73 and 0.80, respectively; P < 0.001). Conclusion Hepatic ceramide synthesis is associated with elevations in circulating FFAs and hepatic triacylglycerol during the induction of hyperlipidemia in dairy cows. This work supports the emerging evidence for the role of ceramide during hepatic steatosis and insulin antagonism in cows.
Collapse
Affiliation(s)
- J Eduardo Rico
- Department of Animal Science, Cornell University, Ithaca, NY.,Division of Animal and Nutritional Sciences, West Virginia University, Morgantown, WV
| | - Sarah L Giesy
- Department of Animal Science, Cornell University, Ithaca, NY
| | - Norman J Haughey
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD
| | | | | |
Collapse
|
40
|
Himmat S, Alzamil A, Aboelnazar N, Hatami S, White C, Dromparis P, Mengel M, Freed D, Nagendran J. A Decrease in Hypoxic Pulmonary Vasoconstriction Correlates With Increased Inflammation During Extended Normothermic Ex Vivo Lung Perfusion. Artif Organs 2017; 42:271-279. [PMID: 29266272 DOI: 10.1111/aor.13017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 07/12/2017] [Accepted: 08/04/2017] [Indexed: 02/06/2023]
Abstract
Normothermic ex vivo lung perfusion (EVLP) is an evolving technology to evaluate function of donor lungs to determine suitability for transplantation. We hypothesize that hypoxic pulmonary vasoconstriction (HPV) during EVLP will provide a more sensitive parameter of lung function to determine donor lung quality for lung transplantation. Eight porcine lungs were procured, and subsequently underwent EVLP with autologous blood and STEEN solution for 10 h. Standard physiologic parameters including dynamic compliance, peak airway pressure, and pulmonary vascular resistance (PVR) remained stable (P = 0.055), mean oxygenation (PO2 /FiO2 ) was 400 ± 18 mm Hg on average throughout perfusion. Response to hypoxia resulted in a robust increase in PVR (ΔPVR) up to 4 h of perfusion, however the HPV response then blunted beyond T6 (P < 0.01). The decrease in HPV response inversely correlated to cytokine concentrations of Interleukin-6 and tumor necrosis factor-α (P < 0.01). Despite acceptable lung oxygenation and standard physiologic parameters during 10 h of EVLP, there is a subclinical deterioration of lung function. HPV challenges can be performed during EVLP as a simple and more sensitive index of pulmonary vascular reactivity.
Collapse
Affiliation(s)
- Sayed Himmat
- Division of Cardiac Surgery, Department of Surgery, University of Alberta, Edmonton, Alberta, Canada.,Mazankowski Alberta Heart Institute, Edmonton, Alberta, Canada
| | - Almothana Alzamil
- Division of Cardiac Surgery, Department of Surgery, University of Alberta, Edmonton, Alberta, Canada.,Mazankowski Alberta Heart Institute, Edmonton, Alberta, Canada
| | - Nader Aboelnazar
- Division of Cardiac Surgery, Department of Surgery, University of Alberta, Edmonton, Alberta, Canada.,Mazankowski Alberta Heart Institute, Edmonton, Alberta, Canada
| | - Sanaz Hatami
- Division of Cardiac Surgery, Department of Surgery, University of Alberta, Edmonton, Alberta, Canada.,Mazankowski Alberta Heart Institute, Edmonton, Alberta, Canada
| | - Christopher White
- Division of Cardiac Surgery, Department of Surgery, University of Alberta, Edmonton, Alberta, Canada.,Mazankowski Alberta Heart Institute, Edmonton, Alberta, Canada
| | - Peter Dromparis
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta, Canada
| | - Michael Mengel
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta, Canada
| | - Darren Freed
- Division of Cardiac Surgery, Department of Surgery, University of Alberta, Edmonton, Alberta, Canada.,Mazankowski Alberta Heart Institute, Edmonton, Alberta, Canada.,Division of Cardiac Surgery, Department of Surgery, Alberta Transplant Institute, Edmonton, Alberta, Canada.,Canadian National Transplant Research Program, Canadian Institute for Health Research, Edmonton, Alberta, Canada
| | - Jayan Nagendran
- Division of Cardiac Surgery, Department of Surgery, University of Alberta, Edmonton, Alberta, Canada.,Mazankowski Alberta Heart Institute, Edmonton, Alberta, Canada.,Division of Cardiac Surgery, Department of Surgery, Alberta Transplant Institute, Edmonton, Alberta, Canada.,Canadian National Transplant Research Program, Canadian Institute for Health Research, Edmonton, Alberta, Canada
| |
Collapse
|
41
|
