1
|
Gheitasi I, Akbari G, Savari F. Physiological and cellular mechanisms of ischemic preconditioning microRNAs-mediated in underlying of ischemia/reperfusion injury in different organs. Mol Cell Biochem 2025; 480:855-868. [PMID: 39001984 DOI: 10.1007/s11010-024-05052-7] [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: 03/28/2024] [Accepted: 06/10/2024] [Indexed: 07/15/2024]
Abstract
Ischemia-reperfusion (I/R) injury, as a pathological phenomenon, takes place when blood supply to an organ is disrupted and then aggravated during restoration of blood flow. Ischemic preconditioning (IPC) is a potent method for attenuating subsequent events of IR damage in numerous organs. IPC protocol is determined by a brief and sequential time periods of I/R before the main ischemia. MicroRNAs are endogenous non-coding RNAs that regulate post-transcriptionally target mRNA translation via degrading it and/or suppressing protein synthesis. This review introduces the physiological and cellular mechanisms of ischemic preconditioning microRNAs-mediated after I/R insult in different organs such as the liver, kidney, heart, brain, and intestine. Data of this review have been collected from the scientific articles published in databases such as Science Direct, Scopus, PubMed, Web of Science, and Scientific Information Database from 2000 to 2023. Based on these literature studies, IPC/IR intervention can affect cellular mechanisms including oxidative stress, apoptosis, angiogenesis, and inflammation through up-regulation or down-regulation of multiple microRNAs and their target genes.
Collapse
Affiliation(s)
- Izadpanah Gheitasi
- Department of Physiology, Faculty of Medicine, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Ghaidafeh Akbari
- Department of Physiology, Faculty of Medicine, Yasuj University of Medical Sciences, Yasuj, Iran.
| | - Feryal Savari
- Department of Medical Basic Sciences, Shoushtar Faculty of Medical Sciences, Shoushtar, Iran.
| |
Collapse
|
2
|
Schulman S, Makatsariya A, Khizroeva J, Bitsadze V, Kapanadze D. The Basic Principles of Pathophysiology of Venous Thrombosis. Int J Mol Sci 2024; 25:11447. [PMID: 39519000 PMCID: PMC11547114 DOI: 10.3390/ijms252111447] [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: 09/07/2024] [Revised: 10/19/2024] [Accepted: 10/20/2024] [Indexed: 11/16/2024] Open
Abstract
The past few decades have brought tremendous insight into the molecular and pathophysiological mechanisms responsible for thrombus generation. For a clinician, it is usually sufficient to explain the incident of deep vein thrombosis (DVT) with provoking factors such as trauma with vascular injury, immobilization, hormonal factors, or inherited or acquired coagulation defects. About half of DVTs are, however, lacking such triggers and are called unprovoked. Venous stasis and hypoxia at the valve sinus level may start a chain of reactions. The concept of immunothrombosis has added a new dimension to the old etiological triad of venous stasis, vessel wall injury, and changes in blood components. This is particularly important in COVID-19, where hyperinflammation, cytokines, and neutrophil extracellular traps are associated with the formation of microthrombi in the lungs. To better understand the mechanisms behind DVT and reach beyond the above-mentioned simplifications, animal models and clinical epidemiological studies have brought insight into the complex interplay between leukocytes, platelets, endothelium, cytokines, complements, and coagulation factors and inhibitors. These pathways and the interplay will be reviewed here, as well as the roles of cancer, anticancer drugs, and congenital thrombophilic defects on the molecular level in hypercoagulability and venous thromboembolism.
Collapse
Affiliation(s)
- Sam Schulman
- Department of Medicine, Thrombosis and Atherosclerosis Research Institute, McMaster University, 1280 Main St W, Hamilton, ON L8S 4L8, Canada
- Department of Obstetrics, Gynecology and Perinatal Medicine, The I.M. Sechenov First Moscow State Medical University (Sechenov University), Trubetskaya Str 8-2, 119435 Moscow, Russia; (A.M.); (J.K.); (V.B.)
| | - Alexander Makatsariya
- Department of Obstetrics, Gynecology and Perinatal Medicine, The I.M. Sechenov First Moscow State Medical University (Sechenov University), Trubetskaya Str 8-2, 119435 Moscow, Russia; (A.M.); (J.K.); (V.B.)
| | - Jamilya Khizroeva
- Department of Obstetrics, Gynecology and Perinatal Medicine, The I.M. Sechenov First Moscow State Medical University (Sechenov University), Trubetskaya Str 8-2, 119435 Moscow, Russia; (A.M.); (J.K.); (V.B.)
| | - Victoria Bitsadze
- Department of Obstetrics, Gynecology and Perinatal Medicine, The I.M. Sechenov First Moscow State Medical University (Sechenov University), Trubetskaya Str 8-2, 119435 Moscow, Russia; (A.M.); (J.K.); (V.B.)
| | - Daredzhan Kapanadze
- Center of Pathology of Pregnancy and Hemostasis «Medlabi», Tbilisi 340112, Georgia;
| |
Collapse
|
3
|
Kim HY, Jang HJ, Muthamil S, Shin UC, Lyu JH, Kim SW, Go Y, Park SH, Lee HG, Park JH. Novel insights into regulators and functional modulators of adipogenesis. Biomed Pharmacother 2024; 177:117073. [PMID: 38981239 DOI: 10.1016/j.biopha.2024.117073] [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: 04/15/2024] [Revised: 06/27/2024] [Accepted: 06/29/2024] [Indexed: 07/11/2024] Open
Abstract
Adipogenesis is a process that differentiates new adipocytes from precursor cells and is tightly regulated by several factors, including many transcription factors and various post-translational modifications. Recently, new roles of adipogenesis have been suggested in various diseases. However, the molecular mechanisms and functional modulation of these adipogenic genes remain poorly understood. This review summarizes the regulatory factors and modulators of adipogenesis and discusses future research directions to identify novel mechanisms regulating adipogenesis and the effects of adipogenic regulators in pathological conditions. The master adipogenic transcriptional factors PPARγ and C/EBPα were identified along with other crucial regulatory factors such as SREBP, Kroxs, STAT5, Wnt, FOXO1, SWI/SNF, KLFs, and PARPs. These transcriptional factors regulate adipogenesis through specific mechanisms, depending on the adipogenic stage. However, further studies related to the in vivo role of newly discovered adipogenic regulators and their function in various diseases are needed to develop new potent therapeutic strategies for metabolic diseases and cancer.
Collapse
Affiliation(s)
- Hyun-Yong Kim
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju, Jeollanam-do 58245, Republic of Korea; New Drug Development Center, Osong Medical Innovation Foundation, 123, Osongsaengmyeong-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungcheongbuk-do 28160, Republic of Korea.
| | - Hyun-Jun Jang
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju, Jeollanam-do 58245, Republic of Korea; Research Group of Personalized Diet, Korea Food Research Institute, Wanju-gun, Jeollabuk-do 55365, Republic of Korea.
| | - Subramanian Muthamil
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju, Jeollanam-do 58245, Republic of Korea.
| | - Ung Cheol Shin
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju, Jeollanam-do 58245, Republic of Korea.
| | - Ji-Hyo Lyu
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju, Jeollanam-do 58245, Republic of Korea.
| | - Seon-Wook Kim
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju, Jeollanam-do 58245, Republic of Korea.
| | - Younghoon Go
- Korean Medicine (KM)-application Center, Korea Institute of Oriental Medicine, Daegu 41062, Republic of Korea.
| | - Seong-Hoon Park
- Genetic and Epigenetic Toxicology Research Group, Korea Institute of Toxicology, Daejeon 34141, Republic of Korea.
| | - Hee Gu Lee
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea.
| | - Jun Hong Park
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju, Jeollanam-do 58245, Republic of Korea; University of Science & Technology (UST), KIOM campus, Korean Convergence Medicine Major, Daejeon 34054, Republic of Korea.
| |
Collapse
|
4
|
Tsaousi M, Sokou R, Pouliakis A, Politou M, Iacovidou N, Boutsikou T, Sulaj A, Karapati E, Tsantes AG, Tsantes AE, Valsami S, Iliodromiti Z. Hemostatic Status of Neonates with Perinatal Hypoxia, Studied via NATEM in Cord Blood Samples. CHILDREN (BASEL, SWITZERLAND) 2024; 11:799. [PMID: 39062248 PMCID: PMC11276384 DOI: 10.3390/children11070799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 06/25/2024] [Accepted: 06/28/2024] [Indexed: 07/28/2024]
Abstract
BACKGROUND Perinatal hypoxia may result in coagulation dysfunction. Diminished blood flow or oxygen to the fetus/neonate during the perinatal period can cause bone marrow and liver function impairment, leading to thrombocytopenia, impaired synthesis of clotting and fibrinolytic factors, and increased destruction of platelets in the small blood vessels. The goal of the present study was to evaluate the hemostatic status of newborns with perinatal hypoxia via the non-activated thromboelastometry (NATEM) assay in cord blood samples. METHODS 134 hypoxic neonates born in our maternity unit over a 1.5-year period were enrolled in this observational cohort study, and 189 healthy neonates served as the control group. Participation in the study was voluntary and parents signed informed consent prior to recruitment. Demographic and clinical data were recorded on admission, and the NATEM method was performed on cord blood samples. The following NATEM values were evaluated: clotting time (CT), alpha angle (α-angle), clot formation time (CFT), clot amplitude at 5 and 10 min. (A5, A10), maximum clot firmness (MCF), clot lysis index at 60 min. after CT (LI60), and maximum clot elasticity (MCE). Statistical analysis was conducted utilizing the SAS for Windows 9.4 software platform. RESULTS Neonates with perinatal hypoxia exhibited decreased fibrinolytic potential in comparison to healthy neonates, as indicated by increased LI60, and this difference was statistically significant (LΙ60: 94 (92-96) Vs 93 (91-95), p value = 0.0001). There were no statistically significant differences noted among the remaining NATEM variables. CONCLUSION Our findings indicate decreased fibrinolytic potential in hypoxic neonates in comparison to healthy neonates, suggesting that NATEM could serve as an effective tool for promptly identifying hemostasis dysfunction in this group of neonates.
Collapse
Affiliation(s)
- Marina Tsaousi
- Neonatal Department, Aretaieio Hospital, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece; (M.T.); (R.S.); (T.B.); (A.S.); (E.K.); (Z.I.)
| | - Rozeta Sokou
- Neonatal Department, Aretaieio Hospital, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece; (M.T.); (R.S.); (T.B.); (A.S.); (E.K.); (Z.I.)
| | - Abraham Pouliakis
- 2nd Department of Pathology, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece;
| | - Marianna Politou
- Hematology Laboratory Blood Bank, Aretaieio Hospital, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece; (M.P.); (S.V.)
| | - Nicoletta Iacovidou
- Neonatal Department, Aretaieio Hospital, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece; (M.T.); (R.S.); (T.B.); (A.S.); (E.K.); (Z.I.)
| | - Theodora Boutsikou
- Neonatal Department, Aretaieio Hospital, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece; (M.T.); (R.S.); (T.B.); (A.S.); (E.K.); (Z.I.)
| | - Alma Sulaj
- Neonatal Department, Aretaieio Hospital, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece; (M.T.); (R.S.); (T.B.); (A.S.); (E.K.); (Z.I.)
| | - Eleni Karapati
- Neonatal Department, Aretaieio Hospital, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece; (M.T.); (R.S.); (T.B.); (A.S.); (E.K.); (Z.I.)
| | - Andreas G. Tsantes
- Laboratory of Haematology and Blood Bank Unit, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece; (A.G.T.); (A.E.T.)
| | - Argirios E. Tsantes
- Laboratory of Haematology and Blood Bank Unit, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece; (A.G.T.); (A.E.T.)
| | - Serena Valsami
- Hematology Laboratory Blood Bank, Aretaieio Hospital, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece; (M.P.); (S.V.)
| | - Zoi Iliodromiti
- Neonatal Department, Aretaieio Hospital, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece; (M.T.); (R.S.); (T.B.); (A.S.); (E.K.); (Z.I.)
| |
Collapse
|
5
|
Nakanishi T, Kuragano T. Growing concerns about using hypoxia-inducible factor prolyl hydroxylase inhibitors for the treatment of renal anemia. Clin Kidney J 2024; 17:sfae051. [PMID: 38516524 PMCID: PMC10956400 DOI: 10.1093/ckj/sfae051] [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] [Received: 02/15/2024] [Indexed: 03/23/2024] Open
Abstract
Hypoxia-inducible factor prolyl hydroxylase inhibitors (HIF-PHIs) have emerged as a novel therapeutic class for treating anemia in patients with chronic kidney disease. Small molecule analogs of α-ketoglutarate (AKG), an essential substrate for 2-oxoglutarate-dependent dioxygenases (2-OGDDs), including prolyl hydroxylase domain proteins (PHDs), inhibit PHDs pharmacologically and thereby prevent HIF degradation. HIF stabilization alleviates anemia through several stimulatory effects on erythropoiesis, but it also affects the expression of many anemia-unrelated genes whose protein products exert important functions in vivo. Therefore, the pleiotropic effects of HIF stabilization under normoxic conditions deserve to be examined in more detail. Specifically, we believe that particular attention should be given to epigenetic modifications among the various AKG-based metabolic systems that may be altered by HIF-PHIs. It is noteworthy that AKG has been reported to exert health-protective actions. AKG-based metabolic systems include enzymes associated with the tricarboxylic acid cycle and amino acid metabolism, as well as 2-OGDD-mediated processes, which play important roles in many biological reactions. In this review, we examine the multifaceted effects of HIF-PHIs, encompassing not only their on-target effect of HIF stabilization but also their off-target inhibitory effects on various AKG-based metabolic systems. Furthermore, we examine its potential relevance to cardiovascular complications, based on clinical and animal studies suggesting its involvement in vascular calcification, thrombogenesis and heart failure. In conclusion, although HIF-PHIs offer a promising avenue for anemia treatment in CKD patients, their broader impact on multiple biological systems raises substantial concerns. The intricate interplay between HIF stabilization, AKG competition and cardiovascular complications warrants extensive, long-term investigations to ensure the safety and usefulness of HIF-PHIs in clinical practice.
Collapse
Affiliation(s)
- Takeshi Nakanishi
- Division of Kidney, Dialysis and Cardiology, Department of Internal Medicine, Hyogo Medical University, Nishinomiya, Hyogo, Japan
- Department of Nephrology, Gojinkai Sumiyoshigawa Hospital, Kobe, Hyogo, Japan
| | - Takahiro Kuragano
- Division of Kidney, Dialysis and Cardiology, Department of Internal Medicine, Hyogo Medical University, Nishinomiya, Hyogo, Japan
| |
Collapse
|
6
|
Wada T, Gando S. Phenotypes of Disseminated Intravascular Coagulation. Thromb Haemost 2024; 124:181-191. [PMID: 37657485 PMCID: PMC10890912 DOI: 10.1055/a-2165-1142] [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: 07/30/2023] [Accepted: 08/30/2023] [Indexed: 09/03/2023]
Abstract
Two phenotypes of disseminated intravascular coagulation (DIC) are systematically reviewed. DIC is classified into thrombotic and fibrinolytic phenotypes characterized by thrombosis and hemorrhage, respectively. Major pathology of DIC with thrombotic phenotype is the activation of coagulation, insufficient anticoagulation with endothelial injury, and plasminogen activator inhibitor-1-mediated inhibition of fibrinolysis, leading to microvascular fibrin thrombosis and organ dysfunction. DIC with fibrinolytic phenotype is defined as massive thrombin generation commonly observed in any type of DIC, combined with systemic pathologic hyperfibrinogenolysis caused by underlying disorder that results in severe bleeding due to excessive plasmin formation. Three major pathomechanisms of systemic hyperfibrinogenolysis have been considered: (1) acceleration of tissue-type plasminogen activator (t-PA) release from hypoxic endothelial cells and t-PA-rich storage pools, (2) enhancement of the conversion of plasminogen to plasmin due to specific proteins and receptors that are expressed on cancer cells and endothelial cells, and (3) alternative pathways of fibrinolysis. DIC with fibrinolytic phenotype can be diagnosed by DIC diagnosis followed by the recognition of systemic pathologic hyperfibrin(ogen)olysis. Low fibrinogen levels, high fibrinogen and fibrin degradation products (FDPs), and the FDP/D-dimer ratio are important for the diagnosis of systemic pathologic hyperfibrin(ogen)olysis. Currently, evidence-based treatment strategies for DIC with fibrinolytic phenotypes are lacking. Tranexamic acid appears to be one of the few methods to be effective in the treatment of systemic pathologic hyperfibrin(ogen)olysis. International cooperation for the elucidation of pathomechanisms, establishment of diagnostic criteria, and treatment strategies for DIC with fibrinolytic phenotype are urgent issues in the field of thrombosis and hemostasis.
