1
|
Cao B, Yu W, Diao Z, Ma Z, Yan S, Yang L, Huang X, Yang J. KLK8: charting new territories in left ventricular hypertrophy biomarker research. Biomark Med 2025; 19:277-286. [PMID: 40171640 PMCID: PMC11980516 DOI: 10.1080/17520363.2025.2483153] [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: 02/19/2024] [Accepted: 03/19/2025] [Indexed: 04/04/2025] Open
Abstract
OBJECTIVE This study investigates the diagnostic potential of serum Kallikrein-related peptidase 8 (KLK8) in detecting left ventricular hypertrophy (LVH). METHODS A total of 62 hypertensive patients with LVH, 60 without LVH, and 60 healthy controls were analyzed. LVH was defined by echocardiography using LVMI thresholds (>95 g/m2 for females, >115 g/m2 for males). Serum KLK8 levels were measured via ELISA, and receiver operating characteristic (ROC) curve analysis assessed its diagnostic performance. RESULTS KLK8 levels were significantly higher in hypertensive patients with LVH (8.59 ± 1.59 ng/mL) than in those without LVH (5.80 ± 1.35 ng/mL) and healthy controls (3.68 ± 0.82 ng/mL). KLK8 positively correlated with blood pressure and cardiac structural parameters, including IVST, LVPWT, and LVEDD. ROC analysis revealed high sensitivity and specificity, indicating KLK8's potential as a biomarker for early LVH detection in hypertension. CONCLUSION In hypertensive patients, KLK8 demonstrates good diagnostic value in predicting LVH.
Collapse
Affiliation(s)
- Buqing Cao
- Department of Laboratory Medicine, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, China
| | - Wenhong Yu
- Department of Laboratory Medicine, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, China
| | - Zhihong Diao
- Department of Laboratory Medicine, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, China
| | - Zhenli Ma
- Department of Laboratory Medicine, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, China
| | - Shineng Yan
- Department of Laboratory Medicine, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, China
| | - Lihua Yang
- Department of Laboratory Medicine, The 923rd Hospital of the Joint Logistics Support Force of the People’s Liberation Army of China, Nanning, China
| | - Xiaoqun Huang
- Department of Laboratory Medicine, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, China
| | - Jingmei Yang
- Department of Laboratory Medicine, The 923rd Hospital of the Joint Logistics Support Force of the People’s Liberation Army of China, Nanning, China
| |
Collapse
|
2
|
Edwards CV, Ferri GM, Villegas-Galaviz J, Ghosh S, Singh Bawa P, Wang F, Klimtchuk E, Ajayi TB, Morgan GJ, Prokaeva T, Staron A, Ruberg FL, Sanchorawala V, Giadone RM, Murphy GJ. Abnormal global longitudinal strain and reduced serum inflammatory markers in cardiac AL amyloidosis patients without significant amyloid fibril deposition. Amyloid 2025:1-14. [PMID: 40134188 DOI: 10.1080/13506129.2025.2478397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 03/05/2025] [Accepted: 03/06/2025] [Indexed: 03/27/2025]
Abstract
BACKGROUND Cardiac dysfunction in AL amyloidosis is thought to be partly related to the direct impact of AL LCs on cardiomyocyte function, with the degree of dysfunction at diagnosis as a major determinant of clinical outcomes. Nonetheless, mechanisms underlying LC-induced myocardial toxicity remain unclear. METHODS We identified gene expression changes correlating with human cardiac cell exposure to cardiomyopathy-associated AL LCs. We then confirmed these findings in a clinical dataset focusing on clinical parameters associated with pathways dysregulated at the gene expression level. RESULTS Upon exposure to cardiomyopathy-associated AL LCs, cardiac cells exhibited gene expression changes related to myocardial contractile function and inflammation, leading us to hypothesise that there could be clinically detectable changes in global longitudinal strain (GLS) on echocardiogram and serum inflammatory markers in patients. Thus, we identified 29 patients with normal interventricular septum diameter (IVSd) but abnormal cardiac biomarkers, suggestive of LC-induced cardiac dysfunction. These patients display early cardiac biomarker staging, abnormal GLS, and significantly reduced serum inflammatory markers compared to patients with clinically evident amyloid fibril deposition. CONCLUSION Collectively, our findings highlight early molecular and functional signatures of cardiac AL amyloidosis, with potential impact for developing improved patient biomarkers and novel therapeutics.
Collapse
Affiliation(s)
- Camille V Edwards
- Section of Hematology and Oncology, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine and Boston Medical Center, Boston, Massachusetts, USA
- Amyloidosis Center, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
- Center for Regenerative Medicine, Boston University Chobanian & Avedisian School of Medicine and Boston Medical Center, Boston, Massachusetts, USA
| | - Grace M Ferri
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine and Boston Medical Center, Boston, Massachusetts, USA
| | - Josue Villegas-Galaviz
- Amyloidosis Center, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
| | - Sabrina Ghosh
- Center for Regenerative Medicine, Boston University Chobanian & Avedisian School of Medicine and Boston Medical Center, Boston, Massachusetts, USA
| | - Pushpinder Singh Bawa
- Center for Regenerative Medicine, Boston University Chobanian & Avedisian School of Medicine and Boston Medical Center, Boston, Massachusetts, USA
| | - Feiya Wang
- Center for Regenerative Medicine, Boston University Chobanian & Avedisian School of Medicine and Boston Medical Center, Boston, Massachusetts, USA
| | - Elena Klimtchuk
- Amyloidosis Center, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
| | - Tinuola B Ajayi
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine and Boston Medical Center, Boston, Massachusetts, USA
| | - Gareth J Morgan
- Amyloidosis Center, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
| | - Tatiana Prokaeva
- Amyloidosis Center, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
| | - Andrew Staron
- Section of Hematology and Oncology, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine and Boston Medical Center, Boston, Massachusetts, USA
- Amyloidosis Center, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
| | - Frederick L Ruberg
- Amyloidosis Center, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
- Section of Cardiovascular Medicine, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine and Boston Medical Center, Boston, Massachusetts, USA
| | - Vaishali Sanchorawala
- Section of Hematology and Oncology, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine and Boston Medical Center, Boston, Massachusetts, USA
- Amyloidosis Center, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
| | - Richard M Giadone
- Center for Regenerative Medicine, Boston University Chobanian & Avedisian School of Medicine and Boston Medical Center, Boston, Massachusetts, USA
| | - George J Murphy
- Section of Hematology and Oncology, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine and Boston Medical Center, Boston, Massachusetts, USA
- Center for Regenerative Medicine, Boston University Chobanian & Avedisian School of Medicine and Boston Medical Center, Boston, Massachusetts, USA
| |
Collapse
|
3
|
Song J, Du J, Zhao Q, Gao Y, Tan X, Cong B. KLK8 modulates macrophage function following myocardial infarction by promoting the paracrine of epidermal growth factor from cardiac fibroblasts. Life Sci 2025; 364:123445. [PMID: 39914589 DOI: 10.1016/j.lfs.2025.123445] [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: 10/12/2024] [Revised: 01/26/2025] [Accepted: 02/03/2025] [Indexed: 02/17/2025]
Abstract
AIMS Tissue kallikrein-related peptidase 8 (KLK8) plays a significant role in the regulation of cardiac remodeling following myocardial infarction (MI). However, the impact of KLK8 on macrophage (MΦ) function in the context of MI remains to be elucidated. MATERIALS AND METHODS MI was induced through the ligation of the left anterior descending coronary artery for a duration of 1 h, followed by reperfusion. The morphological and molecular alterations in the heart were assessed at 24 h and 14 days post-ischemic injury. Adult rat cardiac fibroblasts and bone marrow-derived macrophages were employed to explore the underlying molecular mechanisms in vitro. KEY FINDINGS In the acute phase of MI (24 h post-MI), KLK8 was observed to diminish the inflammatory response and mitigate tissue damage within the ischemic ventricle. Conversely, during the reparative phase of MI (14 days post-MI), KLK8 was found to enhance the accumulation of the M2 MΦs, elevate pro-fibrotic factors, and intensify cardiac fibrosis. The in vitro analysis revealed that KLK8 did not exert a direct effect on MΦs; rather, it facilitated the paracrine secretion of epidermal growth factor (EGF) from the cardiac fibroblasts. This EGF may play a role in inhibiting the pro-inflammatory activation of the MΦs and promoting their polarization towards the M2 phenotype under conditions of inflammatory stress. SIGNIFICANCE In summary, KLK8 modulates MΦ function through the paracrine of EGF derived from cardiac fibroblasts, which may have implications for cardiac injury and remodeling following MI.
Collapse
Affiliation(s)
- Jinchao Song
- Department of Anesthesiology, Shidong Hospital Affiliated to the University of Shanghai for Science and Technology, Shanghai, China; Department of Physiology, College of Basic Medical Sciences, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Jiankui Du
- Department of Physiology, College of Basic Medical Sciences, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Qian Zhao
- College of Basic Medical Sciences, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Yuan Gao
- Department of Anesthesiology, Shidong Hospital Affiliated to the University of Shanghai for Science and Technology, Shanghai, China
| | - Xing Tan
- Department of Physiology, College of Basic Medical Sciences, Naval Medical University (Second Military Medical University), Shanghai, China; Department of Marine Biomedicine and Polar Medicine, Naval Medical Center, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Binhai Cong
- Department of Physiology, College of Basic Medical Sciences, Naval Medical University (Second Military Medical University), Shanghai, China.
| |
Collapse
|
4
|
Sun Y, Hu Z, Han J, Li G. SP1-mediated transcriptional repression of SFRP5 is correlated with cardiac fibroblast activation and atrial myocyte apoptosis in the development of atrial fibrillation. Exp Cell Res 2024; 443:114326. [PMID: 39536929 DOI: 10.1016/j.yexcr.2024.114326] [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: 07/08/2024] [Revised: 11/05/2024] [Accepted: 11/10/2024] [Indexed: 11/16/2024]
Abstract
Secreted frizzled related protein 5 (SFRP5) is a recognized cardioprotective protein with diminished expression in atrial fibrillation (AF). This study investigates SFRP5's function in AF-related cardiac fibrosis and cardiomyocyte apoptosis, exploring the underlying dysregulation causes. Utilizing C57BL/6 mice, mouse cardiac fibroblasts (CFs), and HC-1 mouse atrial myocyte cell line, AF models were induced by angiotensin Ⅱ (Ang Ⅱ). SFRP5 levels were consistently decreased in plasma samples from clinical patients, modeled mice, and CF culture supernatants. Treatment with recombinant SFRP5 restored its levels, mitigating Ang Ⅱ-induced AF in mice and ameliorating atrial tissue fibrosis and oxidative stress. In vitro, SFRP5 recombinant protein suppressed CF activation and fibrosis-related markers. The study identified Sp1 transcription factor (SP1) binding to the SFRP5 promoter, causing transcriptional repression. SP1 knockdown reinstated SFRP5 levels in mice and CFs, thus suppressing fibrosis. Additionally, SP1 knockdown attenuated Ang Ⅱ-induced apoptosis in HC-1 cells, but this effect was counteracted by concurrent SFRP5 knockdown. In conclusion, this investigation underscores that SP1 mediates SFRP5 loss during AF by transcriptional repression, contributing to fibrosis and myocyte apoptosis. These findings illuminate potential therapeutic interventions targeting the SFRP5-SP1 axis in AF-related cardiac complications.
Collapse
Affiliation(s)
- Yanyan Sun
- Department of Cardiology, Henan Provincial Chest Hospital, Zhengzhou, 450000, Henan, PR China.
| | - Zhenzhen Hu
- Department of Cardiology, Henan Provincial Chest Hospital, Zhengzhou, 450000, Henan, PR China
| | - Jie Han
- Department of Cardiology, Henan Provincial Chest Hospital, Zhengzhou, 450000, Henan, PR China
| | - Gang Li
- Department of Cardiology, Henan Provincial Chest Hospital, Zhengzhou, 450000, Henan, PR China
| |
Collapse
|
5
|
Wenta T, Nastaly P, Lipinska B, Manninen A. Remodeling of the extracellular matrix by serine proteases as a prerequisite for cancer initiation and progression. Matrix Biol 2024; 134:197-219. [PMID: 39500383 DOI: 10.1016/j.matbio.2024.10.007] [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: 04/30/2024] [Revised: 10/16/2024] [Accepted: 10/31/2024] [Indexed: 11/25/2024]
Abstract
The extracellular matrix (ECM) serves as a physical scaffold for tissues that is composed of structural proteins such as laminins, collagens, proteoglycans and fibronectin, forming a three dimensional network, and a wide variety of other matrix proteins with ECM-remodeling and signaling functions. The activity of ECM-associated signaling proteins is tightly regulated. Thus, the ECM serves as a reservoir for water and growth regulatory signals. The ECM architecture is dynamically modulated by multiple serine proteases that process both structural and signaling proteins to regulate physiological processes such as organogenesis and tissue homeostasis but they also contribute to pathological events, especially cancer progression. Here, we review the current literature regarding the role of ECM remodeling by serine proteases (KLKs, uPA, furin, HtrAs, granzymes, matriptase, hepsin) in tumorigenesis.