Fernández-Alfonso MS, Somoza B, Tsvetkov D, Kuczmanski A, Dashwood M, Gil-Ortega M. Role of Perivascular Adipose Tissue in Health and Disease. Compr Physiol 2017; 8:23-59. [PMID: 29357124 DOI: 10.1002/cphy.c170004] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Perivascular adipose tissue (PVAT) is cushion of fat tissue surrounding blood vessels, which is phenotypically different from other adipose tissue depots. PVAT is composed of adipocytes and stromal vascular fraction, constituted by different populations of immune cells, endothelial cells, and adipose-derived stromal cells. It expresses and releases an important number of vasoactive factors with paracrine effects on vascular structure and function. In healthy individuals, these factors elicit a net anticontractile and anti-inflammatory paracrine effect aimed at meeting hemodynamic and metabolic demands of specific organs and regions of the body. Pathophysiological situations, such as obesity, diabetes or hypertension, induce changes in its amount and in the expression pattern of vasoactive factors leading to a PVAT dysfunction in which the beneficial paracrine influence of PVAT is shifted to a pro-oxidant, proinflammatory, contractile, and trophic environment leading to functional and structural cardiovascular alterations and cardiovascular disease. Many different PVATs surrounding a variety of blood vessels have been described and exhibit regional differences. Both protective and deleterious influence of PVAT differs regionally depending on the specific vascular bed contributing to variations in the susceptibility of arteries and veins to vascular disease. PVAT therefore, might represent a novel target for pharmacological intervention in cardiovascular disease. © 2018 American Physiological Society. Compr Physiol 8:23-59, 2018.
Collapse
Affiliation(s)
| | - Beatriz Somoza
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU-San Pablo, Madrid, Spain
| | - Dmitry Tsvetkov
- Department of Anestesiology, Perioperative and Pain Medicine, HELIOS Klinikum, Berlin-Buch GmbH, Germany.,Institute of Experimental and Clinical Pharmacology and Toxicology, Department of Pharmacology and Experimental Therapy, Eberhard Karls University Hospitals and Clinics, and Interfaculty Center of Pharmacogenomics and Drug Research, Tübingen, Germany
| | - Artur Kuczmanski
- Department of Anestesiology, Perioperative and Pain Medicine, HELIOS Klinikum, Berlin-Buch GmbH, Germany
| | - Mick Dashwood
- Royal Free Hospital Campus, University College Medical School, London, United Kingdom
| | - Marta Gil-Ortega
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU-San Pablo, Madrid, Spain
| |
Collapse
|
42
|
Kulkarni H, Mamtani M, Blangero J, Curran JE. Lipidomics in the Study of Hypertension in Metabolic Syndrome. Curr Hypertens Rep 2017; 19:7. [PMID: 28168678 DOI: 10.1007/s11906-017-0705-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
43
|
Chamorro V, Pandolfi R, Moreno L, Barreira B, Martínez-Ramas A, Morales-Cano D, Ruiz-Cabello J, Lorente JA, Duarte J, Cogolludo Á, Alvarez-Sala JL, Perez-Vizcaino F. Effects of Quercetin in a Rat Model of Hemorrhagic Traumatic Shock and Reperfusion. Molecules 2016; 21:molecules21121739. [PMID: 27999410 PMCID: PMC6273949 DOI: 10.3390/molecules21121739] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Revised: 11/30/2016] [Accepted: 12/07/2016] [Indexed: 11/16/2022] Open
Abstract
Background: We hypothesized that treatment with quercetin could result in improved hemodynamics, lung inflammatory parameters and mortality in a rat model of hemorrhagic shock. Methods: Rats were anesthetized (80 mg/kg ketamine plus 8 mg/kg xylazine i.p.). The protocol included laparotomy for 15 min (trauma), hemorrhagic shock (blood withdrawal to reduce the mean arterial pressure to 35 mmHg) for 75 min and resuscitation by re-infusion of all the shed blood plus lactate Ringer for 90 min. Intravenous quercetin (50 mg/kg) or vehicle were administered during resuscitation. Results: There was a trend for increased survival 84.6% (11/13) in the treated group vs. the shock group 68.4% (13/19, p > 0.05 Kaplan–Meier). Quercetin fully prevented the development of lung edema. The activity of aSMase was increased in the shock group compared to the sham group and the quercetin prevented this effect. However, other inflammatory markers such as myeloperoxidase activity, interleukin-6 in plasma or bronchoalveolar fluid were similar in the sham and shock groups. We found no bacterial DNA in plasma in these animals. Conclusions: Quercetin partially prevented the changes in blood pressure and lung injury in shock associated to hemorrhage and reperfusion.