Collapse
Affiliation(s)
- Takeshi Wada
- Department of Anesthesiology and Critical Care Medicine, Division of Acute and Critical Care Medicine, Hokkaido University Faculty of Medicine, Sapporo, Japan
| | - Satoshi Gando
- Department of Anesthesiology and Critical Care Medicine, Division of Acute and Critical Care Medicine, Hokkaido University Faculty of Medicine, Sapporo, Japan
- Department of Acute and Critical Care Medicine, Sapporo Higashi Tokushukai Hospital, Sapporo, Japan
| |
Collapse
|
7
|
Liu Y, Li X, Chen S, Zhu C, Shi Y, Dang S, Zhang W, Li W. Pan-cancer analysis of SERPINE family genes as biomarkers of cancer prognosis and response to therapy. Front Mol Biosci 2024; 10:1277508. [PMID: 38274096 PMCID: PMC10808646 DOI: 10.3389/fmolb.2023.1277508] [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: 08/14/2023] [Accepted: 12/29/2023] [Indexed: 01/27/2024] Open
Abstract
Background: Serine protease inhibitor E (SERPINE) family genes participate in the tumor growth, cancer cell survival and metastasis. However, the SERPINE family members role in the prognosis and their clinical therapeutic potentials in various human cancer types have not been elaborately explored. Methods: We preliminarily analyzed expression levels and prognostic values of SERPINE family genes, and investigated the correlation between SERPINEs expression and tumor microenvironment (TME), Stemness score, clinical characteristic, immune infiltration, tumor mutational burden (TMB), immune subtype, and drug sensitivity in pan-cancer, which based on updated public databases and integrated some bioinformatics analysis methods. In addition, we conducted the enrichment analysis of SERPINEs from DAVID and KOBAS databases. Results: SERPINE1, SERPINE2, and SERPINE3 expression were upregulated in nine cancers, twelve cancers, and six cancers, respectively. The expression of SERPINE family genes was associated with the prognosis in several cancers from The Cancer Genome Atlas (TCGA). Furthermore, SERPINE family genes expression also had a significant relation to stromal and immune scores, and RNA stemness score and DNA stemness score in pan-cancer. SERPINE1 and SERPINE2 expression significantly increased in tumor advanced stage in colon adenocarcinoma (COAD). Results showed that SERPINE1 and SERPINE2 expression were negatively related with B cells and Monocytes, respectively. SERPINE2 expression had a significantly positive relation with B cells and Macrophages. In terms of TMB, SERPINE1, SERPINE2, and SERPINE3 were found to associated with TMB in seven cancers, fourteen cancers, and four cancers, respectively. Moreover, all SERPINE gene family members were significantly correlated with immune subtypes. SERPINE1 expression had a significantly positive or negative correlation with drug sensitivity. Conclusion: The study indicated the great potential of SERPINE family genes as biomarkers for prognosis and provided valuable strategies for further investigation of SERPINE family genes as potential targets in cancer.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Wei Li
- Department of Cancer Center, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
8
|
Alhelf M, Rashed L, Doss RW, Mohamed SM, Abd Elazeem NA. Long noncoding RNA (taurine upregulated gene 1) and micro RNA-377: emerging players in the development of metabolic syndrome among psoriasis patients. BENI-SUEF UNIVERSITY JOURNAL OF BASIC AND APPLIED SCIENCES 2023; 12:68. [DOI: 10.1186/s43088-023-00404-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 06/29/2023] [Indexed: 09/02/2023] Open
Abstract
Abstract
Background
Psoriasis (PsO) is an immune-mediated dermatosis and systemic inflammatory condition that can affect the skin, joints, and other organs and tissues with a range of comorbidities. The activation of proinflammatory cytokines is the primary cause of the development of skin lesions in PsO. Patients with PsO have a higher risk of developing cardiovascular metabolic comorbidities; among these is the metabolic syndrome (MetS). Particularly, MetS is characterized by abdominal obesity, hypertension, hyperglycemia, and hyperlipidemia, has been linked to PsO. The connection between PsO and MetS is believed to be caused by PsO generating systemic inflammation, which then results in elevated inflammatory adipokines, endothelial dysfunction, and insulin resistance. Micro RNA-377 and long noncoding RNA taurine upregulated 1 (TUG1) are both involved in the control of a variety of inflammatory disorders in humans and can be employed as biomarkers for the diagnosis and prognosis of psoriasis. The aim of the present study is to establish a panel of biomarkers for the early diagnosis of MetS incidence in psoriasis and thereby, reducing its lethal consequences.
Results
In this study, 120 patients: 40 psoriatic patients, 40 psoriatic patients with metabolic syndrome, and 40 healthy subjects were conducted. Expressions of Long noncoding RNA Taurine Upregulated Gene-1 (TUG1), miRNA-377 and Peroxisome Proliferator-Activated Receptor-γ (PPAR-γ) were assessed in tissue lesion by real-time PCR. ELISA technique was carried out for the detection of serum levels of plasminogen activator inhibitor-1 (PAI-1) and transforming growth factor β (TGFβ). Moreover, miRNA-377 expression was significantly elevated with the simultaneous down-regulation of both TUG-1 and PPAR-γ in PsO-MetS group when compared to those of PsO and control groups. Furthermore, PAI-1 and TGFβ levels were higher in PsO-MetS than PsO.
Conclusions
The dysregulated levels of TUG-1, miRNA-377, PPAR-γ, PAI-1, and TGFβ, biomarkers may provide information about their potential role in the emergence of MetS in psoriasis patients.
Collapse
|
9
|
Lv R, Liu X, Zhang Y, Dong N, Wang X, He Y, Yue H, Yin Q. Pathophysiological mechanisms and therapeutic approaches in obstructive sleep apnea syndrome. Signal Transduct Target Ther 2023; 8:218. [PMID: 37230968 DOI: 10.1038/s41392-023-01496-3] [Citation(s) in RCA: 136] [Impact Index Per Article: 68.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 05/09/2023] [Accepted: 05/11/2023] [Indexed: 05/27/2023] Open
Abstract
Obstructive sleep apnea syndrome (OSAS) is a common breathing disorder in sleep in which the airways narrow or collapse during sleep, causing obstructive sleep apnea. The prevalence of OSAS continues to rise worldwide, particularly in middle-aged and elderly individuals. The mechanism of upper airway collapse is incompletely understood but is associated with several factors, including obesity, craniofacial changes, altered muscle function in the upper airway, pharyngeal neuropathy, and fluid shifts to the neck. The main characteristics of OSAS are recurrent pauses in respiration, which lead to intermittent hypoxia (IH) and hypercapnia, accompanied by blood oxygen desaturation and arousal during sleep, which sharply increases the risk of several diseases. This paper first briefly describes the epidemiology, incidence, and pathophysiological mechanisms of OSAS. Next, the alterations in relevant signaling pathways induced by IH are systematically reviewed and discussed. For example, IH can induce gut microbiota (GM) dysbiosis, impair the intestinal barrier, and alter intestinal metabolites. These mechanisms ultimately lead to secondary oxidative stress, systemic inflammation, and sympathetic activation. We then summarize the effects of IH on disease pathogenesis, including cardiocerebrovascular disorders, neurological disorders, metabolic diseases, cancer, reproductive disorders, and COVID-19. Finally, different therapeutic strategies for OSAS caused by different causes are proposed. Multidisciplinary approaches and shared decision-making are necessary for the successful treatment of OSAS in the future, but more randomized controlled trials are needed for further evaluation to define what treatments are best for specific OSAS patients.
Collapse
Affiliation(s)
- Renjun Lv
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, China
| | - Xueying Liu
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China
| | - Yue Zhang
- Department of Geriatrics, the 2nd Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Na Dong
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, China
| | - Xiao Wang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, China
| | - Yao He
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, China
| | - Hongmei Yue
- Department of Pulmonary and Critical Care Medicine, The First Hospital of Lanzhou University, Lanzhou, 730000, China.
| | - Qingqing Yin
- Department of Geriatric Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China.
| |
Collapse
|
10
|
Lai XX, Zhang CP, Wu YX, Yang Y, Zhang MQ, Qin WJ, Wang RX, Shu H. Comparative transcriptome analysis reveals physiological responses in liver tissues of Epinephelus coioides under acute hypoxia stress. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2022; 43:101005. [PMID: 35653833 DOI: 10.1016/j.cbd.2022.101005] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 05/16/2022] [Accepted: 05/16/2022] [Indexed: 06/15/2023]
Abstract
Hypoxia is a common stressor for aquatic animals, including Epinephelus coioides, with a considerable impact on sustainable aquaculture. E. coioides is a widely consumed fish in China owing to its high nutritious value and taste. However, water hypoxia caused by high density culture process has become a great threat to E. coioides culture, and its response to hypoxia stress has not been discussed before. Therefore, the aim of this study was to examine the response of E. coioides to acute hypoxia using transcriptomic techniques. To this end, RNA sequencing was performed on the liver tissues of fish exposed to normoxic and hypoxic conditions for 1 h. The results presented 503 differentially expressed genes (DEGs) in the liver tissue of fish exposed to hypoxic condition compared with those in the normoxic group. Enrichment analysis using the Gene Ontology database showed that the DEGs were mainly enriched for functions related to cell apoptosis signaling pathways, insulin resistance, antioxidant enzymes, and glycolysis/gluconeogenesis signaling pathways. KEGG enrichment analysis showed that HIF-1, PI3K-AKT, IL-17, NF-kappa B, and MAPK signaling pathways were significantly enriched by the DEGs. The DEGs were mainly involved in immune response, inflammatory response, cell apoptosis regulation, energy metabolism, and substance metabolism. Additionally, the hypoxia response in E. coioides was mainly regulated via the PI3K-AKT-HIF-1 signaling axis. Overall, the findings of this study contribute to the understanding of hypoxia stress response in E. coioides, and provides target genes for breeding hypoxia-tolerant Epinephelus spp.
Collapse
Affiliation(s)
- Xing-Xing Lai
- School of Life Sciences, Guangzhou University, Guangzhou 51006, China.
| | - Cui-Ping Zhang
- School of Life Sciences, Guangzhou University, Guangzhou 51006, China
| | - Yu-Xin Wu
- School of Life Sciences, Guangzhou University, Guangzhou 51006, China
| | - Yang Yang
- School of Life Sciences, Sun Yat-sen University, Guangzhou 51006, China
| | - Ming-Qing Zhang
- School of Life Sciences, Guangzhou University, Guangzhou 51006, China
| | - Wei-Jian Qin
- School of Life Sciences, Guangzhou University, Guangzhou 51006, China
| | - Rui-Xuan Wang
- School of Life Sciences and Food Engineering, Hanshan Normal University, Chaozhou 521041, China.
| | - Hu Shu
- School of Life Sciences, Guangzhou University, Guangzhou 51006, China.
| |
Collapse
|
11
|
Badran M, Gozal D. PAI-1: A Major Player in the Vascular Dysfunction in Obstructive Sleep Apnea? Int J Mol Sci 2022; 23:5516. [PMID: 35628326 PMCID: PMC9141273 DOI: 10.3390/ijms23105516] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 05/05/2022] [Accepted: 05/12/2022] [Indexed: 02/04/2023] Open
Abstract
Obstructive sleep apnea is a chronic and prevalent condition that is associated with endothelial dysfunction, atherosclerosis, and imposes excess overall cardiovascular risk and mortality. Despite its high prevalence and the susceptibility of CVD patients to OSA-mediated stressors, OSA is still under-recognized and untreated in cardiovascular practice. Moreover, conventional OSA treatments have yielded either controversial or disappointing results in terms of protection against CVD, prompting the need for the identification of additional mechanisms and associated adjuvant therapies. Plasminogen activator inhibitor-1 (PAI-1), the primary inhibitor of tissue-type plasminogen activator (tPA) and urinary-type plasminogen activator (uPA), is a key regulator of fibrinolysis and cell migration. Indeed, elevated PAI-1 expression is associated with major cardiovascular adverse events that have been attributed to its antifibrinolytic activity. However, extensive evidence indicates that PAI-1 can induce endothelial dysfunction and atherosclerosis through complex interactions within the vasculature in an antifibrinolytic-independent matter. Elevated PAI-1 levels have been reported in OSA patients. However, the impact of PAI-1 on OSA-induced CVD has not been addressed to date. Here, we provide a comprehensive review on the mechanisms by which OSA and its most detrimental perturbation, intermittent hypoxia (IH), can enhance the transcription of PAI-1. We also propose causal pathways by which PAI-1 can promote atherosclerosis in OSA, thereby identifying PAI-1 as a potential therapeutic target in OSA-induced CVD.
Collapse
Affiliation(s)
- Mohammad Badran
- Department of Child Health and Child Health Research Institute, School of Medicine, University of Missouri, 400 N Keene St, Suite 010, Columbia, MO 65201, USA;
| | - David Gozal
- Department of Child Health and Child Health Research Institute, School of Medicine, University of Missouri, 400 N Keene St, Suite 010, Columbia, MO 65201, USA;
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, MO 65201, USA
| |
Collapse
|
12
|
Multiphysics and multiscale modeling of microthrombosis in COVID-19. PLoS Comput Biol 2022; 18:e1009892. [PMID: 35255089 PMCID: PMC8901059 DOI: 10.1371/journal.pcbi.1009892] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 02/02/2022] [Indexed: 12/21/2022] Open
Abstract
Emerging clinical evidence suggests that thrombosis in the microvasculature of patients with Coronavirus disease 2019 (COVID-19) plays an essential role in dictating the disease progression. Because of the infectious nature of SARS-CoV-2, patients’ fresh blood samples are limited to access for in vitro experimental investigations. Herein, we employ a novel multiscale and multiphysics computational framework to perform predictive modeling of the pathological thrombus formation in the microvasculature using data from patients with COVID-19. This framework seamlessly integrates the key components in the process of blood clotting, including hemodynamics, transport of coagulation factors and coagulation kinetics, blood cell mechanics and adhesive dynamics, and thus allows us to quantify the contributions of many prothrombotic factors reported in the literature, such as stasis, the derangement in blood coagulation factor levels and activities, inflammatory responses of endothelial cells and leukocytes to the microthrombus formation in COVID-19. Our simulation results show that among the coagulation factors considered, antithrombin and factor V play more prominent roles in promoting thrombosis. Our simulations also suggest that recruitment of WBCs to the endothelial cells exacerbates thrombogenesis and contributes to the blockage of the blood flow. Additionally, we show that the recent identification of flowing blood cell clusters could be a result of detachment of WBCs from thrombogenic sites, which may serve as a nidus for new clot formation. These findings point to potential targets that should be further evaluated, and prioritized in the anti-thrombotic treatment of patients with COVID-19. Altogether, our computational framework provides a powerful tool for quantitative understanding of the mechanism of pathological thrombus formation and offers insights into new therapeutic approaches for treating COVID-19 associated thrombosis. Emerging clinical evidence suggests that thrombosis in the microvasculature of patients with Coronavirus disease 2019 (COVID-19) plays an essential role in dictating the disease progression. We employ a novel multiphysics and multiscale computational framework to investigate the underlying mechanism of the pathological formation of microthrombi and circulating cell clusters in COVID-19. We quantify the contributions of many prothrombotic factors reported in the literature, such as stasis, the derangement in blood coagulation factor levels and activities, inflammatory responses of endothelial cells and leukocytes to the microthrombus formation in COVID-19, through which we identify the potential targets that should be further evaluated, and prioritized in the anti-thrombotic treatment of patients with COVID-19.