Collapse
Affiliation(s)
- Tomasz Wenta
- Department of General and Medical Biochemistry, Faculty of Biology, University of Gdansk, Poland.
| | - Paulina Nastaly
- Laboratory of Translational Oncology, Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Gdansk, Poland
| | - Barbara Lipinska
- Department of General and Medical Biochemistry, Faculty of Biology, University of Gdansk, Poland
| | - Aki Manninen
- Disease Networks Research Unit, Faculty of Biochemistry and Molecular Medicine & Biocenter Oulu, University of Oulu, Oulu, Finland.
| |
Collapse
|
6
|
Zhang M, Zhang Y, He J, Wang X, Wang Y, Li L, Tao L, Zhang M, Shen X. Gastrodin attenuates diabetic cardiomyopathy characterized by myocardial fibrosis by inhibiting the KLK8-PAR1 signaling axis. Chin Med 2024; 19:164. [PMID: 39578836 PMCID: PMC11583739 DOI: 10.1186/s13020-024-01035-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Accepted: 11/07/2024] [Indexed: 11/24/2024] Open
Abstract
BACKGROUND Diabetic cardiomyopathy (DCM), characterized by myocardial fibrosis, is a major cause of mortality and morbidity in diabetic patients; the inhibition of cardiac fibrosis is a fundamental strategy for treating DCM. Gastrodin (GAS), a compound extracted from Gastrodia elata protects against DCM, but the molecular mechanism underlying its antifibrotic effect has not been elucidated. METHODS In vivo, the effects of GAS were investigated using C57BL/6 mice with DCM, which was induced by administering a high-sugar, high-fat (HSF) diet and streptozotocin (STZ). We assessed the cardiac function in these mice and detected histopathological changes in their hearts and the degree of cardiac fibrosis. In vitro, neonatal rat cardiac fibroblasts (CFs) were transformed into myofibroblasts by exposing them to high glucose combined with high palmitic acid (HG-PA), and CFs were induced by pEX-1 (pGCMV/MCS/EGFP/Neo) plasmid-mediated overexpression of KLK8, which contains the rat KLK8 gene. The KLK8 siRNA was knocked down to study the effects of GAS on CF differentiation, collagen synthesis, and cell migration by specific mechanisms of action of GAS. RESULTS GAS attenuated pathological changes in the hearts of DCM mice, rescued impaired cardiac function, and attenuated cardiac fibrosis. Additionally, the results of molecular docking analysis showed that GAS binds to kinin-releasing enzyme-related peptidase 8 (KLK8) to inhibit the increase in protease-activated receptor-1 (PAR-1), thus attenuating myocardial fibrosis. Specifically, GAS attenuated the transformation of neonatal rat CFs to myofibroblasts exposed to HG-PA. Overexpressing KLK8 promoted CF differentiation, collagen synthesis, and cell migration, and KLK8 siRNA attenuated HG-PA-induced CF differentiation, collagen synthesis, and cell migration. Further studies revealed that a PAR-1 antagonist, but not a PAR-2 antagonist, could attenuate CF differentiation, collagen synthesis, and cell migration. Additionally, GAS inhibited KLK8 upregulation and PAR1 activation, thus blocking the differentiation, collagen synthesis, and cell migration of HG-PA-exposed CFs and triggering TGF-β1/Smad3 signaling. CONCLUSION GAS alleviated pathological changes in the hearts of DCM model mice induced by an HSF diet combined with STZ. KLK8 mediated HG-PA-induced differentiation, collagen synthesis, and the migration of CFs. GAS attenuated the differentiation, collagen synthesis, and migration of CFs by inhibiting the KLK8-PAR1 signaling axis, a process in which TGF-β1 and Smad3 are involved.
Collapse
Affiliation(s)
- MingShan Zhang
- The State Key Laboratory of Functions and Applications of Medicinal Plants, The Key Laboratory of Endemic and Ethnic Diseases of Ministry of Education), Guizhou Medical University, No.6 Ankang Avenue, Guian New District, Guiyang, 561113, Guizhou, China
- Department of Physiology, School of Basic Medical Sciences, Guizhou Medical University, Guian New District, Guiyang , 561113, Guizhou, China
- The Department of Pharmacology of Materia Medica (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The high educational key laboratory of Guizhou province for natural medicianl Pharmacology and Druggability), Guian New District, Guiyang, 561113, Guizhou, China
| | - YuFei Zhang
- The State Key Laboratory of Functions and Applications of Medicinal Plants, The Key Laboratory of Endemic and Ethnic Diseases of Ministry of Education), Guizhou Medical University, No.6 Ankang Avenue, Guian New District, Guiyang, 561113, Guizhou, China
- The Department of Pharmacology of Materia Medica (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The high educational key laboratory of Guizhou province for natural medicianl Pharmacology and Druggability), Guian New District, Guiyang, 561113, Guizhou, China
| | - JingGang He
- The State Key Laboratory of Functions and Applications of Medicinal Plants, The Key Laboratory of Endemic and Ethnic Diseases of Ministry of Education), Guizhou Medical University, No.6 Ankang Avenue, Guian New District, Guiyang, 561113, Guizhou, China
- The Department of Pharmacology of Materia Medica (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The high educational key laboratory of Guizhou province for natural medicianl Pharmacology and Druggability), Guian New District, Guiyang, 561113, Guizhou, China
| | - XinRui Wang
- The State Key Laboratory of Functions and Applications of Medicinal Plants, The Key Laboratory of Endemic and Ethnic Diseases of Ministry of Education), Guizhou Medical University, No.6 Ankang Avenue, Guian New District, Guiyang, 561113, Guizhou, China
- The Department of Pharmacology of Materia Medica (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The high educational key laboratory of Guizhou province for natural medicianl Pharmacology and Druggability), Guian New District, Guiyang, 561113, Guizhou, China
| | - YinYin Wang
- The State Key Laboratory of Functions and Applications of Medicinal Plants, The Key Laboratory of Endemic and Ethnic Diseases of Ministry of Education), Guizhou Medical University, No.6 Ankang Avenue, Guian New District, Guiyang, 561113, Guizhou, China
- The Department of Pharmacology of Materia Medica (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The high educational key laboratory of Guizhou province for natural medicianl Pharmacology and Druggability), Guian New District, Guiyang, 561113, Guizhou, China
| | - LinYan Li
- The State Key Laboratory of Functions and Applications of Medicinal Plants, The Key Laboratory of Endemic and Ethnic Diseases of Ministry of Education), Guizhou Medical University, No.6 Ankang Avenue, Guian New District, Guiyang, 561113, Guizhou, China
- The Department of Pharmacology of Materia Medica (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The high educational key laboratory of Guizhou province for natural medicianl Pharmacology and Druggability), Guian New District, Guiyang, 561113, Guizhou, China
| | - Ling Tao
- The State Key Laboratory of Functions and Applications of Medicinal Plants, The Key Laboratory of Endemic and Ethnic Diseases of Ministry of Education), Guizhou Medical University, No.6 Ankang Avenue, Guian New District, Guiyang, 561113, Guizhou, China.
- The Department of Pharmacology of Materia Medica (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The high educational key laboratory of Guizhou province for natural medicianl Pharmacology and Druggability), Guian New District, Guiyang, 561113, Guizhou, China.
| | - Min Zhang
- Department of Physiology, School of Basic Medical Sciences, Guizhou Medical University, Guian New District, Guiyang , 561113, Guizhou, China.
| | - Xiangchun Shen
- The State Key Laboratory of Functions and Applications of Medicinal Plants, The Key Laboratory of Endemic and Ethnic Diseases of Ministry of Education), Guizhou Medical University, No.6 Ankang Avenue, Guian New District, Guiyang, 561113, Guizhou, China.
- The Department of Pharmacology of Materia Medica (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The high educational key laboratory of Guizhou province for natural medicianl Pharmacology and Druggability), Guian New District, Guiyang, 561113, Guizhou, China.
| |
Collapse
|
7
|
Alanazi AH, Selim MS, Yendamuri MR, Zhang D, Narayanan SP, Somanath PR. The impact of diabetes mellitus on blood-tissue barrier regulation and vascular complications: Is the lung different from other organs? Tissue Barriers 2024:2386183. [PMID: 39072526 DOI: 10.1080/21688370.2024.2386183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 07/12/2024] [Accepted: 07/16/2024] [Indexed: 07/30/2024] Open
Abstract
Diabetes Mellitus presents a formidable challenge as one of the most prevalent and complex chronic diseases, exerting significant strain on both patients and the world economy. It is recognized as a common comorbidity among severely ill individuals, often leading to a myriad of micro- and macro-vascular complications. Despite extensive research dissecting the pathophysiology and molecular mechanisms underlying vascular complications of diabetes, relatively little attention has been paid to potential lung-related complications. This review aims to illuminate the impact of diabetes on prevalent respiratory diseases, including chronic obstructive pulmonary disease (COPD), acute respiratory distress syndrome (ARDS), idiopathic pulmonary fibrosis (IPF), tuberculosis (TB), pneumonia infections, and asthma, and compare the vascular complications with other vascular beds. Additionally, we explore the primary mechanistic pathways contributing to these complications, such as the expression modulation of blood-tissue-barrier proteins, resulting in increased paracellular and transcellular permeability, and compromised immune responses rendering diabetes patients more susceptible to infections. The activation of inflammatory pathways leading to cellular injury and hastening the onset of these respiratory complications is also discussed.
Collapse
Affiliation(s)
- Abdulaziz H Alanazi
- Clinical and Experimental Therapeutics, University of Georgia, Augusta, GA, USA
- Charlie Norwood VA Medical Center, Augusta, GA, USA
- Department of Clinical Practice, College of Pharmacy, Northern Border University, Rafha, Saudi Arabia
| | - Mohamed S Selim
- Clinical and Experimental Therapeutics, University of Georgia, Augusta, GA, USA
- Charlie Norwood VA Medical Center, Augusta, GA, USA
| | - Manyasreeprapti R Yendamuri
- Clinical and Experimental Therapeutics, University of Georgia, Augusta, GA, USA
- Charlie Norwood VA Medical Center, Augusta, GA, USA
| | - Duo Zhang
- Clinical and Experimental Therapeutics, University of Georgia, Augusta, GA, USA
- Charlie Norwood VA Medical Center, Augusta, GA, USA
| | - S Priya Narayanan
- Clinical and Experimental Therapeutics, University of Georgia, Augusta, GA, USA
- Charlie Norwood VA Medical Center, Augusta, GA, USA
| | - Payaningal R Somanath
- Clinical and Experimental Therapeutics, University of Georgia, Augusta, GA, USA
- Charlie Norwood VA Medical Center, Augusta, GA, USA
| |
Collapse
|
8
|
Ding J, Fayyaz AI, Ding Y, Liang D, Luo M. Role of Specificity Protein 1 (SP1) in Cardiovascular Diseases: Pathological Mechanisms and Therapeutic Potentials. Biomolecules 2024; 14:807. [PMID: 39062521 PMCID: PMC11274404 DOI: 10.3390/biom14070807] [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: 06/12/2024] [Revised: 07/01/2024] [Accepted: 07/05/2024] [Indexed: 07/28/2024] Open
Abstract
In mammals, specificity protein 1 (SP1) was the first Cys2-His2 zinc finger transcription factor to be isolated within the specificity protein and Krüppel-like factor (Sp/KLF) gene family. SP1 regulates gene expression by binding to Guanine-Cytosine (GC)-rich sequences on promoter regions of target genes, affecting various cellular processes. Additionally, the activity of SP1 is markedly influenced by posttranslational modifications, such as phosphorylation, acetylation, glycosylation, and proteolysis. SP1 is implicated in the regulation of apoptosis, cell hypertrophy, inflammation, oxidative stress, lipid metabolism, plaque stabilization, endothelial dysfunction, fibrosis, calcification, and other pathological processes. These processes impact the onset and progression of numerous cardiovascular disorders, including coronary heart disease, ischemia-reperfusion injury, cardiomyopathy, arrhythmia, and vascular disease. SP1 emerges as a potential target for the prevention and therapeutic intervention of cardiac ailments. In this review, we delve into the biological functions, pathophysiological mechanisms, and potential clinical implications of SP1 in cardiac pathology to offer valuable insights into the regulatory functions of SP1 in heart diseases and unveil novel avenues for the prevention and treatment of cardiovascular conditions.