Collapse
Affiliation(s)
- Virginia Chamorro
- Departamento de Farmacología, Facultad de Medicina, Universidad Complutense de Madrid, Madrid 28040, Spain.
- Centro de Investigaciones Biomédicas en Red de Enfermedades Respiratorias (Ciberes), Madrid 28029, Spain.
- Instituto de Investigación Sanitaria Gregorio Marañón (IISGM), Madrid 28007, Spain.
| | - Rachele Pandolfi
- Departamento de Farmacología, Facultad de Medicina, Universidad Complutense de Madrid, Madrid 28040, Spain.
- Centro de Investigaciones Biomédicas en Red de Enfermedades Respiratorias (Ciberes), Madrid 28029, Spain.
- Instituto de Investigación Sanitaria Gregorio Marañón (IISGM), Madrid 28007, Spain.
| | - Laura Moreno
- Departamento de Farmacología, Facultad de Medicina, Universidad Complutense de Madrid, Madrid 28040, Spain.
- Centro de Investigaciones Biomédicas en Red de Enfermedades Respiratorias (Ciberes), Madrid 28029, Spain.
- Instituto de Investigación Sanitaria Gregorio Marañón (IISGM), Madrid 28007, Spain.
| | - Bianca Barreira
- Centro de Investigaciones Biomédicas en Red de Enfermedades Respiratorias (Ciberes), Madrid 28029, Spain.
- Instituto de Investigación Sanitaria Gregorio Marañón (IISGM), Madrid 28007, Spain.
| | - Andrea Martínez-Ramas
- Centro de Investigaciones Biomédicas en Red de Enfermedades Respiratorias (Ciberes), Madrid 28029, Spain.
- Instituto de Investigación Sanitaria Gregorio Marañón (IISGM), Madrid 28007, Spain.
| | - Daniel Morales-Cano
- Departamento de Farmacología, Facultad de Medicina, Universidad Complutense de Madrid, Madrid 28040, Spain.
- Centro de Investigaciones Biomédicas en Red de Enfermedades Respiratorias (Ciberes), Madrid 28029, Spain.
- Instituto de Investigación Sanitaria Gregorio Marañón (IISGM), Madrid 28007, Spain.
| | - Jesús Ruiz-Cabello
- Centro de Investigaciones Biomédicas en Red de Enfermedades Respiratorias (Ciberes), Madrid 28029, Spain.
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernández Almagro 3, Madrid 28029, Spain.
| | - José Angel Lorente
- Centro de Investigaciones Biomédicas en Red de Enfermedades Respiratorias (Ciberes), Madrid 28029, Spain.
- Servicio de Medicina Intensiva, Hospital Universitario de Getafe, Getafe, Madrid 28905, Spain.
- Universidad Europea de Madrid, Madrid 28905, Spain.
| | - Juan Duarte
- Departamento de Farmacología, Facultad de Farmacia, Universidad de Granada, Granada 18071, Spain.
- Centro de Investigaciones Biomédicas en Red de Enfermedades Cardiovasculares (Ciberec), Madrid 28029, Spain.
| | - Ángel Cogolludo
- Departamento de Farmacología, Facultad de Medicina, Universidad Complutense de Madrid, Madrid 28040, Spain.
- Centro de Investigaciones Biomédicas en Red de Enfermedades Respiratorias (Ciberes), Madrid 28029, Spain.
- Instituto de Investigación Sanitaria Gregorio Marañón (IISGM), Madrid 28007, Spain.
| | - José Luis Alvarez-Sala
- Departamento de Medicina, Facultad de Medicina, Universidad Complutense de Madrid, Madrid 28040, Spain.
- Servicio de Neumología, Instituto de Investigación Sanitaria San Carlos (IdISSC), Madrid 28040, Spain.
| | - Francisco Perez-Vizcaino
- Departamento de Farmacología, Facultad de Medicina, Universidad Complutense de Madrid, Madrid 28040, Spain.
- Centro de Investigaciones Biomédicas en Red de Enfermedades Respiratorias (Ciberes), Madrid 28029, Spain.
- Instituto de Investigación Sanitaria Gregorio Marañón (IISGM), Madrid 28007, Spain.
| |
Collapse
|