Collapse
|
13
|
Rusu I, Turlacu M, Micheu MM. Acute myocardial injury in patients with COVID-19: Possible mechanisms and clinical implications. World J Clin Cases 2022; 10:762-776. [PMID: 35127893 PMCID: PMC8790431 DOI: 10.12998/wjcc.v10.i3.762] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 06/15/2021] [Accepted: 12/21/2021] [Indexed: 02/06/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 infection affects not only the lungs, but also the cardiovascular system, having a major impact on patients' outcomes. Myocardial injury (MI) occurs in the context of coronavirus infectious disease 2019 (COVID-19) and is associated with a higher risk of severe clinical outcome and mortality. COVID-19-related MI can have various clinical manifestations, of which the main ones are myocarditis, stress cardiomyopathy, acute coronary syndrome, and pulmonary embolism. The exact mechanisms of how MI occurs in these patients are not yet fully known. Direct injury, through direct viral myocardial invasion, and indirect injury, through interaction with angiotensin I converting enzyme 2, increased inflammation, and thrombocyte and endothelial dysfunction, could be involved in acute MI in patients with COVID-19. A better understanding of these multiple potential mechanisms may help to develop new targeted therapeutic strategies. The purpose of this review is to provide the current understanding of the potential mechanisms involved in MI induced by COVID-19 and to discuss the current progress in the therapeutic strategies.
Collapse
Affiliation(s)
- Iulia Rusu
- Department of Cardiology, Clinical Emergency Hospital of Bucharest, Bucharest 014461, Romania
| | - Malina Turlacu
- Department of Cardiology, Clinical Emergency Hospital of Bucharest, Bucharest 014461, Romania
| | - Miruna Mihaela Micheu
- Department of Cardiology, Clinical Emergency Hospital of Bucharest, Bucharest 014461, Romania
| |
Collapse
|
14
|
Sasidharakurup H, Kumar G, Nair B, Diwakar S. Mathematical Modeling of Severe Acute Respiratory Syndrome Coronavirus 2 Infection Network with Cytokine Storm, Oxidative Stress, Thrombosis, Insulin Resistance, and Nitric Oxide Pathways. OMICS : A JOURNAL OF INTEGRATIVE BIOLOGY 2021; 25:770-781. [PMID: 34807729 DOI: 10.1089/omi.2021.0155] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is a systemic disease affecting not only the lungs but also multiple organ systems. Clinical studies implicate that SARS-CoV-2 infection causes imbalance of cellular homeostasis and immune response that trigger cytokine storm, oxidative stress, thrombosis, and insulin resistance. Mathematical modeling can offer in-depth understanding of the SARS-CoV-2 infection and illuminate how subcellular mechanisms and feedback loops underpin disease progression and multiorgan failure. We report here a mathematical model of SARS-CoV-2 infection pathway network with cytokine storm, oxidative stress, thrombosis, insulin resistance, and nitric oxide (NO) pathways. The biochemical systems theory model shows autocrine loops with positive feedback enabling excessive immune response, cytokines, transcription factors, and interferons, which can imbalance homeostasis of the system. The simulations suggest that changes in immune response led to uncontrolled release of cytokines and chemokines, including interleukin (IL)-1β, IL-6, and tumor necrosis factor α (TNFα), and affect insulin, coagulation, and NO signaling pathways. Increased production of NETs (neutrophil extracellular traps), thrombin, PAI-1 (plasminogen activator inhibitor-1), and other procoagulant factors led to thrombosis. By analyzing complex biochemical reactions, this model forecasts the key intermediates, potential biomarkers, and risk factors at different stages of COVID-19. These insights can be useful for drug discovery and development, as well as precision treatment of multiorgan implications of COVID-19 as seen in systems medicine.
Collapse
Affiliation(s)
- Hemalatha Sasidharakurup
- Amrita Mind Brain Center and Amrita Vishwa Vidyapeetham, Kollam, India
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam, India
| | - Geetha Kumar
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam, India
- Tata Institute for Genetics and Society, Kodigehalli, Bengaluru, India
| | - Bipin Nair
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam, India
- Tata Institute for Genetics and Society, Kodigehalli, Bengaluru, India
| | - Shyam Diwakar
- Amrita Mind Brain Center and Amrita Vishwa Vidyapeetham, Kollam, India
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam, India
- School of Engineering, Amrita Vishwa Vidyapeetham, Kollam, India
| |
Collapse
|
15
|
Sadia K, Ashraf MZ, Mishra A. Therapeutic Role of Sirtuins Targeting Unfolded Protein Response, Coagulation, and Inflammation in Hypoxia-Induced Thrombosis. Front Physiol 2021; 12:733453. [PMID: 34803727 PMCID: PMC8602789 DOI: 10.3389/fphys.2021.733453] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 10/19/2021] [Indexed: 12/23/2022] Open
Abstract
Thrombosis remains one of the leading causes of morbidity and mortality across the world. Many pathological milieus in the body resulting from multiple risk factors escort thrombosis. Hypoxic condition is one such risk factor that disturbs the integrity of endothelial cells to cause an imbalance between anticoagulant and procoagulant proteins. Hypoxia generates reactive oxygen species (ROS) and triggers inflammatory pathways to augment the coagulation cascade. Hypoxia in cells also activates unfolded protein response (UPR) signaling pathways in the endoplasmic reticulum (ER), which tries to restore ER homeostasis and function. But the sustained UPR linked with inflammation, generation of ROS and apoptosis stimulates the severity of thrombosis in the body. Sirtuins, a group of seven proteins, play a vast role in bringing down inflammation, oxidative and ER stress and apoptosis. As a result, sirtuins might provide a therapeutic approach towards the treatment or prevention of hypoxia-induced thrombosis. Sirtuins modulate hypoxia-inducible factors (HIFs) and counteract ER stress-induced apoptosis by attenuating protein kinase RNA-like endoplasmic reticulum kinase (PERK)/Eukaryotic translation initiation factor 2α (eIF2α) pathway activation. It prevents ER-stress mediated inflammation by targeting X-Box Binding Protein 1 (XBP1) and inhibiting nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κβ) signaling through deacetylation. Sirtuins also obstruct nucleotide-binding domain, leucine-rich-containing family, pyrin domain containing 3 (NLRP3) inflammasome activation to reduce the expression of several pro-inflammatory molecules. It protects cells against oxidative stress by targeting nuclear factor erythroid 2-related factor 2 (Nrf2), glutathione (GSH), forkhead box O3 (FOXO3), superoxide dismutase (SOD), catalase (CAT), peroxisome proliferator-activated receptor gamma coactivator 1-α (PGC-1α), glucose-6-phosphate dehydrogenase (G6PD), phosphoglucomutase-2 (PGAM2), and NF-κB, to name few. This review, thus, discusses the potential role of sirtuins as a new treatment for hypoxia-induced thrombosis that involves an intersection of UPR and inflammatory pathways in its pathological manifestation.
Collapse
Affiliation(s)
- Khan Sadia
- Department of Biotechnology, Jamia Millia Islamia, New Delhi, India
| | | | - Aastha Mishra
- Council of Scientific and Industrial Research-Institute of Genomics and Integrative Biology, New Delhi, India
| |
Collapse
|
16
|
Zhao J, Xu Y, Xie J, Liu J, Zhang R, Yan X. Roxadustat Does Not Affect Platelet Production, Activation, and Thrombosis Formation. Arterioscler Thromb Vasc Biol 2021; 41:2523-2537. [PMID: 34348487 PMCID: PMC8454499 DOI: 10.1161/atvbaha.121.316495] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 07/26/2021] [Indexed: 11/16/2022]
Abstract
Objective: Roxadustat is a new medication for the treatment of renal anemia. EPO (erythropoietin)-the current treatment standard-has been reported to enhance platelet activation and production. However, to date, the effect of roxadustat on platelets is unclear. To address this deficiency, herein, we have evaluated the effect of roxadustat on platelet production and function. Approach and Results: We performed several mouse platelet functional assays in the presence/absence of in vitro and in vivo roxadustat treatment. Both healthy and 5/6 nephrectomized mice were utilized. The effect of roxadustat on platelet function of healthy volunteers and chronic kidney disease patients was also evaluated. For platelet production, megakaryocyte maturation and proplatelet formation were assayed in vitro. Peripheral platelet and bone marrow megakaryocyte counts were also determined. We found that roxadustat could not stimulate washed platelets directly, and platelet aggregation, spreading, clot retraction, and P-selectin/JON/A exposure were similar with or without in vitro or in vivo roxadustat treatment among both healthy and 5/6 nephrectomized mice. In vivo mouse thrombosis models were additionally performed, and no differences were detected between the vehicle and roxadustat treatment groups. EPO, which was considered a positive control in the present study, promoted platelet function and production as reported previously. Megakaryocyte maturation and proplatelet formation were also not significantly different between control mice and those treated with roxadustat. After receiving roxadustat for 14 days, no difference in the peripheral platelet count was observed in the mice. Conclusions: Administration of roxadustat has no significant impact on platelet production and function.
Collapse
Affiliation(s)
- Jiaxin Zhao
- Department of Cardiology, Ruijin Hospital (J.Z., R.Z., X.Y.), Shanghai Jiao Tong University School of Medicine, China
| | - Yanyan Xu
- Department of Biochemistry and Molecular Cell Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education (Y.X., J.L.), Shanghai Jiao Tong University School of Medicine, China
| | - Jingyuan Xie
- Department of Nephrology, Ruijin Hospital (J.X.), Shanghai Jiao Tong University School of Medicine, China
| | - Junling Liu
- Department of Biochemistry and Molecular Cell Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education (Y.X., J.L.), Shanghai Jiao Tong University School of Medicine, China
| | - Ruiyan Zhang
- Department of Cardiology, Ruijin Hospital (J.Z., R.Z., X.Y.), Shanghai Jiao Tong University School of Medicine, China
| | - Xiaoxiang Yan
- Department of Cardiology, Ruijin Hospital (J.Z., R.Z., X.Y.), Shanghai Jiao Tong University School of Medicine, China
| |
Collapse
|
17
|
An Integrated Approach of the Potential Underlying Molecular Mechanistic Paradigms of SARS-CoV-2-Mediated Coagulopathy. Indian J Clin Biochem 2021; 36:387-403. [PMID: 33875909 PMCID: PMC8047580 DOI: 10.1007/s12291-021-00972-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Accepted: 04/03/2021] [Indexed: 02/06/2023]
Abstract
Coronavirus disease 2019 (Covid-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a pandemic disease which has affected more than 6.2 million people globally, with numbers mounting considerably daily. However, till date, no specific treatment modalities are available for Covid-19 and also not much information is known about this disease. Recent studies have revealed that SARS-CoV-2 infection is associated with the generation of thrombosis and coagulopathy. Fundamentally, it has been believed that a diverse array of signalling pathways might be responsible for the activation of coagulation cascade during SARS-CoV-2 infection. Henceforth, a detailed understanding of these probable underlying molecular mechanistic pathways causing thrombosis in Covid-19 disease deserves an urgent exploration. Therefore, in this review, the hypothetical crosstalk between distinct signalling pathways including apoptosis, inflammation, hypoxia and angiogenesis attributable for the commencement of thrombotic events during SARS-CoV-2 infection has been addressed which might further unravel promising therapeutic targets in Covid-19 disease.
Collapse
|
18
|
Chauhan A, Kaur R, Chakrbarti P, Pal A. " Silent Hypoxemia" Leads to Vicious Cycle of Infection, Coagulopathy and Cytokine Storm in COVID-19: Can Prophylactic Oxygen Therapy Prevent It? Indian J Clin Biochem 2021; 36:468-472. [PMID: 33746377 PMCID: PMC7958103 DOI: 10.1007/s12291-021-00967-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 02/22/2021] [Indexed: 01/06/2023]
Abstract
Humankind is facing its worst pandemic of the twenty-first century, due to infection of a novel coronavirus named as SARS-CoV2, started from Wuhan in China. Till now, 15 million people are infected, causing more than 600,000 deaths. The disease, commonly known as, COVID-19, was initially thought to be associated with ARDS only, but later on revealed to have many unexplained and atypical clinical features like coagulopathy and cytokinemia, leading to multi-organ involvements. The patients also suffer from 'Silent Hypoxemia', where there is no immediate respiratory signs and symptoms even though alarmingly low SpO2 level. We hypothesize that this covert hypoxemia may lead to molecular changes exacerbating coagulopathy and cytokine storm in COVID19 patients, which again, in turn, causes a vicious cycle of more hypoxemia/hypoxia and progression of the infection to more severe stages through HIF-1α dependent pathway. Although molecular mechanisms are yet to be substantiated by scientific evidence, hypoxemia remains an independent worsening factor in serious COVID 19 patients. Keeping all in mind, we propose that even in the early and asymptomatic cases, prophylactic oxygen therapy to be initiated to break the vicious cycle and to reduce the mortality in COVID 19 to save precious human lives.
Collapse
Affiliation(s)
- Anshika Chauhan
- Department of Biochemistry, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Rajandeep Kaur
- Department of Biochemistry, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | | | - Arnab Pal
- Department of Biochemistry, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
19
|
Bikov A, Meszaros M, Schwarz EI. Coagulation and Fibrinolysis in Obstructive Sleep Apnoea. Int J Mol Sci 2021; 22:ijms22062834. [PMID: 33799528 PMCID: PMC8000922 DOI: 10.3390/ijms22062834] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 03/07/2021] [Accepted: 03/08/2021] [Indexed: 12/29/2022] Open
Abstract
Obstructive sleep apnoea (OSA) is a common disease which is characterised by repetitive collapse of the upper airways during sleep resulting in chronic intermittent hypoxaemia and frequent microarousals, consequently leading to sympathetic overflow, enhanced oxidative stress, systemic inflammation, and metabolic disturbances. OSA is associated with increased risk for cardiovascular morbidity and mortality, and accelerated coagulation, platelet activation, and impaired fibrinolysis serve the link between OSA and cardiovascular disease. In this article we briefly describe physiological coagulation and fibrinolysis focusing on processes which could be altered in OSA. Then, we discuss how OSA-associated disturbances, such as hypoxaemia, sympathetic system activation, and systemic inflammation, affect these processes. Finally, we critically review the literature on OSA-related changes in markers of coagulation and fibrinolysis, discuss potential reasons for discrepancies, and comment on the clinical implications and future research needs.
Collapse
Affiliation(s)
- Andras Bikov
- North West Lung Centre, Manchester University NHS Foundation Trust, Manchester M23 9LT, UK
- Division of Infection, Immunity and Respiratory Medicine, University of Manchester, Manchester M13 9MT, UK
- Correspondence: ; Tel.: +44-161-291-2493; Fax: +44-161-291-5730
| | - Martina Meszaros
- Department of Pulmonology, Semmelweis University, 1083 Budapest, Hungary;
- Department of Pulmonology and Sleep Disorders Centre, University Hospital Zurich, 8006 Zurich, Switzerland;
| | - Esther Irene Schwarz
- Department of Pulmonology and Sleep Disorders Centre, University Hospital Zurich, 8006 Zurich, Switzerland;
- Centre of Competence Sleep & Health Zurich, University of Zurich, 8091 Zurich, Switzerland
| |
Collapse
|
20
|
PAI-1, the Plasminogen System, and Skeletal Muscle. Int J Mol Sci 2020; 21:ijms21197066. [PMID: 32993026 PMCID: PMC7582753 DOI: 10.3390/ijms21197066] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 09/21/2020] [Accepted: 09/23/2020] [Indexed: 12/14/2022] Open
Abstract
The plasminogen system is a critical proteolytic system responsible for the remodeling of the extracellular matrix (ECM). The master regulator of the plasminogen system, plasminogen activator inhibitor-1 (PAI-1), has been implicated for its role in exacerbating various disease states not only through the accumulation of ECM (i.e., fibrosis) but also its role in altering cell fate/behaviour. Examination of PAI-1 has extended through various tissues and cell-types with recent investigations showing its presence in skeletal muscle. In skeletal muscle, the role of this protein has been implicated throughout the regeneration process, and in skeletal muscle pathologies (muscular dystrophy, diabetes, and aging-driven pathology). Needless to say, the complete function of this protein in skeletal muscle has yet to be fully elucidated. Given the importance of skeletal muscle in maintaining overall health and quality of life, it is critical to understand the alterations—particularly in PAI-1—that occur to negatively impact this organ. Thus, we provide a comprehensive review of the importance of PAI-1 in skeletal muscle health and function. We aim to shed light on the relevance of this protein in skeletal muscle and propose potential therapeutic approaches to aid in the maintenance of skeletal muscle health.