Collapse
Affiliation(s)
- Jie Ding
- School of Medicine, Tongji University, Shanghai 200092, China; (J.D.); (D.L.)
- State Key Laboratory of Cardiovascular Diseases, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200120, China
- Shanghai Arrhythmia Research Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200120, China
| | - Aminah I. Fayyaz
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI 48201, USA; (A.I.F.); (Y.D.)
| | - Yuchuan Ding
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI 48201, USA; (A.I.F.); (Y.D.)
| | - Dandan Liang
- School of Medicine, Tongji University, Shanghai 200092, China; (J.D.); (D.L.)
- State Key Laboratory of Cardiovascular Diseases, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200120, China
- Shanghai Arrhythmia Research Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200120, China
| | - Ming Luo
- School of Medicine, Tongji University, Shanghai 200092, China; (J.D.); (D.L.)
| |
Collapse
|
9
|
Song J, Du J, Tan X, Chen H, Cong B. Bradykinin attenuates endothelial-mesenchymal transition following cardiac ischemia-reperfusion injury. Eur J Pharmacol 2024; 971:176556. [PMID: 38574840 DOI: 10.1016/j.ejphar.2024.176556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 04/01/2024] [Accepted: 04/02/2024] [Indexed: 04/06/2024]
Abstract
AIMS Endothelial-mesenchymal transition (EndMT) is a crucial pathological process contributing to cardiac fibrosis. Bradykinin has been found to protect the heart against fibrosis. Whether bradykinin regulates EndMT has not been determined. MATERIALS AND METHODS Rats were subjected to ligation of the left anterior descending coronary artery for 1 h and subsequent reperfusion to induce cardiac ischemia-reperfusion (IR) injury. Bradykinin (0.5 μg/h) was infused by an osmotic pump implanted subcutaneously at the onset of reperfusion. Fourteen days later, the functional, histological, and molecular analyses were performed to investigate the changes in cardiac fibrosis and EndMT. Human coronary artery endothelial cells were utilized to determine the molecular mechanisms in vitro. RESULTS Bradykinin treatment improved cardiac function and decreased fibrosis following cardiac IR injury, accompanied by ameliorated EndMT and increased nitric oxide (NO) production. In vitro experiments found that bradykinin mitigated transforming growth factor β1 (TGFβ1)-induced EndMT. Significantly, the bradykinin B2 receptor antagonist or endothelial nitric oxide synthase inhibitor abolished the effects of bradykinin on EndMT inhibition, indicating that the bradykinin B2 receptor and NO might mediate the effects of bradykinin on EndMT inhibition. CONCLUSION Bradykinin plays an essential role in the process of cardiac fibrosis. Bradykinin preserves the cellular signature of endothelial cells, preventing them from EndMT following cardiac IR injury, possibly mediated by bradykinin B2 receptor activation and NO production.
Collapse
Affiliation(s)
- Jinchao Song
- Department of Anesthesiology, Shidong Hospital Affiliated to the University of Shanghai for Science and Technology, Shanghai, China; Department of Physiology, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Jiankui Du
- Department of Physiology, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Xing Tan
- Department of Physiology, Naval Medical University (Second Military Medical University), Shanghai, China; Department of Marine Biomedicine and Polar Medicine, Naval Medical Center, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Haiyan Chen
- Department of Anesthesiology, Shidong Hospital Affiliated to the University of Shanghai for Science and Technology, Shanghai, China
| | - Binhai Cong
- Department of Physiology, Naval Medical University (Second Military Medical University), Shanghai, China.
| |
Collapse
|
10
|
Bao YN, Yang Q, Shen XL, Yu WK, Zhou L, Zhu QR, Shan QY, Wang ZC, Cao G. Targeting tumor suppressor p53 for organ fibrosis therapy. Cell Death Dis 2024; 15:336. [PMID: 38744865 PMCID: PMC11094089 DOI: 10.1038/s41419-024-06702-w] [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: 10/18/2023] [Revised: 04/18/2024] [Accepted: 04/22/2024] [Indexed: 05/16/2024]
Abstract
Fibrosis is a reparative and progressive process characterized by abnormal extracellular matrix deposition, contributing to organ dysfunction in chronic diseases. The tumor suppressor p53 (p53), known for its regulatory roles in cell proliferation, apoptosis, aging, and metabolism across diverse tissues, appears to play a pivotal role in aggravating biological processes such as epithelial-mesenchymal transition (EMT), cell apoptosis, and cell senescence. These processes are closely intertwined with the pathogenesis of fibrotic disease. In this review, we briefly introduce the background and specific mechanism of p53, investigate the pathogenesis of fibrosis, and further discuss p53's relationship and role in fibrosis affecting the kidney, liver, lung, and heart. In summary, targeting p53 represents a promising and innovative therapeutic approach for the prevention and treatment of organ fibrosis.
Collapse
Affiliation(s)
- Yi-Ni Bao
- School of Pharmacy, Zhejiang Chinese Medical University, No. 548 Binwen Road, Hangzhou, Zhejiang, 310053, China
| | - Qiao Yang
- School of Pharmacy, Zhejiang Chinese Medical University, No. 548 Binwen Road, Hangzhou, Zhejiang, 310053, China
| | - Xin-Lei Shen
- School of Pharmacy, Zhejiang Chinese Medical University, No. 548 Binwen Road, Hangzhou, Zhejiang, 310053, China
| | - Wen-Kai Yu
- School of Pharmacy, Zhejiang Chinese Medical University, No. 548 Binwen Road, Hangzhou, Zhejiang, 310053, China
| | - Li Zhou
- School of Pharmacy, Zhejiang Chinese Medical University, No. 548 Binwen Road, Hangzhou, Zhejiang, 310053, China
| | - Qing-Ru Zhu
- School of Pharmacy, Zhejiang Chinese Medical University, No. 548 Binwen Road, Hangzhou, Zhejiang, 310053, China
| | - Qi-Yuan Shan
- School of Pharmacy, Zhejiang Chinese Medical University, No. 548 Binwen Road, Hangzhou, Zhejiang, 310053, China
| | - Zhi-Chao Wang
- School of Pharmacy, Zhejiang Chinese Medical University, No. 548 Binwen Road, Hangzhou, Zhejiang, 310053, China
| | - Gang Cao
- School of Pharmacy, Zhejiang Chinese Medical University, No. 548 Binwen Road, Hangzhou, Zhejiang, 310053, China.
| |
Collapse
|
11
|
Fu TL, Li GR, Li DH, He RY, Liu BH, Xiong R, Xu CZ, Lu ZL, Song CK, Qiu HL, Wang WJ, Zou SS, Yi K, Li N, Geng Q. Mangiferin alleviates diabetic pulmonary fibrosis in mice via inhibiting endothelial-mesenchymal transition through AMPK/FoxO3/SIRT3 axis. Acta Pharmacol Sin 2024; 45:1002-1018. [PMID: 38225395 PMCID: PMC11053064 DOI: 10.1038/s41401-023-01202-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 11/17/2023] [Indexed: 01/17/2024]
Abstract
Diabetes mellitus results in numerous complications. Diabetic pulmonary fibrosis (DPF), a late pulmonary complication of diabetes, has not attracted as much attention as diabetic nephropathy and cardiomyopathy. Mangiferin (MF) is a natural small molecular compound that exhibits a variety of pharmacological effects including anti-inflammatory, anti-cancer, anti-diabetes, and anti-fibrosis effects. In this study, we investigated whether long-term diabetes shock induces DPF, and explored whether MF had a protective effect against DPF. We first examined the lung tissues and sections of 20 diabetic patients obtained from discarded lung surgical resection specimens and found that pulmonary fibrosis mainly accumulated around the pulmonary vessels, accompanied by significantly enhanced endothelial-mesenchymal transition (EndMT). We established a mouse model of DPF by STZ injections. Ten days after the final STZ injection, the mice were administered MF (20, 60 mg/kg, i.g.) every 3 days for 4 weeks, and kept feeding until 16 weeks and euthanized. We showed that pulmonary fibrotic lesions were developed in the diabetic mice, which began around the pulmonary vessels, while MF administration did not affect long-term blood glucose levels, but dose-dependently alleviated diabetes-induced pulmonary fibrosis. In human umbilical vein endothelial cells (HUVECs), exposure to high glucose (33.3 mM) induced EndMT, which was dose-dependently inhibited by treatment with MF (10, 50 μM). Furthermore, MF treatment promoted SIRT3 expression in high glucose-exposed HUVECs by directly binding to AMPK to enhance the activity of FoxO3, which finally reversed diabetes-induced EndMT. We conclude that MF attenuates DPF by inhibiting EndMT through the AMPK/FoxO3/SIRT3 axis. MF could be a potential candidate for the early prevention and treatment of DPF.
Collapse
Affiliation(s)
- Ting-Lv Fu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Guo-Rui Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Dong-Hang Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Ru-Yuan He
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Bo-Hao Liu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, 130061, China
| | - Rui Xiong
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Chen-Zhen Xu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Zi-Long Lu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Cong-Kuan Song
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Hong-Liang Qiu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Wen-Jie Wang
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Shi-Shi Zou
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Ke Yi
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Ning Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Qing Geng
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
12
|
Su Q, Huang W, Huang Y, Dai R, Chang C, Li QY, Liu H, Li Z, Zhao Y, Wu Q, Pan DG. Single-cell insights: pioneering an integrated atlas of chromatin accessibility and transcriptomic landscapes in diabetic cardiomyopathy. Cardiovasc Diabetol 2024; 23:139. [PMID: 38664790 PMCID: PMC11046823 DOI: 10.1186/s12933-024-02233-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 04/11/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND Diabetic cardiomyopathy (DCM) poses a growing health threat, elevating heart failure risk in diabetic individuals. Understanding DCM is crucial, with fibroblasts and endothelial cells playing pivotal roles in driving myocardial fibrosis and contributing to cardiac dysfunction. Advances in Multimodal single-cell profiling, such as scRNA-seq and scATAC-seq, provide deeper insights into DCM's unique cell states and molecular landscape for targeted therapeutic interventions. METHODS Single-cell RNA and ATAC data from 10x Multiome libraries were processed using Cell Ranger ARC v2.0.1. Gene expression and ATAC data underwent Seurat and Signac filtration. Differential gene expression and accessible chromatin regions were identified. Transcription factor activity was estimated with chromVAR, and Cis-coaccessibility networks were calculated using Cicero. Coaccessibility connections were compared to the GeneHancer database. Gene Ontology analysis, biological process scoring, cell-cell communication analysis, and gene-motif correlation was performed to reveal intricate molecular changes. Immunofluorescent staining utilized various antibodies on paraffin-embedded tissues to verify the findings. RESULTS This study integrated scRNA-seq and scATAC-seq data obtained from hearts of WT and DCM mice, elucidating molecular changes at the single-cell level throughout the diabetic cardiomyopathy progression. Robust and accurate clustering analysis of the integrated data revealed altered cell proportions, showcasing decreased endothelial cells and macrophages, coupled with increased fibroblasts and myocardial cells in the DCM group, indicating enhanced fibrosis and endothelial damage. Chromatin accessibility analysis unveiled unique patterns in cell types, with heightened transcriptional activity in myocardial cells. Subpopulation analysis highlighted distinct changes in cardiomyocytes and fibroblasts, emphasizing pathways related to fatty acid metabolism and cardiac contraction. Fibroblast-centered communication analysis identified interactions with endothelial cells, implicating VEGF receptors. Endothelial cell subpopulations exhibited altered gene expressions, emphasizing contraction and growth-related pathways. Candidate regulators, including Tcf21, Arnt, Stat5a, and Stat5b, were identified, suggesting their pivotal roles in DCM development. Immunofluorescence staining validated marker genes of cell subpopulations, confirming PDK4, PPARγ and Tpm1 as markers for metabolic pattern-altered cardiomyocytes, activated fibroblasts and endothelial cells with compromised proliferation. CONCLUSION Our integrated scRNA-seq and scATAC-seq analysis unveils intricate cell states and molecular alterations in diabetic cardiomyopathy. Identified cell type-specific changes, transcription factors, and marker genes offer valuable insights. The study sheds light on potential therapeutic targets for DCM.