Collapse
|
21
|
Porembskaya O, Toropova Y, Tomson V, Lobastov K, Laberko L, Kravchuk V, Saiganov S, Brill A. Pulmonary Artery Thrombosis: A Diagnosis That Strives for Its Independence. Int J Mol Sci 2020; 21:ijms21145086. [PMID: 32708482 PMCID: PMC7404175 DOI: 10.3390/ijms21145086] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 07/15/2020] [Accepted: 07/16/2020] [Indexed: 12/13/2022] Open
Abstract
According to a widespread theory, thrombotic masses are not formed in the pulmonary artery (PA) but result from migration of blood clots from the venous system. This concept has prevailed in clinical practice for more than a century. However, a new technologic era has brought forth more diagnostic possibilities, and it has been shown that thrombotic masses in the PA could, in many cases, be found without any obvious source of emboli. Chronic obstructive pulmonary disease, asthma, sickle cell anemia, emergency and elective surgery, viral pneumonia, and other conditions could be complicated by PA thrombosis development without concomitant deep vein thrombosis (DVT). Different pathologies have different causes for local PA thrombotic process. As evidenced by experimental results and clinical observations, endothelial and platelet activation are the crucial mechanisms of this process. Endothelial dysfunction can impair antithrombotic function of the arterial wall through downregulation of endothelial nitric oxide synthase (eNOS) or via stimulation of adhesion receptor expression. Hypoxia, proinflammatory cytokines, or genetic mutations may underlie the procoagulant phenotype of the PA endothelium. Both endotheliocytes and platelets could be activated by protease mediated receptor (PAR)- and receptors for advanced glycation end (RAGE)-dependent mechanisms. Hypoxia, in particular induced by high altitudes, could play a role in thrombotic complications as a trigger of platelet activity. In this review, we discuss potential mechanisms of PA thrombosis in situ.
Collapse
Affiliation(s)
- Olga Porembskaya
- Mechnikov North-Western State Medical University, Saint Petersburg 191015, Russia; (V.K.); (S.S.)
- Institute of Experimental Medicine, Saint Petersburg 197376, Russia
- Correspondence: (O.P.); (A.B.); Tel.: +7-92-1310-6629 (O.P.); Tel.: +44-12-1415-8679 (A.B.)
| | - Yana Toropova
- Institute of Experimental Medicine, Almazov National Medical Research Center, Saint Petersburg 197341, Russia;
| | | | - Kirill Lobastov
- Pirogov Russian National Research Medical University, Moscow 117997, Russia; (K.L.); (L.L.)
| | - Leonid Laberko
- Pirogov Russian National Research Medical University, Moscow 117997, Russia; (K.L.); (L.L.)
| | - Viacheslav Kravchuk
- Mechnikov North-Western State Medical University, Saint Petersburg 191015, Russia; (V.K.); (S.S.)
| | - Sergey Saiganov
- Mechnikov North-Western State Medical University, Saint Petersburg 191015, Russia; (V.K.); (S.S.)
| | - Alexander Brill
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham B152TT, UK
- Department of Pathophysiology, Sechenov First Moscow State Medical University (Sechenov University), Moscow 119991, Russia
- Correspondence: (O.P.); (A.B.); Tel.: +7-92-1310-6629 (O.P.); Tel.: +44-12-1415-8679 (A.B.)
| |
Collapse
|
22
|
Vidali S, Morosetti D, Cossu E, Luisi MLE, Pancani S, Semeraro V, Consales G. D-dimer as an indicator of prognosis in SARS-CoV-2 infection: a systematic review. ERJ Open Res 2020; 6:00260-2020. [PMID: 32685436 PMCID: PMC7357271 DOI: 10.1183/23120541.00260-2020] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 05/18/2020] [Indexed: 02/07/2023] Open
Abstract
Background Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) stimulates pro-thrombotic changes. This, combined with its tropism for endothelium and lung structures, may explain its association with thrombotic events, reduction of pulmonary gas exchange, acute respiratory distress syndrome (ARDS) and a composite end-point (intensive care unit, invasive ventilation, death). This study aims to highlight the correlation between elevated D-dimer (an indirect thrombosis marker) and the increased rate of poor prognosis-associated conditions, and to introduce D-dimer-labelled anticoagulant administration as a potentially useful tool to prevent complications and positively influence coronavirus disease 2019 (COVID-19) course. Methods An online database search (PubMed, Google Scholar, Scopus, Web of Science and Cochrane) was performed between 13 March and 10 April 2020. The most relevant keywords were "D-dimer", "SARS-CoV-2", "COVID-19", "thrombosis" and "ARDS". Selection was independently conducted by three reviewers. References and previews of accepted articles were evaluated. Data inclusion/extraction inaccuracy was limited by the work of three reviewers. Selection bias reduction was addressed by thoughtfully designing the search protocol. Quality assessment was performed with the Newcastle-Ottawa Scale. The systematic review protocol was not registered because we anticipated the very limited available evidence on the topic and due to the urgency of the study. Results 16 studies were evaluated. Good-quality criteria were reached in 13 out of 16 studies. D-dimer was increased and significantly higher in COVID-19 patients compared with healthy controls, in COVID-19 patients with severe disease or a composite end-point compared with non-severe disease, in ARDS compared with non-ARDS patients and in deceased ARDS patients compared with ARDS patients who survived (all p<0.001). COVID-19 patients treated with anticoagulants demonstrated lower mortality compared with those not treated (p=0.017). Conclusions Correlations exist between COVID-19 infection, severe elevation of D-dimer levels, and increase in the rate of complications and composite end-point. The appropriateness of early and continuous D-dimer monitoring and labelled anticoagulation as management tools for COVID-19 disease deserves accurate investigation, to prevent complications and reduce interventions.
Collapse
Affiliation(s)
- Sofia Vidali
- Diagnostic and Interventional Imaging, University Hospital Policlinico Tor Vergata, Rome, Italy
| | - Daniele Morosetti
- Diagnostic and Interventional Imaging, University Hospital Policlinico Tor Vergata, Rome, Italy
| | - Elsa Cossu
- Diagnostic and Interventional Imaging, University Hospital Policlinico Tor Vergata, Rome, Italy
| | | | | | - Vittorio Semeraro
- Diagnostic and Interventional Imaging, Santissima Annunziata Hospital, Taranto, Italy
| | - Guglielmo Consales
- Anesthesiology and Resuscitation, Santo Stefano Hospital, Prato, Italy.,Anesthesiology and Resuscitation, Azienda USL Toscana Centro, Florence, Italy
| |
Collapse
|
23
|
Rodríguez-Lorenzo S, Ferreira Francisco DM, Vos R, van Het Hof B, Rijnsburger M, Schroten H, Ishikawa H, Beaino W, Bruggmann R, Kooij G, de Vries HE. Altered secretory and neuroprotective function of the choroid plexus in progressive multiple sclerosis. Acta Neuropathol Commun 2020; 8:35. [PMID: 32192527 PMCID: PMC7083003 DOI: 10.1186/s40478-020-00903-y] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 02/29/2020] [Indexed: 12/13/2022] Open
Abstract
The choroid plexus (CP) is a key regulator of the central nervous system (CNS) homeostasis through its secretory, immunological and barrier properties. Accumulating evidence suggests that the CP plays a pivotal role in the pathogenesis of multiple sclerosis (MS), but the underlying mechanisms remain largely elusive. To get a comprehensive view on the role of the CP in MS, we studied transcriptomic alterations of the human CP in progressive MS and non-neurological disease controls using RNA sequencing. We identified 17 genes with significantly higher expression in progressive MS patients relative to that in controls. Among them is the newly described long non-coding RNA HIF1A-AS3. Next to that, we uncovered disease-affected pathways related to hypoxia, secretion and neuroprotection, while only subtle immunological and no barrier alterations were observed. In an ex vivo CP explant model, a subset of the upregulated genes responded in a similar way to hypoxic conditions. Our results suggest a deregulation of the Hypoxia-Inducible Factor (HIF)-1 pathway in progressive MS CP. Importantly, cerebrospinal fluid levels of the hypoxia-responsive secreted peptide PAI-1 were higher in MS patients with high disability relative to those with low disability. These findings provide for the first time a complete overview of the CP transcriptome in health and disease, and suggest that the CP environment becomes hypoxic in progressive MS patients, highlighting the altered secretory and neuroprotective properties of the CP under neuropathological conditions. Together, these findings provide novel insights to target the CP and promote the secretion of neuroprotective factors into the CNS of progressive MS patients.
Collapse
Affiliation(s)
- Sabela Rodríguez-Lorenzo
- Department of Molecular Cell Biology and Immunology, Amsterdam Neuroscience, MS Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, de Boelelaan 1117, 1007 MB, Amsterdam, Netherlands
| | | | - Ricardo Vos
- Department of Radiology & Nuclear Medicine, Amsterdam UMC, Amsterdam, the Netherlands
| | - Bert van Het Hof
- Department of Molecular Cell Biology and Immunology, Amsterdam Neuroscience, MS Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, de Boelelaan 1117, 1007 MB, Amsterdam, Netherlands
| | - Merel Rijnsburger
- Department of Molecular Cell Biology and Immunology, Amsterdam Neuroscience, MS Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, de Boelelaan 1117, 1007 MB, Amsterdam, Netherlands
| | - Horst Schroten
- Pediatric Infectious Diseases, University Children's Hospital Manheim, Medical Faculty Manheim, Heidelberg University, Manheim, Germany
| | - Hiroshi Ishikawa
- Laboratory of Clinical Regenerative Medicine, Department of Neurosurgery, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Wissam Beaino
- Department of Radiology & Nuclear Medicine, Amsterdam UMC, Amsterdam, the Netherlands
| | - Rémy Bruggmann
- Interfaculty Bioinformatics Unit and Swiss Institute of Bioinformatics, University of Bern, Bern, Switzerland
| | - Gijs Kooij
- Department of Molecular Cell Biology and Immunology, Amsterdam Neuroscience, MS Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, de Boelelaan 1117, 1007 MB, Amsterdam, Netherlands
| | - Helga E de Vries
- Department of Molecular Cell Biology and Immunology, Amsterdam Neuroscience, MS Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, de Boelelaan 1117, 1007 MB, Amsterdam, Netherlands.
- Medical Biochemistry, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam, 1105 AZ, the Netherlands.
| |
Collapse
|
24
|
Gupta N, Zhao YY, Evans CE. The stimulation of thrombosis by hypoxia. Thromb Res 2019; 181:77-83. [DOI: 10.1016/j.thromres.2019.07.013] [Citation(s) in RCA: 372] [Impact Index Per Article: 62.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 06/19/2019] [Accepted: 07/14/2019] [Indexed: 11/28/2022]
|
25
|
Evans CE. Hypoxia and HIF activation as a possible link between sepsis and thrombosis. Thromb J 2019; 17:16. [PMID: 31423111 PMCID: PMC6693167 DOI: 10.1186/s12959-019-0205-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 07/23/2019] [Indexed: 01/01/2023] Open
Abstract
Risk factors for thrombosis include hypoxia and sepsis, but the mechanisms that control sepsis-induced thrombus formation are incompletely understood. A recent article published in Thrombosis Journal: (i) reviews the role of endothelial cells in the pathogenesis of sepsis-associated microthrombosis; (ii) describes a novel ‘two-path unifying theory’ of hemostatic discorders; and (iii) refers to hypoxia as a consequence of microthrombus formation in sepsis patients. The current article adds to this review by describing how sepsis and thrombus formation could be linked through hypoxia and activation of hypoxia-inducible transcription factors (HIFs). In other words, hypoxia and HIF activation may be a cause as well as a consequence of thrombosis in sepsis patients. While microthrombosis reduces microvascular blood flow causing local hypoxia and tissue ischemia, sepsis-induced increases in HIF1 activation could conversely increase the expression of coagulant factors and integrins that promote thrombus formation, and stimulate the formation of pro-thrombotic neutrophil extracellular traps. A better understanding of the role of cell-specific HIFs in thrombus formation could lead to the development of novel prophylactic therapies for individuals at risk of thrombosis.
Collapse
Affiliation(s)
- Colin E Evans
- 1Program for Lung and Vascular Biology, Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL USA.,2Department of Pediatrics, Division of Critical Care, Northwestern University Feinberg School of Medicine, Chicago, IL USA
| |
Collapse
|
26
|
Sato S, Basse AL, Schönke M, Chen S, Samad M, Altıntaş A, Laker RC, Dalbram E, Barrès R, Baldi P, Treebak JT, Zierath JR, Sassone-Corsi P. Time of Exercise Specifies the Impact on Muscle Metabolic Pathways and Systemic Energy Homeostasis. Cell Metab 2019; 30:92-110.e4. [PMID: 31006592 DOI: 10.1016/j.cmet.2019.03.013] [Citation(s) in RCA: 184] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 02/06/2019] [Accepted: 03/26/2019] [Indexed: 12/11/2022]
Abstract
While the timing of food intake is important, it is unclear whether the effects of exercise on energy metabolism are restricted to unique time windows. As circadian regulation is key to controlling metabolism, understanding the impact of exercise performed at different times of the day is relevant for physiology and homeostasis. Using high-throughput transcriptomic and metabolomic approaches, we identify distinct responses of metabolic oscillations that characterize exercise in either the early rest phase or the early active phase in mice. Notably, glycolytic activation is specific to exercise at the active phase. At the molecular level, HIF1α, a central regulator of glycolysis during hypoxia, is selectively activated in a time-dependent manner upon exercise, resulting in carbohydrate exhaustion, usage of alternative energy sources, and adaptation of systemic energy expenditure. Our findings demonstrate that the time of day is a critical factor to amplify the beneficial impact of exercise on both metabolic pathways within skeletal muscle and systemic energy homeostasis.
Collapse
Affiliation(s)
- Shogo Sato
- Center for Epigenetics and Metabolism, INSERM U1233, Department of Biological Chemistry, School of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Astrid Linde Basse
- Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Integrative Physiology, University of Copenhagen, Copenhagen, Denmark
| | - Milena Schönke
- Department of Molecular Medicine and Surgery, Integrative Physiology, Karolinska Institutet, Stockholm, Sweden
| | - Siwei Chen
- Institute for Genomics and Bioinformatics, University of California, Irvine, Irvine, CA, USA
| | - Muntaha Samad
- Institute for Genomics and Bioinformatics, University of California, Irvine, Irvine, CA, USA
| | - Ali Altıntaş
- Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Integrative Physiology, University of Copenhagen, Copenhagen, Denmark
| | - Rhianna C Laker
- Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Integrative Physiology, University of Copenhagen, Copenhagen, Denmark
| | - Emilie Dalbram
- Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Integrative Physiology, University of Copenhagen, Copenhagen, Denmark
| | - Romain Barrès
- Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Integrative Physiology, University of Copenhagen, Copenhagen, Denmark
| | - Pierre Baldi
- Institute for Genomics and Bioinformatics, University of California, Irvine, Irvine, CA, USA
| | - Jonas T Treebak
- Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Integrative Physiology, University of Copenhagen, Copenhagen, Denmark
| | - Juleen R Zierath
- Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Integrative Physiology, University of Copenhagen, Copenhagen, Denmark; Department of Molecular Medicine and Surgery, Integrative Physiology, Karolinska Institutet, Stockholm, Sweden; Department of Physiology and Pharmacology, Integrative Physiology, Karolinska Institutet, Stockholm, Sweden
| | - Paolo Sassone-Corsi
- Center for Epigenetics and Metabolism, INSERM U1233, Department of Biological Chemistry, School of Medicine, University of California, Irvine, Irvine, CA, USA.
| |
Collapse
|
27
|
Hyperglycaemia cause vascular inflammation through advanced glycation end products/early growth response-1 axis in gestational diabetes mellitus. Mol Cell Biochem 2019; 456:179-190. [DOI: 10.1007/s11010-019-03503-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 02/01/2019] [Indexed: 12/27/2022]
|
28
|
Lin EPY, Hsiao TH, Lu JY, Wong SH, Lu TP, Peck K, Takahashi T, Yang PC. Translating Gene Signatures Into a Pathologic Feature: Tumor Necrosis Predicts Disease Relapse in Operable and Stage I Lung Adenocarcinoma. JCO Precis Oncol 2018; 2:1-13. [DOI: 10.1200/po.18.00043] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Purpose The high 5-year disease relapse rate in patients with stage I lung adenocarcinoma indicates the need for additional risk stratification parameters. This study assessed whether gene signatures translate into a pathologic feature for better disease stratification. Materials and Methods The mutual interdependence and risk stratification power of three gene signatures, cell cycle, hypoxia, and mammalian target of rapamycin (mTOR), were investigated in nine cohorts of patients with lung adenocarcinoma and five cohorts of patients with lung squamous cell carcinoma. The translation from gene signatures to a pathologic feature, tumor necrosis, was validated in The Cancer Genome Atlas lung adenocarcinoma cohort. The translation of signature score to pathway activity was further investigated by integrative analyses using The Cancer Genome Atlas and The Cancer Protein Atlas lung adenocarcinoma data sets. Results The results showed that the three gene signatures were mutually interdependent in lung adenocarcinoma but not in lung squamous cell carcinoma. The signature activities were higher in necrosis-positive tumors than in necrosis-negative tumors. The signature score correlated with the expression level of the representative protein that implicated the activity of each pathway. These signatures stratified patients with operable and stage I lung adenocarcinomas into different risk groups independent of age and stage. Furthermore, the signatures translated to a pathologic feature, tumor necrosis, which predicted shorter overall and relapse-free survival in patients with operable and stage I lung adenocarcinomas. Conclusion This study showed that gene signatures could translate into a pathologic feature, tumor necrosis, with risk stratification ability in patients with operable and stage I lung adenocarcinomas.
Collapse
Affiliation(s)
- Emily Pei-Ying Lin
- Emily Pei-Ying Lin, Tzu-Hung Hsiao, and Pan-Chyr Yang, National Taiwan University Hospital; Tzu-Pin Lu, National Taiwan University; Emily Pei-Ying Lin, Tzu-Hung Hsiao, Jo-Yang Lu, Siao-Han Wong, Konan Peck, and Pan-Chyr Yang, Academia Sinica, Taipei; Tzu-Hung Hsiao, Taichung Veterans General Hospital, Taichung, Taiwan; and Takashi Takahashi, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tzu-Hung Hsiao
- Emily Pei-Ying Lin, Tzu-Hung Hsiao, and Pan-Chyr Yang, National Taiwan University Hospital; Tzu-Pin Lu, National Taiwan University; Emily Pei-Ying Lin, Tzu-Hung Hsiao, Jo-Yang Lu, Siao-Han Wong, Konan Peck, and Pan-Chyr Yang, Academia Sinica, Taipei; Tzu-Hung Hsiao, Taichung Veterans General Hospital, Taichung, Taiwan; and Takashi Takahashi, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Jo-yang Lu
- Emily Pei-Ying Lin, Tzu-Hung Hsiao, and Pan-Chyr Yang, National Taiwan University Hospital; Tzu-Pin Lu, National Taiwan University; Emily Pei-Ying Lin, Tzu-Hung Hsiao, Jo-Yang Lu, Siao-Han Wong, Konan Peck, and Pan-Chyr Yang, Academia Sinica, Taipei; Tzu-Hung Hsiao, Taichung Veterans General Hospital, Taichung, Taiwan; and Takashi Takahashi, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Siao-Han Wong
- Emily Pei-Ying Lin, Tzu-Hung Hsiao, and Pan-Chyr Yang, National Taiwan University Hospital; Tzu-Pin Lu, National Taiwan University; Emily Pei-Ying Lin, Tzu-Hung Hsiao, Jo-Yang Lu, Siao-Han Wong, Konan Peck, and Pan-Chyr Yang, Academia Sinica, Taipei; Tzu-Hung Hsiao, Taichung Veterans General Hospital, Taichung, Taiwan; and Takashi Takahashi, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tzu-Pin Lu
- Emily Pei-Ying Lin, Tzu-Hung Hsiao, and Pan-Chyr Yang, National Taiwan University Hospital; Tzu-Pin Lu, National Taiwan University; Emily Pei-Ying Lin, Tzu-Hung Hsiao, Jo-Yang Lu, Siao-Han Wong, Konan Peck, and Pan-Chyr Yang, Academia Sinica, Taipei; Tzu-Hung Hsiao, Taichung Veterans General Hospital, Taichung, Taiwan; and Takashi Takahashi, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Konan Peck
- Emily Pei-Ying Lin, Tzu-Hung Hsiao, and Pan-Chyr Yang, National Taiwan University Hospital; Tzu-Pin Lu, National Taiwan University; Emily Pei-Ying Lin, Tzu-Hung Hsiao, Jo-Yang Lu, Siao-Han Wong, Konan Peck, and Pan-Chyr Yang, Academia Sinica, Taipei; Tzu-Hung Hsiao, Taichung Veterans General Hospital, Taichung, Taiwan; and Takashi Takahashi, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takashi Takahashi
- Emily Pei-Ying Lin, Tzu-Hung Hsiao, and Pan-Chyr Yang, National Taiwan University Hospital; Tzu-Pin Lu, National Taiwan University; Emily Pei-Ying Lin, Tzu-Hung Hsiao, Jo-Yang Lu, Siao-Han Wong, Konan Peck, and Pan-Chyr Yang, Academia Sinica, Taipei; Tzu-Hung Hsiao, Taichung Veterans General Hospital, Taichung, Taiwan; and Takashi Takahashi, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Pan-Chyr Yang
- Emily Pei-Ying Lin, Tzu-Hung Hsiao, and Pan-Chyr Yang, National Taiwan University Hospital; Tzu-Pin Lu, National Taiwan University; Emily Pei-Ying Lin, Tzu-Hung Hsiao, Jo-Yang Lu, Siao-Han Wong, Konan Peck, and Pan-Chyr Yang, Academia Sinica, Taipei; Tzu-Hung Hsiao, Taichung Veterans General Hospital, Taichung, Taiwan; and Takashi Takahashi, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
29
|
VCE-004.8, A Multitarget Cannabinoquinone, Attenuates Adipogenesis and Prevents Diet-Induced Obesity. Sci Rep 2018; 8:16092. [PMID: 30382123 PMCID: PMC6208444 DOI: 10.1038/s41598-018-34259-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 10/06/2018] [Indexed: 01/01/2023] Open
Abstract
Over the past few years, the endocannabinoid system (ECs) has emerged as a crucial player for the regulation of food intake and energy metabolism, and its pharmacological manipulation represents a novel strategy for the management of metabolic diseases. The discovery that VCE-004.8, a dual PPARγ and CB2 receptor agonist, also inhibits prolyl-hydroxylases (PHDs) and activates the HIF pathway provided a rationale to investigate its effect in in vitro models of adipogenesis and in a murine model of metabolic syndrome, all processes critically regulated by these targets of VCE-004.8. In accordance with its different binding mode to PPARγ compared to rosiglitazone (RGZ), VCE-004.8 neither induced adipogenic differentiation, nor affected osteoblastogenesis. Daily administration of VCE-004.8 (20 mg/kg) to HFD mice for 3-wks induced a significant reduction in body weight gain, total fat mass, adipocyte volume and plasma triglycerides levels. VCE-004.8 could also significantly ameliorate glucose tolerance, reduce leptin levels (a marker of adiposity) and increase adiponectin and incretins (GLP-1 and GIP) levels. Remarkably, VCE-004.8 increased the FGF21 mRNA expression in white and brown adipose, as well as in a BAT cell line, qualifying cannabinoaminoquinones as a class of novel therapeutic candidates for the management of obesity and its common metabolic co-morbidities.
Collapse
|
30
|
Evans CE, Spier AB, Zhao YY. Sepsis-induced thrombus formation and cell-specific HIFs. Thromb Res 2018; 171:187-189. [PMID: 30220434 DOI: 10.1016/j.thromres.2018.08.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 08/01/2018] [Accepted: 08/24/2018] [Indexed: 01/11/2023]
Affiliation(s)
- Colin E Evans
- Program for Lung and Vascular Biology, Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA; Department of Pediatrics, Division of Critical Care, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| | - Addie B Spier
- Metro Infectious Disease Consultants, Chicago, IL, USA
| | - You-Yang Zhao
- Program for Lung and Vascular Biology, Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA; Department of Pediatrics, Division of Critical Care, Northwestern University Feinberg School of Medicine, Chicago, IL, USA; Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA; Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA; Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
31
|
Bondu V, Bitting C, Poland VL, Hanson JA, Harkins MS, Lathrop S, Nolte KB, Lawrence DA, Buranda T. Upregulation of P2Y 2R, Active uPA, and PAI-1 Are Essential Components of Hantavirus Cardiopulmonary Syndrome. Front Cell Infect Microbiol 2018; 8:169. [PMID: 29930915 PMCID: PMC6001748 DOI: 10.3389/fcimb.2018.00169] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 05/03/2018] [Indexed: 12/13/2022] Open
Abstract
Sin Nombre virus (SNV) causes hantavirus cardiopulmonary pulmonary syndrome (HCPS) with the loss of pulmonary vascular endothelial integrity, and pulmonary edema without causing cytopathic effects on the vascular endothelium. HCPS is associated primarily with a dysregulated immune response. We previously found occult signs of hemostatic imbalance in the form of a sharp >30-100 fold increase in the expression of plasminogen activator inhibitor type 1 (PAI-1), in serial blood plasma draws of terminal stage-patients. However, the mechanism of the increase in PAI-1 remains unclear. PAI-1 is a primary inhibitor of fibrinolysis caused by tissue plasminogen activator (tPA) and urokinase plasminogen activator plasma (uPA). Here, we investigate factors that contribute to PAI-1 upregulation during HCPS. Using zymography, we found evidence of PAI-1-refractory uPA activity and no tPA activity in plasma samples drawn from HCPS patients. The sole prevalence of uPA activity suggested that severe inflammation drove PAI-1 activity. We have recently reported that the P2Y2 receptor (P2Y2R) mediates SNV infectivity by interacting in cis with β3 integrins, which activates the latter during infection. P2Y2R is a known effector for several biological processes relevant to HCPS pathogenesis, such as upregulation of tissue factor (TF), a primary initiator of the coagulation cascade, stimulating vascular permeability and leukocyte homing to sites of infection. As P2Y2R is prone to upregulation under conditions of inflammation, we compared the expression level of P2Y2R in formalin fixed tissues of HCPS decedents using a TaqMan assay and immunohistochemistry. Our TaqMan results show that the expression of P2Y2R is upregulated significantly in HCPS cases compared to non- HCPS controls (P < 0.001). Immunohistochemistry showed that lung macrophages were the primary reservoir of high and coincident localization of P2Y2R, uPA, PAI-1, and TF antigens. We also observed increased staining for SNV antigens in the same tissue segments where P2Y2R expression was upregulated. Conversely, sections of low P2Y2R expression showed weak manifestations of macrophages, SNV, PAI-1, and TF. Coincident localization of P2Y2R and PAI-1 on macrophage deposits suggests an inflammation-dependent mechanism of increasing pro-coagulant activity in HCPS in the absence of tissue injury.
Collapse
Affiliation(s)
- Virginie Bondu
- Department of Pathology, University of New Mexico School of Medicine, Albuquerque, NM, United States
| | - Casey Bitting
- Department of Pathology, University of New Mexico School of Medicine, Albuquerque, NM, United States
| | - Valerie L Poland
- Office of the Medical Investigator, University of New Mexico School of Medicine, Albuquerque, NM, United States
| | - Joshua A Hanson
- Department of Pathology, University of New Mexico School of Medicine, Albuquerque, NM, United States
| | - Michelle S Harkins
- Division of Infectious Disease, Pulmonary, Critical Care, and Sleep, Department of Internal Medicine, School of Medicine, University of New Mexico, Albuquerque, NM, United States
| | - Sarah Lathrop
- Department of Pathology, University of New Mexico School of Medicine, Albuquerque, NM, United States.,Office of the Medical Investigator, University of New Mexico School of Medicine, Albuquerque, NM, United States
| | - Kurt B Nolte
- Department of Pathology, University of New Mexico School of Medicine, Albuquerque, NM, United States.,Office of the Medical Investigator, University of New Mexico School of Medicine, Albuquerque, NM, United States
| | - Daniel A Lawrence
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Tione Buranda
- Department of Pathology, University of New Mexico School of Medicine, Albuquerque, NM, United States
| |
Collapse
|
32
|
Giraud S, Steichen C, Allain G, Couturier P, Labourdette D, Lamarre S, Ameteau V, Tillet S, Hannaert P, Thuillier R, Hauet T. Dynamic transcriptomic analysis of Ischemic Injury in a Porcine Pre-Clinical Model mimicking Donors Deceased after Circulatory Death. Sci Rep 2018; 8:5986. [PMID: 29654283 PMCID: PMC5899088 DOI: 10.1038/s41598-018-24282-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 03/28/2018] [Indexed: 02/06/2023] Open
Abstract
Due to organ shortage, clinicians are prone to consider alternative type of organ donors among them donors deceased after circulatory death (DCD). However, especially using these organs which are more prone to graft dysfunction, there is a need to better understand mechanistic events ocuring during ischemia phase and leading to ischemia/reperfusion injuries (IRI). The aim of this study is to provide a dynamic transcriptomic analysis of preclinical porcine model kidneys subjected to ischemic stress mimicking DCD donor. We compared cortex and corticomedullary junction (CMJ) tissues from porcine kidneys submitted to 60 min warm ischemia (WI) followed by 0, 6 or 24 hours of cold storage in University of Wisconsin solution versus control non-ischemic kidneys (n = 5 per group). 29 cortex genes and 113 CMJ genes were significantly up or down-regulated after WI versus healthy kidneys, and up to 400 genes were regulated after WI followed by 6 or 24 hours of cold storage (p < 0.05). Functionnal enrichment analysis (home selected gene kinetic classification, Gene-ontology-biological processes and Gene-ontology-molecular-function) revealed relevant genes implication during WI and cold storage. We uncovered targets which we will further validate as biomarkers and new therapeutic targets to optimize graft kidney quality before transplantation and improve whole transplantation outcome.