Collapse
Affiliation(s)
- Qiang Su
- Department of Cardiology, People's Hospital of Guilin, Guilin, China
- Department of Cardiology, Jiangbin Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Wanzhong Huang
- Department of Cardiology, Jiangbin Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Yuan Huang
- Department of Cardiology, Jiangbin Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Rixin Dai
- Department of Cardiology, Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Chen Chang
- Department of Cardiology, Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Qiu-Yan Li
- Department of Cardiology, People's Hospital of Guilin, Guilin, China
| | - Hao Liu
- Institute of Bioengineering, Biotrans Technology Co., LTD, Shanghai, China
- United New Drug Research and Development Center, Biotrans Technology Co., LTD, Changsha, China
| | - Zhenhao Li
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- BoYu Intelligent Health Innovation Laboratory, Hangzhou, China
| | - Yuxiang Zhao
- Institute of Bioengineering, Biotrans Technology Co., LTD, Shanghai, China.
- United New Drug Research and Development Center, Biotrans Technology Co., LTD, Changsha, China.
| | - Qiang Wu
- Senior Department of Cardiology, the Sixth Medical Centre, Chinese PLA General Hospital, Beijing, China.
| | - Di-Guang Pan
- Department of Cardiology, People's Hospital of Guilin, Guilin, China.
| |
Collapse
|
13
|
He W, Tang M, Gu R, Wu X, Mu X, Nie X. The Role of p53 in Regulating Chronic Inflammation and PANoptosis in Diabetic Wounds. Aging Dis 2024; 16:AD.2024.0212. [PMID: 38377027 PMCID: PMC11745441 DOI: 10.14336/ad.2024.0212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Accepted: 02/12/2024] [Indexed: 02/22/2024] Open
Abstract
Diabetic wounds represent a formidable challenge in the clinical management of diabetes mellitus, markedly diminishing the patient's quality of life. These wounds arise from a multifaceted etiology, with the pathophysiological underpinnings remaining elusive and complex. Diabetes precipitates neuropathies and vasculopathies in the lower extremities, culminating in infections, ulcerations, and extensive tissue damage. The hallmarks of non-healing diabetic wounds include senescence, persistent inflammation, heightened apoptosis, and attenuated cellular proliferation. The TP53 gene, a pivotal tumor suppressor frequently silenced in human malignancies, orchestrates cellular proliferation, senescence, DNA repair, and apoptosis. While p53 is integral in cell cycle regulation, its role in initial tissue repair appears to be deleterious. In typical cutaneous wounds, p53 levels transiently dip, swiftly reverting to baseline. Yet in diabetic wounds, protracted p53 activation impedes healing via two distinct pathways: i) activating the p53-p21-Retinoblastoma (RB) axis, which halts the cell cycle, and ii) upregulating the cGAS-STING and nuclear factor-kappaB (NF-κB) cascades, instigating ferroptosis and pyroptosis. Furthermore, p53 intersects with various metabolic pathways, including glycolysis, gluconeogenesis, oxidative phosphorylation, and autophagy. In diabetic wounds, p53 may drive metabolic reprogramming, thus potentially derailing macrophage polarization. This review synthesizes case studies investigating the therapeutic modulation of p53 in diabetic wounds care. In summation, p53 modulates chronic inflammation and cellular aging within diabetic cutaneous wounds and is implicated in a novel cell death modality, encompassing ferroptosis and pyroptosis, which hinders the reparative process.
Collapse
Affiliation(s)
- Wenjie He
- College of Pharmacy, Zunyi Medical University, Zunyi 563006, China.
- Key Lab of the Basic Pharmacology of the Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563006, China.
| | - Ming Tang
- Department of Structural Biology, St. Jude Children’s Research Hospital, Memphis 38105, USA.
| | - Rifang Gu
- School Medical Office, Zunyi Medical University, Zunyi 563006, China.
| | - Xingqian Wu
- College of Pharmacy, Zunyi Medical University, Zunyi 563006, China.
- Key Lab of the Basic Pharmacology of the Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563006, China.
| | - Xinrui Mu
- College of Pharmacy, Zunyi Medical University, Zunyi 563006, China.
- Key Lab of the Basic Pharmacology of the Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563006, China.
| | - Xuqiang Nie
- College of Pharmacy, Zunyi Medical University, Zunyi 563006, China.
- Key Lab of the Basic Pharmacology of the Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563006, China.
| |
Collapse
|
14
|
Mimouni M, Lajoix AD, Desmetz C. Experimental Models to Study Endothelial to Mesenchymal Transition in Myocardial Fibrosis and Cardiovascular Diseases. Int J Mol Sci 2023; 25:382. [PMID: 38203553 PMCID: PMC10779210 DOI: 10.3390/ijms25010382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 12/21/2023] [Accepted: 12/25/2023] [Indexed: 01/12/2024] Open
Abstract
Fibrosis is a common feature of cardiovascular diseases and targets multiple organs, such as the heart and vessels. Endothelial to mesenchymal transition is a complex, vital process that occurs during embryonic formation and plays a crucial role in cardiac development. It is also a fundamental process implicated in cardiac fibrosis and repair, but also in other organs. Indeed, in numerous cardiovascular diseases, the endothelial-to-mesenchymal transition has been shown to be involved in the generation of fibroblasts that are able to produce extracellular matrix proteins such as type I collagen. This massive deposition results in tissue stiffening and organ dysfunction. To advance our understanding of this process for the development of new specific diagnostic and therapeutic strategies, it is essential to develop relevant cellular and animal models of this process. In this review, our aim was to gain an in-depth insight into existing in vitro and in vivo models of endothelial to mesenchymal transition in cardiovascular diseases with a focus on cardiac fibrosis. We discuss important parameters impacting endothelial to mesenchymal transition, and we give perspectives for the development of relevant models to decipher the underlying mechanisms and ultimately find new treatments specific to fibrosis happening in cardiovascular diseases.
Collapse
Affiliation(s)
- Mohammed Mimouni
- Biocommunication in Cardio-Metabolism (BC2M), University of Montpellier, 34000 Montpellier, France
| | - Anne-Dominique Lajoix
- Biocommunication in Cardio-Metabolism (BC2M), University of Montpellier, 34000 Montpellier, France
| | - Caroline Desmetz
- Biocommunication in Cardio-Metabolism (BC2M), University of Montpellier, 34000 Montpellier, France
| |
Collapse
|
15
|
Ji-hong Y, Yu M, Ling-hong Y, Jing-jing G, Ling-li X, Lv W, Yong-mei J. Baicalein attenuates bleomycin-induced lung fibroblast senescence and lung fibrosis through restoration of Sirt3 expression. PHARMACEUTICAL BIOLOGY 2023; 61:288-297. [PMID: 36815239 PMCID: PMC9970214 DOI: 10.1080/13880209.2022.2160767] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 11/09/2022] [Accepted: 12/15/2022] [Indexed: 06/18/2023]
Abstract
CONTEXT Fibroblast senescence was reported to contribute to the pathological development of idiopathic pulmonary fibrosis (IPF), and baicalein is reported to attenuate IPF. OBJECTIVE This study explores whether baicalein attenuates lung fibrosis by regulating lung fibroblast senescence. MATERIALS AND METHODS Institute of Cancer Research (ICR) mice were randomly assigned to control, bleomycin (BLM), baicalein and BLM + baicalein groups. Lung fibrosis was established by a single intratracheal dose of BLM (3 mg/kg). The baicalein group received baicalein orally (100 mg/kg/day). Sirtuin 3 (Sirt3) siRNA (50 μg) was injected through the tail vein once a week for 2 weeks to explore its effect on the anti-pulmonary fibrosis of baicalein. RESULTS BLM-treated mice exhibited obvious lung fibrosis and fibroblast senescence by showing increased levels of collagen deposition (27.29% vs. 4.14%), hydroxyproline (208.05 vs. 40.16 ng/mg), collagen I (25.18 vs. 9.15 μg/mg), p53, p21, p16, MCP-1, PAI-1, TNF-α, MMP-10 and MMP-12 in lung tissues, which were attenuated by baicalein. Baicalein also mitigated BLM-mediated activation of TGF-β1/Smad signalling pathway. Baicalein restored the BLM-induced downregulation of Sirt3 expression in lung tissues and silencing of Sirt3 abolished the inhibitory role of baicalein against BLM-induced lung fibrosis, fibroblast senescence and activation of TGF-β1/Smad signalling pathway. CONCLUSIONS Baicalein preserved the BLM-induced downregulation of lung Sirt3 expression, and thus the suppression of TGF-β1/Smad signalling pathway and lung fibrosis, which might provide an experimental basis for treatment of IPF.
Collapse
Affiliation(s)
- Yuan Ji-hong
- Department of Acute and Critical Care, Shanghai Seventh People’s Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ma Yu
- Department of Anesthesiology, Shanghai Baoshan Traditional Chinese Medicine-integrated Hospital, Shanghai, China
| | - Yuan Ling-hong
- Department of Acute and Critical Care, Changxing Branch of Xinhua Hospital Affiliated to School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Gong Jing-jing
- Department of Nephrology, Shanghai Seventh People’s Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xu Ling-li
- Department of Acute and Critical Care, Shanghai Seventh People’s Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wang Lv
- Department of Emergency and Critical Care Medicine, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Jin Yong-mei
- Department of Nursing, Shanghai Seventh People’s Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
16
|
Zhang X, Zhou H, Chang X. Involvement of mitochondrial dynamics and mitophagy in diabetic endothelial dysfunction and cardiac microvascular injury. Arch Toxicol 2023; 97:3023-3035. [PMID: 37707623 DOI: 10.1007/s00204-023-03599-w] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 08/30/2023] [Indexed: 09/15/2023]
Abstract
Endothelial cells (ECs), found in the innermost layer of blood vessels, are crucial for maintaining the structure and function of coronary microcirculation. Dysregulated coronary microcirculation poses a fundamental challenge in diabetes-related myocardial microvascular injury, impacting myocardial blood perfusion, thrombogenesis, and inflammation. Extensive research aims to understand the mechanistic connection and functional relationship between cardiac EC dysfunction and the development, diagnosis, and treatment of diabetes-related myocardial microvascular injury. Despite the low mitochondrial content in ECs, mitochondria act as sensors of environmental and cellular stress, influencing EC viability, structure, and function. Mitochondrial dynamics and mitophagy play a vital role in orchestrating mitochondrial responses to various stressors by regulating morphology, localization, and degradation. Impaired mitochondrial dynamics or reduced mitophagy is associated with EC dysfunction, serving as a potential molecular basis and promising therapeutic target for diabetes-related myocardial microvascular injury. This review introduces newly recognized mechanisms of damaged coronary microvasculature in diabetes-related microvascular injury and provides updated insights into the molecular aspects of mitochondrial dynamics and mitophagy. Additionally, novel targeted therapeutic approaches against diabetes-related microvascular injury or endothelial dysfunction, focusing on mitochondrial fission and mitophagy in endothelial cells, are summarized.
Collapse
Affiliation(s)
- Xiao Zhang
- Dermatology, Liaocheng Hospital of Traditional Chinese Medicine, Liaocheng, 252000, China
| | - Hao Zhou
- Department of Cardiology, The Sixth Medical Center of People's Liberation Army General Hospital, Beijing, 100048, China.
| | - Xing Chang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, 5 Beixiagge, Xicheng District, Beijing, 100053, China.
| |
Collapse
|
17
|
Xu QF, Zhang H, Zhao Y, Liu D, Wei J, Jiang L, Liu YJ, Zhu XY. Increased R-spondin 3 contributes to aerobic exercise-induced protection against renal vascular endothelial hyperpermeability and acute kidney injury. Acta Physiol (Oxf) 2023; 239:e14036. [PMID: 37607126 DOI: 10.1111/apha.14036] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 07/11/2023] [Accepted: 08/08/2023] [Indexed: 08/24/2023]
Abstract
AIM Exercise training exerts protective effects against sepsis-associated multiple organ dysfunction. This study aimed to investigate whether aerobic exercise protected against sepsis-associated acute kidney injury (AKI) via modulating R-spondin 3 (RSPO3) expression. METHODS To investigate the effects of aerobic exercise on lipopolysaccharide (LPS)-induced AKI, LPS (20 mg/kg) was intraperitoneally injected after six weeks of treadmill training. To investigate the role of RSPO3 in LPS-induced AKI, wild-type (WT) or inducible endothelial cell-specific RSPO3 knockout (RSPO3EC-/- ) mice were intraperitoneally injected with 12 mg/kg LPS. RSPO3 was intraperitoneally injected 30 min before LPS treatment. RESULTS Aerobic exercise-trained mice were more resistant to LPS-induced body weight loss and hypothermia and had a significant higher survival rate than sedentary mice exposed to LPS. Exercise training restored the LPS-induced decreases in serum and renal RSPO3 levels. Exercise or RSPO3 attenuated, whereas inducible endothelial cell-specific RSPO3 knockout exacerbated LPS-induced renal glycocalyx loss, endothelial hyperpermeability, inflammation, and AKI. Bioinformatics analysis results revealed significant increases in the expression of matrix metalloproteinases (MMPs) in kidney tissues of mice exposed to sepsis or endotoxaemia, which was validated in renal tissue from LPS-exposed mice and LPS-treated human microvascular endothelial cells (HMVECs). Both RSPO3 and MMPs inhibitor restored LPS-induced downregulation of tight junction protein, adherens junction protein, and glycocalyx components, thus ameliorating LPS-induced endothelial leakage. Exercise or RSPO3 reversed LPS-induced upregulation of MMPs in renal tissues. CONCLUSION Increased renal expression of RSPO3 contributes to aerobic exercise-induced protection against LPS-induced renal endothelial hyperpermeability and AKI by suppressing MMPs-mediated disruption of glycocalyx and tight and adherens junctions.