Collapse
Affiliation(s)
- Sebastien Giraud
- Inserm U1082 IRTOMIT, Poitiers, F-86000, France.,Université de Poitiers, Faculté de Médecine et de Pharmacie, Poitiers, F-86000, France.,CHU Poitiers, Service de Biochimie, Poitiers, F-86000, France
| | - Clara Steichen
- Inserm U1082 IRTOMIT, Poitiers, F-86000, France.,Université de Poitiers, Faculté de Médecine et de Pharmacie, Poitiers, F-86000, France
| | - Geraldine Allain
- Inserm U1082 IRTOMIT, Poitiers, F-86000, France.,Université de Poitiers, Faculté de Médecine et de Pharmacie, Poitiers, F-86000, France.,CHU Poitiers, Service de chirurgie cardio-thoracique, Poitiers, 86000, France
| | - Pierre Couturier
- Inserm U1082 IRTOMIT, Poitiers, F-86000, France.,CHU Poitiers, Service de Biochimie, Poitiers, F-86000, France.,MOPICT, IBiSA plateforme 'Experimental Surgery and Transplantation', Domaine du Magneraud, Surgères, F-17700, France
| | | | - Sophie Lamarre
- LISBP, Université de Toulouse, CNRS, INRA, INSA, Toulouse, F- 31077, France
| | - Virginie Ameteau
- Inserm U1082 IRTOMIT, Poitiers, F-86000, France.,Université de Poitiers, Faculté de Médecine et de Pharmacie, Poitiers, F-86000, France
| | - Solenne Tillet
- Inserm U1082 IRTOMIT, Poitiers, F-86000, France.,Université de Poitiers, Faculté de Médecine et de Pharmacie, Poitiers, F-86000, France
| | | | - Raphael Thuillier
- Inserm U1082 IRTOMIT, Poitiers, F-86000, France.,Université de Poitiers, Faculté de Médecine et de Pharmacie, Poitiers, F-86000, France.,CHU Poitiers, Service de Biochimie, Poitiers, F-86000, France
| | - Thierry Hauet
- Inserm U1082 IRTOMIT, Poitiers, F-86000, France. .,Université de Poitiers, Faculté de Médecine et de Pharmacie, Poitiers, F-86000, France. .,CHU Poitiers, Service de Biochimie, Poitiers, F-86000, France. .,MOPICT, IBiSA plateforme 'Experimental Surgery and Transplantation', Domaine du Magneraud, Surgères, F-17700, France. .,FHU SUPORT 'SUrvival oPtimization in ORgan Transplantation', Poitiers, F-86000, France.
| |
Collapse
|
33
|
Regulation of Tumor Progression by Programmed Necrosis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:3537471. [PMID: 29636841 PMCID: PMC5831895 DOI: 10.1155/2018/3537471] [Citation(s) in RCA: 135] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 11/28/2017] [Indexed: 12/12/2022]
Abstract
Rapidly growing malignant tumors frequently encounter hypoxia and nutrient (e.g., glucose) deprivation, which occurs because of insufficient blood supply. This results in necrotic cell death in the core region of solid tumors. Necrotic cells release their cellular cytoplasmic contents into the extracellular space, such as high mobility group box 1 (HMGB1), which is a nonhistone nuclear protein, but acts as a proinflammatory and tumor-promoting cytokine when released by necrotic cells. These released molecules recruit immune and inflammatory cells, which exert tumor-promoting activity by inducing angiogenesis, proliferation, and invasion. Development of a necrotic core in cancer patients is also associated with poor prognosis. Conventionally, necrosis has been thought of as an unregulated process, unlike programmed cell death processes like apoptosis and autophagy. Recently, necrosis has been recognized as a programmed cell death, encompassing processes such as oncosis, necroptosis, and others. Metabolic stress-induced necrosis and its regulatory mechanisms have been poorly investigated until recently. Snail and Dlx-2, EMT-inducing transcription factors, are responsible for metabolic stress-induced necrosis in tumors. Snail and Dlx-2 contribute to tumor progression by promoting necrosis and inducing EMT and oncogenic metabolism. Oncogenic metabolism has been shown to play a role(s) in initiating necrosis. Here, we discuss the molecular mechanisms underlying metabolic stress-induced programmed necrosis that promote tumor progression and aggressiveness.
Collapse
|
34
|
Fang WF, Chen YM, Lin CY, Huang HL, Yeh H, Chang YT, Huang KT, Lin MC. Histone deacetylase 2 (HDAC2) attenuates lipopolysaccharide (LPS)-induced inflammation by regulating PAI-1 expression. JOURNAL OF INFLAMMATION-LONDON 2018; 15:3. [PMID: 29344006 PMCID: PMC5763578 DOI: 10.1186/s12950-018-0179-6] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 01/04/2018] [Indexed: 12/22/2022]
Abstract
Background Sepsis is a life-threatening organ dysfunction caused by dysregulated host response to infection, and is primarily characterized by an uncontrolled systemic inflammatory response. In the present study, we developed an effective adjunct therapy mediated by a novel mechanism, to attenuate overt inflammation. LPS-treated macrophages were adopted as an in vitro model of endotoxin-induced inflammation during sepsis. Experiments were carried out using primary mouse peritoneal macrophages and the murine macrophage cell line RAW264.7, to elucidate the mechanisms by which HDAC2 modulates endotoxin-induced inflammation. Results Results revealed that PAI-1, TNF, and MIP-2 expression were inhibited by theophylline, an HDAC2 enhancer, in a RAW macrophage cell line, following LPS-induced inflammation. Thus, HDAC2 plays an important role in immune defense by regulating the expression of inflammatory genes via the c-Jun/PAI-1 pathway. During LPS-induced inflammation, overexpression of HDAC2 was found to inhibit PAI-1, TNF, and MIP-2 expression. Following LPS stimulation, HDAC2 knockdown increased nuclear translocation and DNA binding of c-Jun to the PAI-1 gene promoter, thereby activating PAI-1 gene transcription. Furthermore, inhibition of PAI-1 by TM5275 alone or in combination with theophylline notably suppressed TNF and MIP-2 expression. Conclusion HDAC2 can attenuate lipopolysaccharide-induced inflammation by regulating c-Jun and PAI-1 expression in macrophages.
Collapse
Affiliation(s)
- Wen-Feng Fang
- 1Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, 833 Taiwan.,2Department of Respiratory Therapy, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, 123 Ta-Pei Rd, Niao-Sung Dist, Kaohsiung, 833 Taiwan.,3Department of Respiratory Care, Chang Gung University of Science and Technology, Chiayi, 813 Taiwan
| | - Yu-Mu Chen
- 1Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, 833 Taiwan
| | - Chiung-Yu Lin
- 1Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, 833 Taiwan
| | - Hui-Lin Huang
- 1Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, 833 Taiwan
| | - Hua Yeh
- 1Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, 833 Taiwan
| | - Ya-Ting Chang
- 1Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, 833 Taiwan
| | - Kuo-Tung Huang
- 1Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, 833 Taiwan
| | - Meng-Chih Lin
- 1Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, 833 Taiwan.,2Department of Respiratory Therapy, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, 123 Ta-Pei Rd, Niao-Sung Dist, Kaohsiung, 833 Taiwan
| |
Collapse
|
35
|
Strowitzki MJ, Ritter AS, Radhakrishnan P, Harnoss JM, Opitz VM, Biller M, Wehrmann J, Keppler U, Scheer J, Wallwiener M, Schmidt T, Ulrich A, Schneider M. Pharmacological HIF-inhibition attenuates postoperative adhesion formation. Sci Rep 2017; 7:13151. [PMID: 29030625 PMCID: PMC5640636 DOI: 10.1038/s41598-017-13638-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 09/29/2017] [Indexed: 02/06/2023] Open
Abstract
Peritoneal adhesions represent a common complication of abdominal surgery, and tissue hypoxia is a main determinant in adhesion formation. Reliable therapeutic options to reduce peritoneal adhesions are scarce. We investigated whether the formation of postsurgical adhesions can be affected by pharmacological interference with hypoxia-inducible factors (HIFs). Mice were treated with a small molecule HIF-inhibitor, YC-1 (3-[5′-Hydroxymethyl-2′-furyl]-1-benzyl-indazole), or vehicle three days before and seven days after induction of peritoneal adhesions or, alternatively, once during induction of peritoneal adhesions. Pretreatment or single intraperitoneal lavage with YC-1 significantly reduced postoperative adhesion formation without prompting systemic adverse effects. Expression analyses of cytokines in peritoneal tissue and fluid and in vitro assays applying macrophages and peritoneal fibroblasts indicated that this effect was cooperatively mediated by various putatively HIF-1α-dependent mechanisms, comprising attenuated pro-inflammatory activation of macrophages, impaired recruitment and activation of peritoneal fibroblasts, mitigated epithelial-mesenchymal-transition (EMT), as well as enhanced fibrinolysis and impaired angiogenesis. Thus, this study identifies prevention of postsurgical peritoneal adhesions as a novel and promising field for the application of HIF inhibitors in clinical practice.
Collapse
Affiliation(s)
- Moritz J Strowitzki
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Alina S Ritter
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Praveen Radhakrishnan
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Jonathan M Harnoss
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany.,Cancer Immunology, Genentech, Inc., South San Francisco, USA
| | - Vanessa M Opitz
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Marvin Biller
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Julian Wehrmann
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Ulrich Keppler
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Jana Scheer
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Markus Wallwiener
- Department of General Gynaecology and Obstetrics, University of Heidelberg, Heidelberg, Germany
| | - Thomas Schmidt
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Alexis Ulrich
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Martin Schneider
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany.
| |
Collapse
|
36
|
Rabieian R, Boshtam M, Zareei M, Kouhpayeh S, Masoudifar A, Mirzaei H. Plasminogen Activator Inhibitor Type-1 as a Regulator of Fibrosis. J Cell Biochem 2017; 119:17-27. [PMID: 28520219 DOI: 10.1002/jcb.26146] [Citation(s) in RCA: 121] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 05/17/2017] [Indexed: 12/17/2022]
Abstract
Fibrosis is known as a frequent and irreversible pathological condition which is associated with organ failure. Tissue fibrosis is a central process in a variety of chronic progressive diseases such as diabetes, hypertension, and persistent inflammation. This state could contribute to chronic injury and the initiation of tissue repair. Fibrotic disorders represent abnormal wound healing with defective matrix turnover and clearance that lead to excessive accumulation of extracellular matrix components. A variety of identified growth factors, cytokines, and persistently activated myofibroblasts have critical roles in the pathogenesis of fibrosis. Irrespective of etiology, the transforming growth factor-β pathway is the major driver of fibrotic response. Plasminogen activator inhibitor-1 (PAI-1) is a crucial downstream target of this pathway. Transforming growth factor-β positively regulates PAI-1 gene expression via two main pathways including Smad-mediated canonical and non-canonical pathways. Overexpression of PAI-1 reduces extracellular matrix degradation via perturbing the plasminogen activation system. Indeed, elevated PAI-1 levels inhibit proteolytic activity of tissue plasminogen activator and urokinase plasminogen activator which could contribute to a variety of inflammatory elements in the injury site and to excessive matrix deposition. This review summarizes the current knowledge of critical pathways that regulate PAI-1 gene expression and suggests effective approaches for the treatment of fibrotic disease. J. Cell. Biochem. 119: 17-27, 2018. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Reyhaneh Rabieian
- Department of Genetics and Molecular Biology, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Maryam Boshtam
- Isfahan Cardiovascular Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mahshid Zareei
- Department of Biology, School of Sciences, University of Isfahan, Isfahan, Iran
| | - Shirin Kouhpayeh
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Aria Masoudifar
- Department of Molecular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Hamed Mirzaei
- Department of Medical Biotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
37
|
Andrews RN, Metheny-Barlow LJ, Peiffer AM, Hanbury DB, Tooze JA, Bourland JD, Hampson RE, Deadwyler SA, Cline JM. Cerebrovascular Remodeling and Neuroinflammation is a Late Effect of Radiation-Induced Brain Injury in Non-Human Primates. Radiat Res 2017; 187:599-611. [PMID: 28398880 PMCID: PMC5508216 DOI: 10.1667/rr14616.1] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Fractionated whole-brain irradiation (fWBI) is a mainstay of treatment for patients with intracranial neoplasia; however late-delayed radiation-induced normal tissue injury remains a major adverse consequence of treatment, with deleterious effects on quality of life for affected patients. We hypothesize that cerebrovascular injury and remodeling after fWBI results in ischemic injury to dependent white matter, which contributes to the observed cognitive dysfunction. To evaluate molecular effectors of radiation-induced brain injury (RIBI), real-time quantitative polymerase chain reaction (RT-qPCR) was performed on the dorsolateral prefrontal cortex (DLPFC, Brodmann area 46), hippocampus and temporal white matter of 4 male Rhesus macaques (age 6-11 years), which had received 40 Gray (Gy) fWBI (8 fractions of 5 Gy each, twice per week), and 3 control comparators. All fWBI animals developed neurologic impairment; humane euthanasia was elected at a median of 6 months. Radiation-induced brain injury was confirmed histopathologically in all animals, characterized by white matter degeneration and necrosis, and multifocal cerebrovascular injury consisting of perivascular edema, abnormal angiogenesis and perivascular extracellular matrix deposition. Herein we demonstrate that RIBI is associated with white matter-specific up-regulation of hypoxia-associated lactate dehydrogenase A (LDHA) and that increased gene expression of fibronectin 1 (FN1), SERPINE1 and matrix metalloprotease 2 (MMP2) may contribute to cerebrovascular remodeling in late-delayed RIBI. Additionally, vascular stability and maturation associated tumor necrosis super family member 15 (TNFSF15) and vascular endothelial growth factor beta (VEGFB) mRNAs were increased within temporal white matter. We also demonstrate that radiation-induced brain injury is associated with decreases in white matter-specific expression of neurotransmitter receptors SYP, GRIN2A and GRIA4. We additionally provide evidence that macrophage/microglial mediated neuroinflammation may contribute to RIBI through increased gene expression of the macrophage chemoattractant CCL2 and macrophage/microglia associated CD68. Global patterns in cerebral gene expression varied significantly between regions examined (P < 0.0001, Friedman's test), with effects most prominent within cerebral white matter.