Collapse
Affiliation(s)
- Qing-Feng Xu
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, The Key Laboratory of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai, China
- Department of Physiology, Navy Medical University, Shanghai, China
| | - Hui Zhang
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Ying Zhao
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, The Key Laboratory of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai, China
| | - Di Liu
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Juan Wei
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, The Key Laboratory of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai, China
| | - Lai Jiang
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yu-Jian Liu
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, The Key Laboratory of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai, China
| | - Xiao-Yan Zhu
- Department of Physiology, Navy Medical University, Shanghai, China
| |
Collapse
|
18
|
Zhan JH, Wei J, Liu L, Xu YT, Ji H, Wang CN, Liu YJ, Zhu XY. Investigation of a UPR-Related Gene Signature Identifies the Pro-Fibrotic Effects of Thrombospondin-1 by Activating CD47/ROS/Endoplasmic Reticulum Stress Pathway in Lung Fibroblasts. Antioxidants (Basel) 2023; 12:2024. [PMID: 38136144 PMCID: PMC10740656 DOI: 10.3390/antiox12122024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 11/17/2023] [Accepted: 11/19/2023] [Indexed: 12/24/2023] Open
Abstract
Unfolded protein response (UPR) signaling and endoplasmic reticulum (ER) stress have been linked to pulmonary fibrosis. However, the relationship between UPR status and pulmonary function and prognosis in idiopathic pulmonary fibrosis (IPF) patients remains largely unknown. Through a series of bioinformatics analyses, we established a correlation between UPR status and pulmonary function in IPF patients. Furthermore, thrombospondin-1 (TSP-1) was identified as a potential biomarker for prognostic evaluation in IPF patients. By utilizing both bulk RNA profiling and single-cell RNA sequencing data, we demonstrated the upregulation of TSP-1 in lung fibroblasts during pulmonary fibrosis. Gene set enrichment analysis (GSEA) results indicated a positive association between TSP-1 expression and gene sets related to the reactive oxygen species (ROS) pathway in lung fibroblasts. TSP-1 overexpression alone induced mild ER stress and pulmonary fibrosis, and it even exacerbated bleomycin-induced ER stress and pulmonary fibrosis. Mechanistically, TSP-1 promoted ER stress and fibroblast activation through CD47-dependent ROS production. Treatment with either TSP-1 inhibitor or CD47 inhibitor significantly attenuated BLM-induced ER stress and pulmonary fibrosis. Collectively, these findings suggest that the elevation of TSP-1 during pulmonary fibrosis is not merely a biomarker but likely plays a pathogenic role in the fibrotic changes in the lung.
Collapse
Affiliation(s)
- Jun-Hui Zhan
- School of Kinesiology, The Key Laboratory of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai 200438, China; (J.-H.Z.); (J.W.); (Y.-T.X.); (H.J.)
| | - Juan Wei
- School of Kinesiology, The Key Laboratory of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai 200438, China; (J.-H.Z.); (J.W.); (Y.-T.X.); (H.J.)
- School of Sports and Health, Nanjing Sport Institute, Nanjing 210014, China;
| | - Lin Liu
- School of Sports and Health, Nanjing Sport Institute, Nanjing 210014, China;
| | - Yi-Tong Xu
- School of Kinesiology, The Key Laboratory of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai 200438, China; (J.-H.Z.); (J.W.); (Y.-T.X.); (H.J.)
| | - Hui Ji
- School of Kinesiology, The Key Laboratory of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai 200438, China; (J.-H.Z.); (J.W.); (Y.-T.X.); (H.J.)
| | - Chang-Nan Wang
- Department of Physiology, Navy Medical University, Shanghai 200433, China;
| | - Yu-Jian Liu
- School of Kinesiology, The Key Laboratory of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai 200438, China; (J.-H.Z.); (J.W.); (Y.-T.X.); (H.J.)
| | - Xiao-Yan Zhu
- Department of Physiology, Navy Medical University, Shanghai 200433, China;
| |
Collapse
|
19
|
Xue C, Chen K, Gao Z, Bao T, Dong L, Zhao L, Tong X, Li X. Common mechanisms underlying diabetic vascular complications: focus on the interaction of metabolic disorders, immuno-inflammation, and endothelial dysfunction. Cell Commun Signal 2023; 21:298. [PMID: 37904236 PMCID: PMC10614351 DOI: 10.1186/s12964-022-01016-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 12/11/2022] [Indexed: 11/01/2023] Open
Abstract
Diabetic vascular complications (DVCs), including macro- and micro- angiopathy, account for a high percentage of mortality in patients with diabetes mellitus (DM). Endothelial dysfunction is the initial and role step for the pathogenesis of DVCs. Hyperglycemia and lipid metabolism disorders contribute to endothelial dysfunction via direct injury of metabolism products, crosstalk between immunity and inflammation, as well as related interaction network. Although physiological and phenotypic differences support their specified changes in different targeted organs, there are still several common mechanisms underlying DVCs. Also, inhibitors of these common mechanisms may decrease the incidence of DVCs effectively. Thus, this review may provide new insights into the possible measures for the secondary prevention of DM. And we discussed the current limitations of those present preventive measures in DVCs research. Video Abstract.
Collapse
Affiliation(s)
- Chongxiang Xue
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, No.5 BeiXianGe Street, Xicheng District, Beijing, 100053, China
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Keyu Chen
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, No.5 BeiXianGe Street, Xicheng District, Beijing, 100053, China
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Zezheng Gao
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, No.5 BeiXianGe Street, Xicheng District, Beijing, 100053, China
- Department of Endocrinology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Tingting Bao
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, No.5 BeiXianGe Street, Xicheng District, Beijing, 100053, China
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | - LiShuo Dong
- Changchun University of Traditional Chinese Medicine, Changchun, 130117, China
| | - Linhua Zhao
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, No.5 BeiXianGe Street, Xicheng District, Beijing, 100053, China.
| | - Xiaolin Tong
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, No.5 BeiXianGe Street, Xicheng District, Beijing, 100053, China.
| | - Xiuyang Li
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, No.5 BeiXianGe Street, Xicheng District, Beijing, 100053, China.
- Department of Endocrinology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China.
| |
Collapse
|
20
|
Song J, Du J, Tan X, Li Y, Yu Q, Liu W, Zhu X, Cong B. Tissue kallikrein-related peptidase8 accentuates cardiac fibrosis after myocardial ischemia-reperfusion injury via regulation of cardiac fibroblasts. Life Sci 2023; 329:121973. [PMID: 37482211 DOI: 10.1016/j.lfs.2023.121973] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 07/12/2023] [Accepted: 07/20/2023] [Indexed: 07/25/2023]
Abstract
AIMS Tissue kallikrein-related peptidase8 (KLK8) has been found to mitigate acute myocardial ischemia-reperfusion (IR) injury. However, the effect of KLK8 on cardiac remodeling in response to IR injury has not been determined. MATERIALS AND METHODS KLK8 overexpressing transgenic rat (KLK8-TG) was used as the animal model. IR injury was induced by ligating the left anterior descending coronary artery for 1 h and subsequent reperfusion. The functional and morphological changes of the heart were examined 14 days after the injury. Neonatal rat cardiac fibroblasts (CFs) were used to investigate the molecular mechanisms in vitro. KEY FINDINGS KLK8 overexpression enhanced cardiac diastolic dysfunction, fibrosis, and hypertrophy after IR injury, indicating that KLK8 accentuated cardiac remodeling in response to IR injury. Moreover, KLK8 overexpression increased epidermal growth factor (EGF) release and promoted the phosphorylation of EGF receptor (EGFR) and ERK1/2 in the heart after IR injury. It was interesting to find that both EGFR antagonist (AG 1478) and MEK inhibitor (PD98059) attenuated the KLK8-induced proliferation and activation of CFs in vitro, indicating that EGFR signaling might mediate the pro-fibrotic action of KLK8. SIGNIFICANCE KLK8 plays a crucial role in cardiac remodeling after myocardial infarction. KLK8 accentuates cardiac fibrosis after IR injury, possibly mediated by EGFR signaling in CFs.
Collapse
Affiliation(s)
- Jinchao Song
- Department of Anesthesiology, Shidong Hospital Affiliated to University of Shanghai for Science and Technology, Shanghai, China; Department of Physiology, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Jiankui Du
- Department of Physiology, Naval Medical University (Second Military Medical University), Shanghai, China; Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xing Tan
- Department of Physiology, Naval Medical University (Second Military Medical University), Shanghai, China; Department of Marine Biomedicine and Polar Medicine, Naval Medical Center, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Yang Li
- Department of Anesthesiology, Shidong Hospital Affiliated to University of Shanghai for Science and Technology, Shanghai, China
| | - Qing Yu
- Department of Physiology, Naval Medical University (Second Military Medical University), Shanghai, China; Department of Cardiology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Wen Liu
- Department of Anesthesiology, Shidong Hospital Affiliated to University of Shanghai for Science and Technology, Shanghai, China
| | - Xiaoyan Zhu
- Department of Physiology, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Binhai Cong
- Department of Physiology, Naval Medical University (Second Military Medical University), Shanghai, China.
| |
Collapse
|
21
|
Li N, Zhang T, Wang R, Sun Y, Chu L, Lu X, Sun K. Homotypic targeted nanoplatform enable efficient chemoimmunotherapy and reduced DOX cardiotoxicity in chemoresistant cancer via TGF-β1 blockade. J Control Release 2023; 361:147-160. [PMID: 37536544 DOI: 10.1016/j.jconrel.2023.07.063] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 07/18/2023] [Accepted: 07/31/2023] [Indexed: 08/05/2023]
Abstract
Doxorubicin (DOX) with broad-spectrum antitumor activity has been reported to induce effective immunogenic cell death (ICD) effect. However, the serious cardiotoxicity and chemoresistance severely restrict the widely clinical application of DOX. Herein, for the first time, a bio-inspired nanoplatform via co-assembly of DOX-conjugated polyethyleneimine (PEI-DOX), cancer cell membrane (CCM) and TGF-β1 siRNA (siTGF-β1) was rationally designed, which can not only overcome the drawbacks of DOX but also display high capability to modulate the tumor microenvironment and prevent the tumor progressing and metastasis. Experimental studies confirmed the pH-sensitivity of PEI-DOX and the homotypic-targeting and immuno-escapable ability of CCM, resulting an enhanced accumulation of DOX and siTGF-β1 in tumor sites. In addition to this, the bio-inspired nanoplatform could also improve the stability and facilitate the endosomal escape of siTGF-β1. All these effects ensured the silence efficiency of siTGF-β1 in tumor sites, which could further modulate the chemoresistant and immunosuppressive tumor microenvironment, resulting a synergistic effect with DOX to prevent tumor progressing and metastasis. Additionally, even trapped in cardiac tissues, siTGF-β1 could inhibit the production of TGF-β1 and ROS induced by DOX, resulting a reduced myocardial damage. Therefore, our newly designed bio-inspired nano-delivery system may be a promising nanoplatform with efficient chemoimmunotherapy to ameliorate DOX-induced cardiotoxicity and combat tumor growth and metastasis in chemoresistant cancer.