Collapse
Affiliation(s)
- Rachel N. Andrews
- Department of Pathology, Section on Comparative Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157
| | - Linda J. Metheny-Barlow
- Department of Radiation Oncology, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157
- Department of Brain Tumor Center of Excellence, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157
| | - Ann M. Peiffer
- Department of Radiation Oncology, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157
- Department of Brain Tumor Center of Excellence, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157
| | - David B. Hanbury
- Department of Psychology, Averett University, Danville, Virginia 24541
| | - Janet A. Tooze
- Department of Biostatistical Sciences, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157
| | - J. Daniel Bourland
- Department of Radiation Oncology, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157
- Department of Brain Tumor Center of Excellence, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157
| | - Robert E. Hampson
- Department of Physiology & Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157
| | - Samuel A. Deadwyler
- Department of Physiology & Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157
| | - J. Mark Cline
- Department of Pathology, Section on Comparative Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157
| |
Collapse
|
38
|
Cummins EP, Keogh CE. Respiratory gases and the regulation of transcription. Exp Physiol 2016; 101:986-1002. [DOI: 10.1113/ep085715] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 05/23/2016] [Indexed: 12/17/2022]
Affiliation(s)
- Eoin P. Cummins
- School of Medicine; University College Dublin; Belfield 4 Dublin Ireland
| | - Ciara E. Keogh
- School of Medicine; University College Dublin; Belfield 4 Dublin Ireland
| |
Collapse
|
39
|
Kaneko M, Minematsu T, Yoshida M, Nishijima Y, Noguchi H, Ohta Y, Nakagami G, Mori T, Sanada H. Compression-induced HIF-1 enhances thrombosis and PAI-1 expression in mouse skin. Wound Repair Regen 2015; 23:657-63. [DOI: 10.1111/wrr.12312] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Accepted: 04/29/2015] [Indexed: 11/26/2022]
Affiliation(s)
- Maki Kaneko
- Department of Gerontological Nursing/Wound Care Management, Graduate School of Medicine; The University of Tokyo; Tokyo Japan
| | - Takeo Minematsu
- Department of Gerontological Nursing/Wound Care Management, Graduate School of Medicine; The University of Tokyo; Tokyo Japan
| | - Mikako Yoshida
- Department of Gerontological Nursing/Wound Care Management, Graduate School of Medicine; The University of Tokyo; Tokyo Japan
| | - Yoshimi Nishijima
- Department of Gerontological Nursing/Wound Care Management, Graduate School of Medicine; The University of Tokyo; Tokyo Japan
| | - Hiroshi Noguchi
- Department of Life Support Technology (Molten), Graduate School of Medicine; The University of Tokyo; Tokyo Japan
| | - Yasunori Ohta
- Department of Pathology, Research Hospital, Institute of Medical Science; The University of Tokyo; Tokyo Japan
| | - Gojiro Nakagami
- Department of Gerontological Nursing/Wound Care Management, Graduate School of Medicine; The University of Tokyo; Tokyo Japan
| | - Taketoshi Mori
- Department of Life Support Technology (Molten), Graduate School of Medicine; The University of Tokyo; Tokyo Japan
| | - Hiromi Sanada
- Department of Gerontological Nursing/Wound Care Management, Graduate School of Medicine; The University of Tokyo; Tokyo Japan
| |
Collapse
|
40
|
Kuchnio A, Moens S, Bruning U, Kuchnio K, Cruys B, Thienpont B, Broux M, Ungureanu AA, Leite de Oliveira R, Bruyère F, Cuervo H, Manderveld A, Carton A, Hernandez-Fernaud JR, Zanivan S, Bartic C, Foidart JM, Noel A, Vinckier S, Lambrechts D, Dewerchin M, Mazzone M, Carmeliet P. The Cancer Cell Oxygen Sensor PHD2 Promotes Metastasis via Activation of Cancer-Associated Fibroblasts. Cell Rep 2015; 12:992-1005. [PMID: 26235614 DOI: 10.1016/j.celrep.2015.07.010] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Revised: 06/23/2015] [Accepted: 07/07/2015] [Indexed: 12/13/2022] Open
Abstract
Several questions about the role of the oxygen sensor prolyl-hydroxylase 2 (PHD2) in cancer have not been addressed. First, the role of PHD2 in metastasis has not been studied in a spontaneous tumor model. Here, we show that global PHD2 haplodeficiency reduced metastasis without affecting tumor growth. Second, it is unknown whether PHD2 regulates cancer by affecting cancer-associated fibroblasts (CAFs). We show that PHD2 haplodeficiency reduced metastasis via two mechanisms: (1) by decreasing CAF activation, matrix production, and contraction by CAFs, an effect that surprisingly relied on PHD2 deletion in cancer cells, but not in CAFs; and (2) by improving tumor vessel normalization. Third, the effect of concomitant PHD2 inhibition in malignant and stromal cells (mimicking PHD2 inhibitor treatment) is unknown. We show that global PHD2 haplodeficiency, induced not only before but also after tumor onset, impaired metastasis. These findings warrant investigation of PHD2's therapeutic potential.
Collapse
Affiliation(s)
- Anna Kuchnio
- Laboratory of Angiogenesis and Neurovascular Link, Department of Oncology, KU Leuven, Herestraat 49, 3000 Leuven, Belgium; Laboratory of Angiogenesis and Neurovascular Link, Vesalius Research Center, VIB, Herestraat 49, 3000 Leuven, Belgium
| | - Stijn Moens
- Laboratory of Angiogenesis and Neurovascular Link, Department of Oncology, KU Leuven, Herestraat 49, 3000 Leuven, Belgium; Laboratory of Angiogenesis and Neurovascular Link, Vesalius Research Center, VIB, Herestraat 49, 3000 Leuven, Belgium
| | - Ulrike Bruning
- Laboratory of Angiogenesis and Neurovascular Link, Department of Oncology, KU Leuven, Herestraat 49, 3000 Leuven, Belgium; Laboratory of Angiogenesis and Neurovascular Link, Vesalius Research Center, VIB, Herestraat 49, 3000 Leuven, Belgium
| | - Karol Kuchnio
- Laboratory of Angiogenesis and Neurovascular Link, Department of Oncology, KU Leuven, Herestraat 49, 3000 Leuven, Belgium; Laboratory of Angiogenesis and Neurovascular Link, Vesalius Research Center, VIB, Herestraat 49, 3000 Leuven, Belgium
| | - Bert Cruys
- Laboratory of Angiogenesis and Neurovascular Link, Department of Oncology, KU Leuven, Herestraat 49, 3000 Leuven, Belgium; Laboratory of Angiogenesis and Neurovascular Link, Vesalius Research Center, VIB, Herestraat 49, 3000 Leuven, Belgium
| | - Bernard Thienpont
- Laboratory for Translational Genetics, Department of Oncology, KU Leuven, Herestraat 49, 3000 Leuven, Belgium; Laboratory for Translational Genetics, Vesalius Research Center, VIB, Herestraat 49, 3000 Leuven, Belgium
| | - Michaël Broux
- Laboratory of Angiogenesis and Neurovascular Link, Department of Oncology, KU Leuven, Herestraat 49, 3000 Leuven, Belgium; Laboratory of Angiogenesis and Neurovascular Link, Vesalius Research Center, VIB, Herestraat 49, 3000 Leuven, Belgium
| | - Andreea Alexandra Ungureanu
- Laboratory of Soft Matter and Biophysics, Department of Physics and Astronomy, KU Leuven, Celestijnenlaan 200D, 3001 Heverlee, Belgium
| | - Rodrigo Leite de Oliveira
- Laboratory of Angiogenesis and Neurovascular Link, Department of Oncology, KU Leuven, Herestraat 49, 3000 Leuven, Belgium; Laboratory of Angiogenesis and Neurovascular Link, Vesalius Research Center, VIB, Herestraat 49, 3000 Leuven, Belgium; Laboratory of Molecular Oncology and Angiogenesis, Department of Oncology, KU Leuven, Herestraat 49, 3000 Leuven, Belgium; Laboratory of Molecular Oncology and Angiogenesis, Vesalius Research Center, VIB, Herestraat 49, 3000 Leuven, Belgium
| | - Françoise Bruyère
- Laboratory of Angiogenesis and Neurovascular Link, Department of Oncology, KU Leuven, Herestraat 49, 3000 Leuven, Belgium; Laboratory of Angiogenesis and Neurovascular Link, Vesalius Research Center, VIB, Herestraat 49, 3000 Leuven, Belgium
| | - Henar Cuervo
- Laboratory of Angiogenesis and Neurovascular Link, Department of Oncology, KU Leuven, Herestraat 49, 3000 Leuven, Belgium; Laboratory of Angiogenesis and Neurovascular Link, Vesalius Research Center, VIB, Herestraat 49, 3000 Leuven, Belgium
| | - Ann Manderveld
- Laboratory of Angiogenesis and Neurovascular Link, Department of Oncology, KU Leuven, Herestraat 49, 3000 Leuven, Belgium; Laboratory of Angiogenesis and Neurovascular Link, Vesalius Research Center, VIB, Herestraat 49, 3000 Leuven, Belgium
| | - An Carton
- Laboratory of Angiogenesis and Neurovascular Link, Department of Oncology, KU Leuven, Herestraat 49, 3000 Leuven, Belgium; Laboratory of Angiogenesis and Neurovascular Link, Vesalius Research Center, VIB, Herestraat 49, 3000 Leuven, Belgium
| | - Juan Ramon Hernandez-Fernaud
- Laboratory of Vascular Proteomics, Cancer Research UK Beatson Institute, Switchback Road, Bearsden, Glasgow G61 1BD, UK
| | - Sara Zanivan
- Laboratory of Vascular Proteomics, Cancer Research UK Beatson Institute, Switchback Road, Bearsden, Glasgow G61 1BD, UK
| | - Carmen Bartic
- Laboratory of Soft Matter and Biophysics, Department of Physics and Astronomy, KU Leuven, Celestijnenlaan 200D, 3001 Heverlee, Belgium; IMEC, Kapeldreef 75, 3001 Heverlee, Belgium
| | - Jean-Michel Foidart
- Laboratory of Tumor and Developmental Biology, GIGA-Cancer, University of Liège, Avenue de l'Hôpital 3, 4000 Liège, Belgium
| | - Agnes Noel
- Laboratory of Tumor and Developmental Biology, GIGA-Cancer, University of Liège, Avenue de l'Hôpital 3, 4000 Liège, Belgium
| | - Stefan Vinckier
- Laboratory of Angiogenesis and Neurovascular Link, Department of Oncology, KU Leuven, Herestraat 49, 3000 Leuven, Belgium; Laboratory of Angiogenesis and Neurovascular Link, Vesalius Research Center, VIB, Herestraat 49, 3000 Leuven, Belgium
| | - Diether Lambrechts
- Laboratory for Translational Genetics, Department of Oncology, KU Leuven, Herestraat 49, 3000 Leuven, Belgium; Laboratory for Translational Genetics, Vesalius Research Center, VIB, Herestraat 49, 3000 Leuven, Belgium
| | - Mieke Dewerchin
- Laboratory of Angiogenesis and Neurovascular Link, Department of Oncology, KU Leuven, Herestraat 49, 3000 Leuven, Belgium; Laboratory of Angiogenesis and Neurovascular Link, Vesalius Research Center, VIB, Herestraat 49, 3000 Leuven, Belgium
| | - Massimiliano Mazzone
- Laboratory of Molecular Oncology and Angiogenesis, Department of Oncology, KU Leuven, Herestraat 49, 3000 Leuven, Belgium; Laboratory of Molecular Oncology and Angiogenesis, Vesalius Research Center, VIB, Herestraat 49, 3000 Leuven, Belgium
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Neurovascular Link, Department of Oncology, KU Leuven, Herestraat 49, 3000 Leuven, Belgium; Laboratory of Angiogenesis and Neurovascular Link, Vesalius Research Center, VIB, Herestraat 49, 3000 Leuven, Belgium.
| |
Collapse
|
41
|
PEDF and its roles in physiological and pathological conditions: implication in diabetic and hypoxia-induced angiogenic diseases. Clin Sci (Lond) 2015; 128:805-23. [PMID: 25881671 PMCID: PMC4557399 DOI: 10.1042/cs20130463] [Citation(s) in RCA: 114] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Pigment epithelium-derived factor (PEDF) is a broadly expressed multifunctional member of the serine proteinase inhibitor (serpin) family. This widely studied protein plays critical roles in many physiological and pathophysiological processes, including neuroprotection, angiogenesis, fibrogenesis and inflammation. The present review summarizes the temporal and spatial distribution patterns of PEDF in a variety of developing and adult organs, and discusses its functions in maintaining physiological homoeostasis. The major focus of the present review is to discuss the implication of PEDF in diabetic and hypoxia-induced angiogenesis, and the pathways mediating PEDF's effects under these conditions. Furthermore, the regulatory mechanisms of PEDF expression, function and degradation are also reviewed. Finally, the therapeutic potential of PEDF as an anti-angiogenic drug is briefly summarized.
Collapse
|
42
|
Eftedal I, Fredriksen HA, Hjelde A, Møllerløkken A. Evaluating PAI-1 as a biomarker for stress in diving: human serum total PAI-1 is unaltered after 2 h dry exposures to 280 kPa hyperbaric air. Physiol Rep 2015; 3:3/6/e12437. [PMID: 26109191 PMCID: PMC4510634 DOI: 10.14814/phy2.12437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Plasminogen activator inhibitor (PAI-1) is induced in the vasculature and secreted into the vascular lumen in response to inflammation and oxidative stress. We have previously reported a fivefold increase in plasma PAI-1 from rats exposed to 708 kPa hyperbaric air. In the current study we assess the potential of human serum total PAI-1 as a biomarker for stress in compressed air diving. Eleven recreational divers, nine males and two females, completed four 2 h hyperbaric air exposures to 280 kPa in a pressure chamber over a period of 2 weeks. The air pressure corresponds to a diving depth of 18 m in water. Serum was collected before the study and again 3 h 30 min after completion of each hyperbaric exposure. All samples were taken in the afternoon to minimize the contribution of circadian variation. The analysis revealed no change in serum total PAI-1 after hyperbaric exposures within the group of divers (P = 0.064), but significant interindividual differences persisted throughout the study (P < 0.0005). A case of decompression sickness after the third round of hyperbaric exposure did not affect PAI-1. In conclusion, compressed air exposure to 280 kPa does not affect serum total PAI-1, and significant interindividual variation in PAI-1 levels may limit its usefulness as a biomarker. This does, however, not give a complete answer regarding PAI-1 in physiologically stressful dives. Further studies with different exposures and timing are needed for that.
Collapse
Affiliation(s)
- Ingrid Eftedal
- Department of Circulation and Medical Imaging; Faculty of Medicine; Norwegian University of Science and Technology; Trondheim Norway
| | - Hallvard Aglen Fredriksen
- Department of Circulation and Medical Imaging; Faculty of Medicine; Norwegian University of Science and Technology; Trondheim Norway
| | - Astrid Hjelde
- Department of Circulation and Medical Imaging; Faculty of Medicine; Norwegian University of Science and Technology; Trondheim Norway
| | - Andreas Møllerløkken
- Department of Circulation and Medical Imaging; Faculty of Medicine; Norwegian University of Science and Technology; Trondheim Norway
| |
Collapse
|
43
|
Hashimoto T, Shibasaki F. Hypoxia-inducible factor as an angiogenic master switch. Front Pediatr 2015; 3:33. [PMID: 25964891 PMCID: PMC4408850 DOI: 10.3389/fped.2015.00033] [Citation(s) in RCA: 154] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 04/07/2015] [Indexed: 12/19/2022] Open
Abstract
Hypoxia-inducible factors (HIFs) regulate the transcription of genes that mediate the response to hypoxia. HIFs are constantly expressed and degraded under normoxia, but stabilized under hypoxia. HIFs have been widely studied in physiological and pathological conditions and have been shown to contribute to the pathogenesis of various vascular diseases. In clinical settings, the HIF pathway has been studied for its role in inhibiting carcinogenesis. HIFs might also play a protective role in the pathology of ischemic diseases. Clinical trials of therapeutic angiogenesis after the administration of a single growth factor have yielded unsatisfactory or controversial results, possibly because the coordinated activity of different HIF-induced factors is necessary to induce mature vessel formation. Thus, manipulation of HIF activity to simultaneously induce a spectrum of angiogenic factors offers a superior strategy for therapeutic angiogenesis. Because HIF-2α plays an essential role in vascular remodeling, manipulation of HIF-2α is a promising approach to the treatment of ischemic diseases caused by arterial obstruction, where insufficient development of collateral vessels impedes effective therapy. Eukaryotic initiation factor 3 subunit e (eIF3e)/INT6 interacts specifically with HIF-2α and induces the proteasome inhibitor-sensitive degradation of HIF-2α, independent of hypoxia and von Hippel-Lindau protein. Treatment with eIF3e/INT6 siRNA stabilizes HIF-2α activity even under normoxic conditions and induces the expression of several angiogenic factors, at levels sufficient to produce functional arteries and veins in vivo. We have demonstrated that administration of eIF3e/INT6 siRNA to ischemic limbs or cold-injured brains reduces ischemic damage in animal models. This review summarizes the current understanding of the relationship between HIFs and vascular diseases. We also discuss novel oxygen-independent regulatory proteins that bind HIF-α and the implications of a new method for therapeutic angiogenesis using HIF stabilizers.
Collapse
Affiliation(s)
- Takuya Hashimoto
- Department of Surgery, Yale University School of Medicine , New Haven, CT , USA ; Division of Vascular Surgery, Department of Surgery, Graduate School of Medicine, The University of Tokyo , Tokyo , Japan
| | - Futoshi Shibasaki
- Department of Molecular Medical Research, Tokyo Metropolitan Institute of Medical Science , Tokyo , Japan
| |
Collapse
|
44
|
Lin CY, Chen HJ, Huang CC, Lai LC, Lu TP, Tseng GC, Kuo TT, Kuok QY, Hsu JL, Sung SY, Hung MC, Sher YP. ADAM9 promotes lung cancer metastases to brain by a plasminogen activator-based pathway. Cancer Res 2014; 74:5229-43. [PMID: 25060522 DOI: 10.1158/0008-5472.can-13-2995] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The transmembrane cell adhesion protein ADAM9 has been implicated in cancer cell migration and lung cancer metastasis to the brain, but the underpinning mechanisms are unclear and clinical support has been lacking. Here, we demonstrate that ADAM9 enhances the ability of tissue plasminogen activator (tPA) to cleave and stimulate the function of the promigratory protein CDCP1 to promote lung metastasis. Blocking this mechanism of cancer cell migration prolonged survival in tumor-bearing mice and cooperated with dexamethasone and dasatinib (a dual Src/Abl kinase inhibitor) treatment to enhance cytotoxic treatment. In clinical specimens, high levels of ADAM9 and CDCP1 correlated with poor prognosis and high risk of mortality in patients with lung cancer. Moreover, ADAM9 levels in brain metastases derived from lung tumors were relatively higher than the levels observed in primary lung tumors. Our results show how ADAM9 regulates lung cancer metastasis to the brain by facilitating the tPA-mediated cleavage of CDCP1, with potential implications to target this network as a strategy to prevent or treat brain metastatic disease.
Collapse
Affiliation(s)
- Chen-Yuan Lin
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan. Division of Hematology and Oncology, China Medical University Hospital, Taichung, Taiwan
| | - Hung-Jen Chen
- Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Cheng-Chung Huang
- Center for Molecular Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Liang-Chuan Lai
- Graduate Institute of Physiology, National Taiwan University, Taipei, Taiwan
| | - Tzu-Pin Lu
- YongLin Biomedical Engineering Center, National Taiwan University, Taipei, Taiwan
| | - Guan-Chin Tseng
- Department of Pathology, China Medical University Hospital, Taichung, Taiwan
| | - Ting-Ting Kuo
- Center for Molecular Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Qian-Yu Kuok
- Center for Molecular Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Jennifer L Hsu
- Center for Molecular Medicine, China Medical University Hospital, Taichung, Taiwan. Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas. Department of Biotechnology, Asia University, Taichung, Taiwan
| | - Shian-Ying Sung
- PhD Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Mien-Chie Hung
- Center for Molecular Medicine, China Medical University Hospital, Taichung, Taiwan. Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas. Department of Biotechnology, Asia University, Taichung, Taiwan. Graduate Institute for Cancer Biology, China Medical University, Taichung, Taiwan.
| | - Yuh-Pyng Sher
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan. Center for Molecular Medicine, China Medical University Hospital, Taichung, Taiwan.
| |
Collapse
|
45
|
Harris AJ, Thompson AR, Whyte MK, Walmsley SR. HIF-mediated innate immune responses: cell signaling and therapeutic implications. HYPOXIA 2014; 2:47-58. [PMID: 27774466 PMCID: PMC5045056 DOI: 10.2147/hp.s50269] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Leukocytes recruited to infected, damaged, or inflamed tissues during an immune response must adapt to oxygen levels much lower than those in the circulation. Hypoxia inducible factors (HIFs) are key mediators of cellular responses to hypoxia and, as in other cell types, HIFs are critical for the upregulation of glycolysis, which enables innate immune cells to produce adenosine triphosphate anaerobically. An increasing body of evidence demonstrates that hypoxia also regulates many other innate immunological functions, including cell migration, apoptosis, phagocytosis of pathogens, antigen presentation and production of cytokines, chemokines, and angiogenic and antimicrobial factors. Many of these functions are mediated by HIFs, which are not only stabilized posttranslationally by hypoxia, but also transcriptionally upregulated by inflammatory signals. Here, we review the role of HIFs in the responses of innate immune cells to hypoxia, both in vitro and in vivo, with a particular focus on myeloid cells, on which the majority of studies have so far been carried out.
Collapse
Affiliation(s)
- Alison J Harris
- Academic Unit of Respiratory Medicine, Department of Infection and Immunity, University of Sheffield, Sheffield, UK
| | - Aa Roger Thompson
- Academic Unit of Respiratory Medicine, Department of Infection and Immunity, University of Sheffield, Sheffield, UK
| | - Moira Kb Whyte
- Academic Unit of Respiratory Medicine, Department of Infection and Immunity, University of Sheffield, Sheffield, UK
| | - Sarah R Walmsley
- Academic Unit of Respiratory Medicine, Department of Infection and Immunity, University of Sheffield, Sheffield, UK
| |
Collapse
|
46
|
Schaible EV, Windschügl J, Bobkiewicz W, Kaburov Y, Dangel L, Krämer T, Huang C, Sebastiani A, Luh C, Werner C, Engelhard K, Thal SC, Schäfer MK. 2-Methoxyestradiol confers neuroprotection and inhibits a maladaptive HIF-1α response after traumatic brain injury in mice. J Neurochem 2014; 129:940-54. [DOI: 10.1111/jnc.12708] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2014] [Revised: 02/27/2014] [Accepted: 03/05/2014] [Indexed: 12/17/2022]
Affiliation(s)
- Eva-Verena Schaible
- Department of Anesthesiology; University Medical Center; Johannes Gutenberg-University; Mainz Germany
| | - Julia Windschügl
- Department of Anesthesiology; University Medical Center; Johannes Gutenberg-University; Mainz Germany
| | - Wiesia Bobkiewicz
- Department of Anesthesiology; University Medical Center; Johannes Gutenberg-University; Mainz Germany
| | - Yordan Kaburov
- Department of Anesthesiology; University Medical Center; Johannes Gutenberg-University; Mainz Germany
| | - Larissa Dangel
- Department of Anesthesiology; University Medical Center; Johannes Gutenberg-University; Mainz Germany
- Focus Program Translational Neuroscience (FTN); Johannes Gutenberg-University; Mainz Germany
| | - Tobias Krämer
- Department of Anesthesiology; University Medical Center; Johannes Gutenberg-University; Mainz Germany
| | - Changsheng Huang
- Department of Anesthesiology; University Medical Center; Johannes Gutenberg-University; Mainz Germany
| | - Anne Sebastiani
- Department of Anesthesiology; University Medical Center; Johannes Gutenberg-University; Mainz Germany
| | - Clara Luh
- Department of Anesthesiology; University Medical Center; Johannes Gutenberg-University; Mainz Germany
| | - Christian Werner
- Department of Anesthesiology; University Medical Center; Johannes Gutenberg-University; Mainz Germany
- Focus Program Translational Neuroscience (FTN); Johannes Gutenberg-University; Mainz Germany
| | - Kristin Engelhard
- Department of Anesthesiology; University Medical Center; Johannes Gutenberg-University; Mainz Germany
- Focus Program Translational Neuroscience (FTN); Johannes Gutenberg-University; Mainz Germany
| | - Serge C. Thal
- Department of Anesthesiology; University Medical Center; Johannes Gutenberg-University; Mainz Germany
| | - Michael K.E. Schäfer
- Department of Anesthesiology; University Medical Center; Johannes Gutenberg-University; Mainz Germany
- Focus Program Translational Neuroscience (FTN); Johannes Gutenberg-University; Mainz Germany
| |
Collapse
|
47
|
Ikeda Y, Tsuchiya H, Hama S, Kajimoto K, Kogure K. Resistin regulates the expression of plasminogen activator inhibitor-1 in 3T3-L1 adipocytes. Biochem Biophys Res Commun 2014; 448:129-33. [PMID: 24667608 DOI: 10.1016/j.bbrc.2014.03.076] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Accepted: 03/17/2014] [Indexed: 11/27/2022]
Abstract
Resistin and plasminogen activator inhibitor-1 (PAI-1) are adipokines, which are secreted from adipocytes. Increased plasma resistin and PAI-1 levels aggravate metabolic syndrome through exacerbation of insulin resistance and induction of chronic inflammation. However, the relationship between resistin and PAI-1 gene expression remains unclear. Previously, we found that resistin regulates lipid metabolism via carbohydrate responsive element-binding protein (ChREBP) during adipocyte maturation (Ikeda et al., 2013) [6]. In this study, to clarify the relationship between expression of resistin and PAI-1, PAI-1 expression in differentiated 3T3-L1 adipocytes was measured after transfection with anti-resistin siRNA. We found that PAI-1 gene expression and secreted PAI-1 protein were significantly decreased by resistin knockdown. Furthermore, phosphorylation of Akt, which can inhibit PAI-1 expression, was accelerated and the activity of protein phosphatase 2A (PP2A) was suppressed in resistin knockdown 3T3-L1 adipocytes. In addition, the expression of glucose transporter type 4, a ChREBP target gene, was reduced and was associated with inhibition of PP2A. The addition of culture medium collected from COS7 cells transfected with a resistin expression plasmid rescued the suppression of PAI-1 expression in resistin knockdown 3T3-L1 adipocytes. Our findings suggest that resistin regulates PAI-1 expression in 3T3-L1 adipocytes via Akt phosphorylation.
Collapse
Affiliation(s)
- Yoshito Ikeda
- Department of Biophysical Chemistry, Kyoto Pharmaceutical University, Kyoto 607-8414, Japan
| | - Hiroyuki Tsuchiya
- Department of Biophysical Chemistry, Kyoto Pharmaceutical University, Kyoto 607-8414, Japan
| | - Susumu Hama
- Department of Biophysical Chemistry, Kyoto Pharmaceutical University, Kyoto 607-8414, Japan
| | - Kazuaki Kajimoto
- Laboratory of Innovative Nanomedicine, Graduate School of Pharmaceutical Sciences, Hokkaido University, Hokkaido 060-0808, Japan
| | - Kentaro Kogure
- Department of Biophysical Chemistry, Kyoto Pharmaceutical University, Kyoto 607-8414, Japan.
| |
Collapse
|
48
|
Famakin BM, Mou Y, Johnson K, Spatz M, Hallenbeck J. A new role for downstream Toll-like receptor signaling in mediating immediate early gene expression during focal cerebral ischemia. J Cereb Blood Flow Metab 2014; 34:258-67. [PMID: 24301291 PMCID: PMC3915199 DOI: 10.1038/jcbfm.2013.182] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Revised: 08/16/2013] [Accepted: 09/20/2013] [Indexed: 12/20/2022]
Abstract
To better understand the role of downstream Toll-like receptor (TLR) signaling during acute cerebral ischemia, we performed cDNA microarrays, on brain RNA, and cytokine arrays, on serum, from wild type (WT), MyD88-/- and TRIF-mutant mice, at baseline and following permanent middle cerebral artery occlusion (pMCAO). The acute stress response pathway was among the top pathways identified by Ingenuity Pathway Analysis of microarray data. We used real-time polymerase chain reaction to confirm the expression of four immediate early genes; EGR1, EGR2, ARC, Nurr77, in this pathway, and insulin degrading enzyme (IDE). Compared to WT, baseline immediate early gene expression was increased up to10-fold in MyD88-/- and TRIF-mutant mice. However, following pMCAO, immediate early gene expression remained unchanged, from this elevated baseline in these mice, but increased up to 12-fold in WT. Furthermore, expression of IDE, which also degrades β-amyloid, decreased significantly only in TRIF-mutant mice. Finally, sE-Selectin, sICAM, sVCAM-1, and MMP-9 levels were significantly decreased only in MyD88-/- compared with WT mice. We thus report a new role for downstream TLR signaling in immediate early gene expression during acute cerebral ischemia. We also show that the TRIF pathway regulates IDE expression; a major enzyme that clears β-amyloid from the brain.
Collapse
Affiliation(s)
- Bolanle M Famakin
- National Institute of Neurological Disorders and Stroke, Stroke Branch, National Institutes of Health, Bethesda, Maryland, USA
| | - Yongshan Mou
- National Institute of Neurological Disorders and Stroke, Stroke Branch, National Institutes of Health, Bethesda, Maryland, USA
| | - Kory Johnson
- National Institute of Neurological Disorders & Stroke, Section on Bioinformatics, Information Technology & Bioinformatics Program, National Institutes of Health, Bethesda, Maryland, USA
| | - Maria Spatz
- National Institute of Neurological Disorders and Stroke, Stroke Branch, National Institutes of Health, Bethesda, Maryland, USA
| | - John Hallenbeck
- National Institute of Neurological Disorders and Stroke, Stroke Branch, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
49
|
Nelis H, D'Herde K, Goossens K, Vandenberghe L, Leemans B, Forier K, Smits K, Braeckmans K, Peelman L, Van Soom A. Equine oviduct explant culture: a basic model to decipher embryo–maternal communication. Reprod Fertil Dev 2014; 26:954-66. [DOI: 10.1071/rd13089] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Accepted: 06/18/2013] [Indexed: 12/27/2022] Open
Abstract
Equine embryos remain for 6 days in the oviduct and thus there is a need for an in vitro model to study embryo–oviductal interactions in the horse, since this subtle way of communication is very difficult to analyse in vivo. Until now, no equine oviduct explant culture model has been characterised both morphologically and functionally. Therefore, we established a culture system for equine oviduct explants that maintained epithelial morphology during 6 days of culture, as revealed by light microscopy and transmission electron microscopy. We demonstrated the presence of highly differentiated, tall columnar, pseudostratified epithelium with basal nuclei, numerous nucleoli, secretory granules and apical cilia, which is very similar to the in vivo situation. Both epithelium and stromal cells originating from the lamina propria are represented in the explants. Moreover, at least 98% of the cells remained membrane intact and fewer than 2% of the cells were apoptotic after 6 days of culture. Although dark-cell degeneration, which is a hypoxia-related type of cell death, was observed in the centre of the explants, quantitative real-time PCR failed to detect upregulation of the hypoxia-related marker genes HIF1A, VEGFA, uPA, GLUT1 and PAI1. Since the explants remained morphologically and functionally intact and since the system is easy to set up, it appears to be an excellent tool for proteome, transcriptome and miRNome analysis in order to unravel embryo–maternal interactions in the horse.
Collapse
|
50
|
Kim JI, Jung KJ, Jang HS, Park KM. Gender-specific role of HDAC11 in kidney ischemia- and reperfusion-induced PAI-1 expression and injury. Am J Physiol Renal Physiol 2013; 305:F61-70. [DOI: 10.1152/ajprenal.00015.2013] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Male gender and the male hormone testosterone increase susceptibility to kidney ischemia and reperfusion (I/R) injury, which is associated with inflammatory responses. Possible involvement of histone deacetylase (HDAC) in inflammatory responses has been suggested. We investigated the gender-specific role of HDACs in plasminogen activator inhibitor type-1 (PAI-1) expression and I/R injury. PAI-1 inhibition protected the kidney from I/R-induced inflammation and functional loss. Among HDACs, only HDAC11 negatively regulated PAI-1 expression in I/R-subjected kidney gender specifically and lipopolysaccharide (LPS)-stimulated mouse monocytes/macrophages. HDAC11 gene silencing increased PAI-1 expression. Chromatin immunoprecipitation assay confirmed binding of HDAC11 to the promoter region of PAI-1 and then release by I/R insult or LPS treatment. I/R-induced HDAC11 release was inhibited by orchiectomy and reversed by dihydrotestosterone treatment. Release of HDAC11 increased acetylation of histone H3. In conclusion, male gender and male hormones accelerate I/R-induced decreases in expression and binding of HDAC11, resulting in an increase in PAI-1 expression. These data provide important insight into gender dimorphism offering HDAC11 as a novel target for I/R injury.
Collapse
Affiliation(s)
- Jee In Kim
- Department of Anatomy and BK21, Kyungpook National University School of Medicine, Daegu, Republic of Korea; and
- Cardiovascular Research Institute, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Kyong-Jin Jung
- Department of Anatomy and BK21, Kyungpook National University School of Medicine, Daegu, Republic of Korea; and
| | - Hee-Seong Jang
- Department of Anatomy and BK21, Kyungpook National University School of Medicine, Daegu, Republic of Korea; and
| | - Kwon Moo Park
- Department of Anatomy and BK21, Kyungpook National University School of Medicine, Daegu, Republic of Korea; and
- Cardiovascular Research Institute, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| |
Collapse
|