Collapse
Affiliation(s)
- Nuannuan Li
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Ministry of Education, Yantai University, Yantai, China
| | - Tianyu Zhang
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Ministry of Education, Yantai University, Yantai, China
| | - Ru Wang
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Ministry of Education, Yantai University, Yantai, China
| | - Yiying Sun
- Yantai Saipute Analyzing Service Co. Ltd, Yantai, Shandong Province, China
| | - Liuxiang Chu
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Ministry of Education, Yantai University, Yantai, China
| | - Xiaoyan Lu
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Ministry of Education, Yantai University, Yantai, China
| | - Kaoxiang Sun
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Ministry of Education, Yantai University, Yantai, China.
| |
Collapse
|
22
|
Wei J, Zhan J, Ji H, Xu Y, Xu Q, Zhu X, Liu Y. Fibroblast Upregulation of Vitamin D Receptor Represents a Self-Protective Response to Limit Fibroblast Proliferation and Activation during Pulmonary Fibrosis. Antioxidants (Basel) 2023; 12:1634. [PMID: 37627629 PMCID: PMC10451996 DOI: 10.3390/antiox12081634] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 08/10/2023] [Accepted: 08/15/2023] [Indexed: 08/27/2023] Open
Abstract
Dysregulation of vitamin D receptor (VDR) is implicated in chronic obstructive pulmonary disease. However, whether VDR dysregulation contributes to the development of pulmonary fibrosis remains largely unknown. Analysis of bulk and single-cell RNA profiling datasets revealed VDR upregulation in lung fibroblasts from patients with pulmonary fibrosis or fibrotic mice, which was validated in lung fibroblasts from bleomycin-exposed mice and bleomycin-treated fibroblasts. Stable VDR knockdown promoted, whereas the VDR agonist paricalcitol suppressed lung fibroblast proliferation and activation. Gene set enrichment analysis (GSEA) showed that the JAK/STAT pathway and unfolded protein response (UPR), a process related to endoplasmic reticulum (ER) stress, were enriched in lung fibroblasts of fibrotic lungs. Stable VDR knockdown stimulated, but paricalcitol suppressed ER stress and JAK1/STAT3 activation in lung fibroblasts. The STAT3 inhibitor blocked bleomycin- or stable VDR knockdown-induced ER stress. Paricalcitol inhibited the bleomycin-induced enrichment of STAT3 to the ATF6 promoter, thereby suppressing ATF6 expression in fibroblasts. Paricalcitol or intrapulmonary VDR overexpression inactivated JAK1/STAT3 and suppressed ER stress in bleomycin-treated mice, thus resulting in the inhibition of fibroblast proliferation and activation. Collectively, this study suggests that fibroblast VDR upregulation may be a self-protective response to limit fibroblast proliferation and activation during pulmonary fibrosis by suppressing the JAK1/STAT3/ER stress pathway.
Collapse
Affiliation(s)
- Juan Wei
- School of Kinesiology, The Key Laboratory of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai 200438, China; (J.W.); (J.Z.); (H.J.); (Y.X.); (Q.X.)
- School of Sports and Health, Nanjing Sport Institute, Nanjing 210014, China
| | - Junhui Zhan
- School of Kinesiology, The Key Laboratory of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai 200438, China; (J.W.); (J.Z.); (H.J.); (Y.X.); (Q.X.)
| | - Hui Ji
- School of Kinesiology, The Key Laboratory of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai 200438, China; (J.W.); (J.Z.); (H.J.); (Y.X.); (Q.X.)
| | - Yitong Xu
- School of Kinesiology, The Key Laboratory of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai 200438, China; (J.W.); (J.Z.); (H.J.); (Y.X.); (Q.X.)
| | - Qingfeng Xu
- School of Kinesiology, The Key Laboratory of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai 200438, China; (J.W.); (J.Z.); (H.J.); (Y.X.); (Q.X.)
| | - Xiaoyan Zhu
- Department of Physiology, Navy Medical University, Shanghai 200433, China
| | - Yujian Liu
- School of Kinesiology, The Key Laboratory of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai 200438, China; (J.W.); (J.Z.); (H.J.); (Y.X.); (Q.X.)
| |
Collapse
|
23
|
Qiu H, Ni C, Jia C, Rong X, Chu M, Wu R, Han B. CircRNA7632 down-regulation alleviates endothelial cell dysfunction in Kawasaki disease via regulating IL-33 expression. Cell Stress Chaperones 2023; 28:363-374. [PMID: 37166618 PMCID: PMC10352195 DOI: 10.1007/s12192-023-01333-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 02/10/2023] [Accepted: 02/21/2023] [Indexed: 05/12/2023] Open
Abstract
Kawasaki disease (KD) is a form of idiopathic vasculitis frequently accompanied by coronary artery lesions, which involves endothelial dysfunction. Recent studies have demonstrated that circular RNAs (circRNAs) are implicated in many cardiovascular diseases. However, few studies have examined the role of circRNAs on endothelial dysfunction in KD. In this study, we investigated the role of circ7632 on endothelial-mesenchymal transition (EndoMT) in KD and then explored the underlying mechanism. Children diagnosed with KD and age-matched healthy controls (HC) were included. Sera samples were collected. Primary human umbilical vein endothelial cells (HUVECs) were obtained and incubated with 15% HC and KD serum for 48 h. The mRNA and protein expression of mesenchymal markers vimentin and α-smooth muscle actin (α-SMA) and endothelial marker zonula occludens-1 (ZO-1) in HUVECs transfected with plasmid-circ7632 and si-circ7632 were detected by RT-qPCR and Western blot analysis. CCK8, scratch test, and migration test were performed to examine the effect of circ7632 on the cell proliferation and migration. The circ7632 level was higher in HUVECs treated by KD serum than in HUVECs treated with HC serum. Overexpression of circ7632 significantly increased vimentin and α-SMA expression, decreased ZO-1 expression, and also decreased cell proliferation. Down-regulation of circ7632 expression got the opposite results. RNA-seq analysis, and confirmatory experiment displayed that down-regulation of circ7632 decreased IL-33 expression, and IL-33 silencing mitigated KD serum-mediated EndoMT. Our study revealed that circ7632 level was elevated in KD serum-treated HUVECs. Circ7632 down-regulation could alleviate EndoMT likely through decreasing IL-33 expression. The circ7632 may become a potential therapeutic target for KD.
Collapse
Affiliation(s)
- Huixian Qiu
- Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Chao Ni
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Chang Jia
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Xing Rong
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Maoping Chu
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Rongzhou Wu
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Bo Han
- Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China.
- Department of Pediatric Cardiology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250021, Shandong, China.
| |
Collapse
|
24
|
Xu DH, Du JK, Liu SY, Zhang H, Yang L, Zhu XY, Liu YJ. Upregulation of KLK8 contributes to CUMS-induced hippocampal neuronal apoptosis by cleaving NCAM1. Cell Death Dis 2023; 14:278. [PMID: 37076499 PMCID: PMC10115824 DOI: 10.1038/s41419-023-05800-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 04/02/2023] [Accepted: 04/06/2023] [Indexed: 04/21/2023]
Abstract
Neuronal apoptosis has been well-recognized as a critical mediator in the pathogenesis of depressive disorders. Tissue kallikrein-related peptidase 8 (KLK8), a trypsin-like serine protease, has been implicated in the pathogenesis of several psychiatric disorders. The present study aimed to explore the potential function of KLK8 in hippocampal neuronal cell apoptosis associated with depressive disorders in rodent models of chronic unpredictable mild stress (CUMS)-induced depression. It was found that depression-like behavior in CUMS-induced mice was associated with hippocampal KLK8 upregulation. Transgenic overexpression of KLK8 exacerbated, whereas KLK8 deficiency attenuated CUMS-induced depression-like behaviors and hippocampal neuronal apoptosis. In HT22 murine hippocampal neuronal cells and primary hippocampal neurons, adenovirus-mediated overexpression of KLK8 (Ad-KLK8) was sufficient to induce neuron apoptosis. Mechanistically, it was identified that the neural cell adhesion molecule 1 (NCAM1) may associate with KLK8 in hippocampal neurons as KLK8 proteolytically cleaved the NCAM1 extracellular domain. Immunofluorescent staining exhibited decreased NCAM1 in hippocampal sections obtained from mice or rats exposed to CUMS. Transgenic overexpression of KLK8 exacerbated, whereas KLK8 deficiency largely prevented CUMS-induced loss of NCAM1 in the hippocampus. Both adenovirus-mediated overexpression of NCAM1 and NCAM1 mimetic peptide rescued KLK8-overexpressed neuron cells from apoptosis. Collectively, this study identified a new pro-apoptotic mechanism in the hippocampus during the pathogenesis of CUMS-induced depression via the upregulation of KLK8, and raised the possibility of KLK8 as a potential therapeutic target for depression.
Collapse
Affiliation(s)
- Dan-Hong Xu
- School of Kinesiology, Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, The Key Laboratory of Exercise and Health Sciences of Ministry of Education Shanghai University of Sport, Shanghai, 200438, China
- Department of Physiology, Navy Medical University, Shanghai, 200433, China
| | - Jian-Kui Du
- National Clinical Research Center for Geriatric Disorders and National International Joint Research Center for Medical Metabolomics, Xiangya Hospital, Central South University, Changsha, Hunan, 41008, China
| | - Shi-Yu Liu
- School of Kinesiology, Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, The Key Laboratory of Exercise and Health Sciences of Ministry of Education Shanghai University of Sport, Shanghai, 200438, China
| | - Hui Zhang
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200092, China
| | - Lu Yang
- School of Kinesiology, Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, The Key Laboratory of Exercise and Health Sciences of Ministry of Education Shanghai University of Sport, Shanghai, 200438, China
| | - Xiao-Yan Zhu
- Department of Physiology, Navy Medical University, Shanghai, 200433, China.
| | - Yu-Jian Liu
- School of Kinesiology, Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, The Key Laboratory of Exercise and Health Sciences of Ministry of Education Shanghai University of Sport, Shanghai, 200438, China.
| |
Collapse
|
25
|
Li Y, Zhao Y, Song L, Xiong L, Li W, Wu W, Miao L. High glucose levels contribute to vascular fibrosis via the activation of the endothelial-to-mesenchymal transition in periodontitis. J Periodontal Res 2023; 58:225-236. [PMID: 36537563 DOI: 10.1111/jre.13084] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 11/02/2022] [Accepted: 12/06/2022] [Indexed: 12/24/2022]
Abstract
OBJECTIVE To determine the changes of Porphyromonas gingivalis (P. gingivalis) growth and metabolism and identify whether the vascular epithelium change could be induced in diabetic periodontitis. BACKGROUND Maintaining favourable vascular function is a precondition for periodontal regeneration. In diabetic periodontitis, high glucose levels could enhance the metabolism of pathogens, and a complex condition involving inflammation and high glucose levels would disrupt homeostasis of the epithelium and promote fibrosis by endothelial-to-mesenchymal transition (EndMT). METHODS Porphyromonas gingivalis was cultured with glucose to judge its metabolic activity. Human umbilical vein endothelial cells (HUVECs) were treated with P. gingivalis-lipopolysaccharide (LPS) (10 μg/ml) and/or high glucose concentrations (25 mM), and transforming growth factor (TGF)-β inhibitor was used to block EndMT. Inflammation level was assessed by flow cytometry. Multiple biological functions including EndMT, angiopoiesis, and cell migration were analysed. Additionally, gene expressions and protein levels were determined with qPCR and western blot, respectively. Finally, blood vessels were cultured ex vivo, and EndMT and fibrosis markers were detected by immunohistochemistry. RESULTS Glucose could promote P. gingivalis growth and biofilm formation as well as the expression of virulence factor genes including FimA, RgpA, RgpB, and Kgp. P. gingivalis-LPS and glucose could increase intracellular reactive oxygen species (ROS) and promote fibrosis via EndMT in HUVECs, along with attenuating angiopoiesis and cell migration, which could be resumed by blocking EndMT with TGF-β inhibitor. Vascular fibrosis was observed after the addition of glucose via EndMT regulation. CONCLUSION Glucose augmented the growth and metabolism of P. gingivalis and promoted fibrosis by the activation of EndMT, as well as the inhibition of angiopoiesis and cell migration.
Collapse
Affiliation(s)
- Yuyang Li
- Department of Cariology and Endodontics, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Yue Zhao
- Department of Cariology and Endodontics, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Lutong Song
- Department of Cariology and Endodontics, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Liping Xiong
- Department of Orthodontics, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Wen Li
- Department of Cariology and Endodontics, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Wenlei Wu
- Department of Periodontology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Leiying Miao
- Department of Cariology and Endodontics, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| |
Collapse
|
26
|
Cheng Y, Wang Y, Yin R, Xu Y, Zhang L, Zhang Y, Yang L, Zhao D. Central role of cardiac fibroblasts in myocardial fibrosis of diabetic cardiomyopathy. Front Endocrinol (Lausanne) 2023; 14:1162754. [PMID: 37065745 PMCID: PMC10102655 DOI: 10.3389/fendo.2023.1162754] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 03/20/2023] [Indexed: 04/03/2023] Open
Abstract
Diabetic cardiomyopathy (DCM), a main cardiovascular complication of diabetes, can eventually develop into heart failure and affect the prognosis of patients. Myocardial fibrosis is the main factor causing ventricular wall stiffness and heart failure in DCM. Early control of myocardial fibrosis in DCM is of great significance to prevent or postpone the progression of DCM to heart failure. A growing body of evidence suggests that cardiomyocytes, immunocytes, and endothelial cells involve fibrogenic actions, however, cardiac fibroblasts, the main participants in collagen production, are situated in the most central position in cardiac fibrosis. In this review, we systematically elaborate the source and physiological role of myocardial fibroblasts in the context of DCM, and we also discuss the potential action and mechanism of cardiac fibroblasts in promoting fibrosis, so as to provide guidance for formulating strategies for prevention and treatment of cardiac fibrosis in DCM.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Dong Zhao
- *Correspondence: Longyan Yang, ; Dong Zhao,
| |
Collapse
|
27
|
Liu M, Peng T, Hu L, Wang M, Guo D, Qi B, Ren G, Wang D, Li Y, Song L, Hu J, Li Y. N-glycosylation-mediated CD147 accumulation induces cardiac fibrosis in the diabetic heart through ALK5 activation. Int J Biol Sci 2023; 19:137-155. [PMID: 36594096 PMCID: PMC9760447 DOI: 10.7150/ijbs.77469] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 10/12/2022] [Indexed: 11/24/2022] Open
Abstract
Emerging evidence has implicated the important role of fibrosis in diabetic cardiomyopathy (DCM), while the underlying mechanism remains unclear. Considering the distinct and overlapping roles of Cluster of Differentiation 147 (CD147) in the pathogenesis of fibrotic diseases, we aim to investigate the role of CD147 in the fibrosis of DCM and explore its underlying mechanism. AAV9-mediated cardiac-specific CD147 silencing attenuated cardiac fibrosis and cardiac function in diabetic mice. CD147 knockdown significantly inhibited high glucose (HG)-induced activation of CFs. Mechanistically, CD147 directly bound to type I transcription growth factor β (TGF-β) receptor I (ALK5), promoting ALK5 activation and endocytosis to induce SMAD2/3 phosphorylation and nuclear translocation. In addition, HG prevented the ubiquitin-proteasome-dependent degradation of CD147 by promoting GNT-V-mediated N-glycosylation. As a result, cardiac-specific CD147 overexpression in control mice mimicked diabetes-induced cardiac fibrosis, aggravating cardiac function. Importantly, CD147 was also upregulated in serum and myocardial specimens from patients with diabetes compared with non-diabetes, accompanied by echocardiographic indices of cardiac dysfunction and excessive collagen deposition. Our study provides the first evidence that CD147 acts as a pivotal factor to promote diabetic cardiac fibrosis, and may contribute to the development of future CD147-based therapeutic strategies for DCM.
Collapse
Affiliation(s)
- Mingchuan Liu
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China
| | - Tingwei Peng
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China
| | - Lang Hu
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China
| | - Min Wang
- Department of Pulmonary and Critical Care Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710038, China
| | - Dong Guo
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China
| | - Bingchao Qi
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China
| | - Gaotong Ren
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China
| | - Di Wang
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China
| | - Yunqing Li
- Brigade 2, College of Basic Medicine, The Fourth Military Medical University, Xi'an, 710038, China
| | - Liqiang Song
- Department of Pulmonary and Critical Care Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710038, China
| | - Jianqiang Hu
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China.,✉ Corresponding authors: Yan Li. Address: Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China. E-mail: . Jianqiang Hu. Address: Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China. E-mail:
| | - Yan Li
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China.,✉ Corresponding authors: Yan Li. Address: Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China. E-mail: . Jianqiang Hu. Address: Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China. E-mail:
| |
Collapse
|
28
|
Xiao H, Ma Y, Zhou Z, Li X, Ding K, Wu Y, Wu T, Chen D. Disease patterns of coronary heart disease and type 2 diabetes harbored distinct and shared genetic architecture. Cardiovasc Diabetol 2022; 21:276. [PMID: 36494812 PMCID: PMC9738029 DOI: 10.1186/s12933-022-01715-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 12/02/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Coronary heart disease (CHD) and type 2 diabetes (T2D) are two complex diseases with complex interrelationships. However, the genetic architecture of the two diseases is often studied independently by the individual single-nucleotide polymorphism (SNP) approach. Here, we presented a genotypic-phenotypic framework for deciphering the genetic architecture underlying the disease patterns of CHD and T2D. METHOD A data-driven SNP-set approach was performed in a genome-wide association study consisting of subpopulations with different disease patterns of CHD and T2D (comorbidity, CHD without T2D, T2D without CHD and all none). We applied nonsmooth nonnegative matrix factorization (nsNMF) clustering to generate SNP sets interacting the information of SNP and subject. Relationships between SNP sets and phenotype sets harboring different disease patterns were then assessed, and we further co-clustered the SNP sets into a genetic network to topologically elucidate the genetic architecture composed of SNP sets. RESULTS We identified 23 non-identical SNP sets with significant association with CHD or T2D (SNP-set based association test, P < 3.70 × [Formula: see text]). Among them, disease patterns involving CHD and T2D were related to distinct SNP sets (Hypergeometric test, P < 2.17 × [Formula: see text]). Accordingly, numerous genes (e.g., KLKs, GRM8, SHANK2) and pathways (e.g., fatty acid metabolism) were diversely implicated in different subtypes and related pathophysiological processes. Finally, we showed that the genetic architecture for disease patterns of CHD and T2D was composed of disjoint genetic networks (heterogeneity), with common genes contributing to it (pleiotropy). CONCLUSION The SNP-set approach deciphered the complexity of both genotype and phenotype as well as their complex relationships. Different disease patterns of CHD and T2D share distinct genetic architectures, for which lipid metabolism related to fibrosis may be an atherogenic pathway that is specifically activated by diabetes. Our findings provide new insights for exploring new biological pathways.
Collapse
Affiliation(s)
- Han Xiao
- grid.11135.370000 0001 2256 9319Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, 100191 China
| | - Yujia Ma
- grid.11135.370000 0001 2256 9319Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, 100191 China
| | - Zechen Zhou
- grid.11135.370000 0001 2256 9319Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, 100191 China
| | - Xiaoyi Li
- grid.11135.370000 0001 2256 9319Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, 100191 China
| | - Kexin Ding
- grid.11135.370000 0001 2256 9319Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, 100191 China
| | - Yiqun Wu
- grid.11135.370000 0001 2256 9319Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, 100191 China
| | - Tao Wu
- grid.11135.370000 0001 2256 9319Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, 100191 China
| | - Dafang Chen
- grid.11135.370000 0001 2256 9319Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, 100191 China
| |
Collapse
|
29
|
Gong F, Qu R, Li Y, Lv Y, Dai J. Astragalus Mongholicus: A review of its anti-fibrosis properties. Front Pharmacol 2022; 13:976561. [PMID: 36160396 PMCID: PMC9490009 DOI: 10.3389/fphar.2022.976561] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 08/18/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Fibrosis-related diseases (FRD) include cerebral fibrosis, pulmonary fibrosis, cardiac fibrosis, liver fibrosis, renal fibrosis, peritoneal fibrosis, etc. The effects of fibrosis can be severe, resulting in organ dysfunction, functional decline, and even organ failure, which can cause serious health problems.Aim: Currently, there is no effective modern medicine for anti-fibrosis in the clinics; however, Chinese medicine has a certain beneficial effect on treating such diseases. Astragalus Mongholicus (AM) has rich medicinal value, and its anti-fibrosis effect has been recently investigated. In recent years, more and more experimental studies have been conducted on the intervention of astragaloside IV (AS-IV), astragalus polysaccharide (APS), astragalus flavone, cycloastragalus alcohol, astragalus water extract and other pharmacological components in fibrosis-related diseases, attracting the interest of researchers. We aim to provide ideas for future research by summarizing recent research advances of AM in treating fibrosis-related diseases.Methods: A literature search was conducted from the core collections of electronic databases such as Baidu Literature, Sciencen.com, Google Scholar, PubMed, and Science Direct using the above keywords and the pharmacological and phytochemical details of the plant.Results: AM can be used to intervene in fibrosis-disease progression by regulating inflammation, oxidative stress, the immune system, and metabolism.Conclusion: AS-IV, APS, and astragalus flavone were studied and discussed in detail. These components have high potential anti-fibrosis activity. Overall, this review aims to gain insight into the AM’s role in treating fibro-related diseases.
Collapse
Affiliation(s)
- Fengying Gong
- Department of Traditional Chinese Medicine, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Rongmei Qu
- Guangdong Provincial Key Laboratory of Medical Biomechanics and Guangdong Engineering Research Center for Translation of Medical 3D Printing Application and National Key Discipline of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Yongchun Li
- Department of Traditional Chinese Medicine, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Ying Lv
- Department of Traditional Chinese Medicine, Nanfang Hospital of Southern Medical University, Guangzhou, China
- *Correspondence: Ying Lv, ; Jingxing Dai,
| | - Jingxing Dai
- Guangdong Provincial Key Laboratory of Medical Biomechanics and Guangdong Engineering Research Center for Translation of Medical 3D Printing Application and National Key Discipline of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- *Correspondence: Ying Lv, ; Jingxing Dai,
| |
Collapse
|
30
|
Sasaki R, Kurebayashi N, Eguchi H, Horimoto Y, Shiga T, Miyazaki S, Kashiyama T, Akamatsu W, Saito M. Involvement of kallikrein-PAR2-proinflammatory pathway in severe trastuzumab-induced cardiotoxicity. Cancer Sci 2022; 113:3449-3462. [PMID: 35879248 PMCID: PMC9530879 DOI: 10.1111/cas.15508] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 07/11/2022] [Accepted: 07/14/2022] [Indexed: 11/28/2022] Open
Abstract
Trastuzumab‐induced cardiotoxicity interferes with continued treatment in approximately 10% of patients with ErbB2‐positive breast cancer, but its mechanism has not been fully elucidated. In this study, we recruited trastuzumab‐treated patients with ≥30% reduction in left ventricular ejection fraction (SP) and noncardiotoxic patients (NP). From each of these patients, we established three cases of induced pluripotent stem cell‐derived cardiomyocytes (pt‐iPSC‐CMs). Reduced contraction and relaxation velocities following trastuzumab treatment were more evident in SP pt‐iPSC‐CMs than NP pt‐iPSC‐CMs, indicating the cardiotoxicity phenotype could be replicated. Differences in ATP production, reactive oxygen species, and autophagy activity were observed between the two groups. Analysis of transcripts revealed enhanced kallikrein5 expression and pro‐inflammatory signaling pathways, such as interleukin‐1β, in SP pt‐iPSC‐CMs after trastuzumab treatment. The kallilkrein5‐protease‐activated receptor 2 (PAR2)‐MAPK signaling pathway was more activated in SP pt‐iPSC‐CMs, and treatment with a PAR2‐antagonist suppressed interleukin‐1β expression. Our data indicate enhanced pro‐inflammatory responses through kallikrein5‐PAR2 signaling and vulnerability to external stresses appear to be the cause of trastuzumab‐induced cardiotoxicity in SP.
Collapse
Affiliation(s)
- Ritsuko Sasaki
- Department of Breast Oncology, Juntendo University Graduate School of Medicine
| | - Nagomi Kurebayashi
- Department of Cellular and Molecular Pharmacology, Juntendo University Graduate School of Medicine
| | - Hidetaka Eguchi
- Intractable Disease Research Center, Juntendo University Graduate School of Medicine
| | - Yoshiya Horimoto
- Department of Breast Oncology, Juntendo University Graduate School of Medicine
| | - Takahiro Shiga
- Center for Genomic and Regenerative Medicine, Juntendo University Graduate School of Medicine
| | - Sakiko Miyazaki
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine
| | - Taku Kashiyama
- Department of Cellular and Molecular Pharmacology, Juntendo University Graduate School of Medicine
| | - Wado Akamatsu
- Center for Genomic and Regenerative Medicine, Juntendo University Graduate School of Medicine
| | - Mitsue Saito
- Department of Breast Oncology, Juntendo University Graduate School of Medicine
| |
Collapse
|
31
|
Phang RJ, Ritchie RH, Hausenloy DJ, Lees JG, Lim SY. Cellular interplay between cardiomyocytes and non-myocytes in diabetic cardiomyopathy. Cardiovasc Res 2022; 119:668-690. [PMID: 35388880 PMCID: PMC10153440 DOI: 10.1093/cvr/cvac049] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 02/16/2022] [Accepted: 03/05/2022] [Indexed: 11/13/2022] Open
Abstract
Patients with Type 2 diabetes mellitus (T2DM) frequently exhibit a distinctive cardiac phenotype known as diabetic cardiomyopathy. Cardiac complications associated with T2DM include cardiac inflammation, hypertrophy, fibrosis and diastolic dysfunction in the early stages of the disease, which can progress to systolic dysfunction and heart failure. Effective therapeutic options for diabetic cardiomyopathy are limited and often have conflicting results. The lack of effective treatments for diabetic cardiomyopathy is due in part, to our poor understanding of the disease development and progression, as well as a lack of robust and valid preclinical human models that can accurately recapitulate the pathophysiology of the human heart. In addition to cardiomyocytes, the heart contains a heterogeneous population of non-myocytes including fibroblasts, vascular cells, autonomic neurons and immune cells. These cardiac non-myocytes play important roles in cardiac homeostasis and disease, yet the effect of hyperglycaemia and hyperlipidaemia on these cell types are often overlooked in preclinical models of diabetic cardiomyopathy. The advent of human induced pluripotent stem cells provides a new paradigm in which to model diabetic cardiomyopathy as they can be differentiated into all cell types in the human heart. This review will discuss the roles of cardiac non-myocytes and their dynamic intercellular interactions in the pathogenesis of diabetic cardiomyopathy. We will also discuss the use of sodium-glucose cotransporter 2 inhibitors as a therapy for diabetic cardiomyopathy and their known impacts on non-myocytes. These developments will no doubt facilitate the discovery of novel treatment targets for preventing the onset and progression of diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Ren Jie Phang
- O'Brien Institute Department, St Vincent's Institute of Medical Research, Fitzroy, Victoria 3065, Australia.,Departments of Surgery and Medicine, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Rebecca H Ritchie
- School of Biosciences, Parkville, Victoria 3010, Australia.,Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Parkville, Victoria 3052, Australia.,Department of Pharmacology, Monash University, Clayton, Victoria 3800, Australia
| | - Derek J Hausenloy
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, Singapore.,Cardiovascular and Metabolic Disorders Programme, Duke-NUS Medical School, Singapore, Singapore.,Yong Loo Lin School of Medicine, National University Singapore, Singapore, Singapore.,The Hatter Cardiovascular Institute, University College London, London, UK.,Cardiovascular Research Center, College of Medical and Health Sciences, Asia University, Taichung City, Taiwan
| | - Jarmon G Lees
- O'Brien Institute Department, St Vincent's Institute of Medical Research, Fitzroy, Victoria 3065, Australia.,Departments of Surgery and Medicine, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Shiang Y Lim
- O'Brien Institute Department, St Vincent's Institute of Medical Research, Fitzroy, Victoria 3065, Australia.,Departments of Surgery and Medicine, University of Melbourne, Parkville, Victoria 3010, Australia.,National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, Singapore
| |
Collapse
|
32
|
Wang FF, Zhang JL, Ji Y, Yan XJ, Sun L, Zhu Y, Jin H. KLF2 mediates the suppressive effect of BDNF on diabetic intimal calcification by inhibiting HK1 induced endothelial-to-mesenchymal transition. Cell Signal 2022; 94:110324. [PMID: 35364229 DOI: 10.1016/j.cellsig.2022.110324] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 03/19/2022] [Accepted: 03/24/2022] [Indexed: 12/19/2022]
Abstract
Diabetic vascular calcification in the arterial intima is closely associated with endothelial-to-mesenchymal transition (EndMT). Glucose metabolism reprogramming is involved in EndMT. Although brain-derived neurotrophic factor (BDNF) and Krüppel-like family of transcription factor 2 (KLF2) play protective roles in the physiological activity of the vascular endothelium, the underlying mechanisms are unclear. Human umbilical vein endothelial cells (HUVECs) were incubated with diabetic osteogenic medium (DOM) to induce EndMT and accelerate osteogenic differentiation. Glycolysis in HUVECs was assessed by monitoring glucose uptake, lactate production, extracellular acidification rate and expression of key glycolytic enzymes. DOM induced EndMT and accelerated osteo-induction in HUVECs, which was alleviated by BDNF/tropomyosin receptor kinase B (TrkB) pathway. Mechanistically, DOM caused hyperactivation of glycolysis in HUVECs and inhibition of the BDNF/TrkB pathway. BDNF preserved KLF2 and downregulated hexokinase 1 (HK1) in HUVECs after DOM treatment. Furthermore, KLF2 interacted with HK1. Increased KLF2 alleviated HK1-mediated glucose metabolism abnormality. HK1 knockdown or a targeted glycolysis inhibitor suppressed EndMT, apoptosis, inflammation and vascular calcification of HUVECs after DOM exposure. This study suggests that KLF2 mediates the suppressive effect of BDNF on diabetic intimal calcification by inhibiting HK1-induced glucose metabolism reprogramming and the EndMT process.
Collapse
Affiliation(s)
- Fang-Fang Wang
- Department of Cardiology, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou 213000, PR China
| | - Jia-Li Zhang
- Department of Gastroenterology Centre, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou 213000, PR China
| | - Yuan Ji
- Department of Cardiology, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou 213000, PR China
| | - Xue-Jiao Yan
- Department of Cardiology, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou 213000, PR China
| | - Ling Sun
- Department of Cardiology, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou 213000, PR China
| | - Yi Zhu
- Department of Cardiology, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou 213000, PR China.
| | - Hong Jin
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, PR China.
| |
Collapse
|
33
|
Peng ML, Fu Y, Wu CW, Zhang Y, Ren H, Zhou SS. Signaling Pathways Related to Oxidative Stress in Diabetic Cardiomyopathy. Front Endocrinol (Lausanne) 2022; 13:907757. [PMID: 35784531 PMCID: PMC9240190 DOI: 10.3389/fendo.2022.907757] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 05/09/2022] [Indexed: 12/19/2022] Open
Abstract
Diabetes is a chronic metabolic disease that is increasing in prevalence and causes many complications. Diabetic cardiomyopathy (DCM) is a complication of diabetes that is associated with high mortality, but it is not well defined. Nevertheless, it is generally accepted that DCM refers to a clinical disease that occurs in patients with diabetes and involves ventricular dysfunction, in the absence of other cardiovascular diseases, such as coronary atherosclerotic heart disease, hypertension, or valvular heart disease. However, it is currently uncertain whether the pathogenesis of DCM is directly attributable to metabolic dysfunction or secondary to diabetic microangiopathy. Oxidative stress (OS) is considered to be a key component of its pathogenesis. The production of reactive oxygen species (ROS) in cardiomyocytes is a vicious circle, resulting in further production of ROS, mitochondrial DNA damage, lipid peroxidation, and the post-translational modification of proteins, as well as inflammation, cardiac hypertrophy and fibrosis, ultimately leading to cell death and cardiac dysfunction. ROS have been shown to affect various signaling pathways involved in the development of DCM. For instance, OS causes metabolic disorders by affecting the regulation of PPARα, AMPK/mTOR, and SIRT3/FOXO3a. Furthermore, OS participates in inflammation mediated by the NF-κB pathway, NLRP3 inflammasome, and the TLR4 pathway. OS also promotes TGF-β-, Rho-ROCK-, and Notch-mediated cardiac remodeling, and is involved in the regulation of calcium homeostasis, which impairs ATP production and causes ROS overproduction. In this review, we summarize the signaling pathways that link OS to DCM, with the intention of identifying appropriate targets and new antioxidant therapies for DCM.
Collapse
Affiliation(s)
- Meng-ling Peng
- Department of Cardiology, The First Hospital of Jilin University, Changchun, China
| | - Yu Fu
- Department of Cardiology, The First Hospital of Jilin University, Changchun, China
| | - Chu-wen Wu
- Department of Cardiology, The First Hospital of Jilin University, Changchun, China
| | - Ying Zhang
- Department of Cardiology, The First Hospital of Jilin University, Changchun, China
| | - Hang Ren
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, China
| | - Shan-shan Zhou
- Department of Cardiology, The First Hospital of Jilin University, Changchun, China
- *Correspondence: Shan-shan Zhou,
| |
Collapse
|
34
|
Wang H, Feng Z, Han X, Xing Y, Zhang X. Downregulation of acylglycerol kinase suppresses high glucose-induced endothelial-mesenchymal transition in HRECs through regulating the LPAR1/TGF-β/Notch signaling pathway. Can J Physiol Pharmacol 2021; 100:142-150. [PMID: 34559978 DOI: 10.1139/cjpp-2021-0265] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The endothelial-mesenchymal transition (EndMT) participates in the progression of diabetic retinopathy (DR), but cell-intrinsic factors modulating this process remain elusive. In this study, we explored the role of lysophosphatidic acid (LPA)-producing enzyme, acylglycerol kinase (AGK) in the EndMT of human retinal microvascular endothelial cells (HRECs) under high glucose (HG) conditions. We found that AGK was significantly elevated in HG-treated cells. In addition, AGK knockdown reversed the HG-induced EndMT in HRECs, which was evidenced by the increased epithelial markers (CD31 and VE-cadherin) and decreased mesenchymal markers (FSP1 and α-SMA). Furthermore, downregulation of AGK inhibited the HG-induced activation of TGF-β/Notch pathways, whereas exogenous TGF-β1 (10 ng/ml) impeded the inhibitory effects of AGK knockdown on HG-induced EndMT in HRECs. Additionally, the silence of AGK abolished the HG-induced upregulation of LPA and its receptor LPAR1, and overexpression of LPAR1 further rescued the AGK knockdown-mediated inhibition of the EndMT process. In conclusion, we demonstrate that downregulation of acylglycerol kinase suppresses high glucose-induced endothelial-mesenchymal transition in HRECs through regulating the LPAR1/TGF-β/Notch signaling pathway, indicating that AGK might be a potential therapeutic target for the treatment of DR.
Collapse
Affiliation(s)
- Haijing Wang
- First Affiliated Hospital of Harbin Medical University, 74559, Harbin, Heilongjiang, China;
| | - Zhuolei Feng
- First Affiliated Hospital of Harbin Medical University, 74559, Harbin, Heilongjiang, China;
| | - Xue Han
- First Affiliated Hospital of Harbin Medical University, 74559, Harbin, Heilongjiang, China;
| | - Yue Xing
- First Affiliated Hospital of Harbin Medical University, 74559, Harbin, Heilongjiang, China;
| | - Xiaomei Zhang
- First Affiliated Hospital of Harbin Medical University, 74559, Harbin, Heilongjiang, China;
| |
Collapse
|
35
|
Hua Q, Sun Z, Liu Y, Shen X, Zhao W, Zhu X, Xu P. KLK8 promotes the proliferation and metastasis of colorectal cancer via the activation of EMT associated with PAR1. Cell Death Dis 2021; 12:860. [PMID: 34552064 PMCID: PMC8458432 DOI: 10.1038/s41419-021-04149-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 07/26/2021] [Accepted: 09/09/2021] [Indexed: 12/24/2022]
Abstract
Kallikrein-related peptidase 8 (KLK8) acts as an oncogene or anti-oncogene in various tumours, and the abnormal expression of KLK8 is involved in the carcinogenesis of several tumours. However, the role of KLK8 in colorectal cancer (CRC) and the underlying mechanism remain largely unclear. In this study, the carcinogenic effect of KLK8 was determined via CCK-8 and colony formation assays in vitro and a xenograft model in nude mice in vivo. The metastasis-promoting effect of KLK8 was investigated with transwell migration and invasion assays and wound-healing assay in vitro and a metastasis model in nude mice in vivo. Bioinformatics analyses and mechanistic experiments were conducted to elucidate the molecular mechanism. Herein, we reported that KLK8 had a promotive effect on the proliferation, migration and invasion of RKO and SW480 cells. Epithelial-mesenchymal transition (EMT) played an important role in the promotive effects of KLK8 on CRC. In addition, protease-activated receptor-1 (PAR-1) antagonist SCH79797 but not protease-activated receptor-2 (PAR-2) antagonist FSLLRY-NH2 attenuated the proliferation, migration and invasion of KLK8-upregulated RKO and SW480 cells. PAR-1 antagonist SCH79797 reduced the tumour volume of xenograft model and decreased the metastatic nodules in the livers of metastasis model. Furthermore, SCH79797 could reverse the positive impact of KLK8 on the EMT process in CRC both in vitro and in vivo. Taken together, these findings demonstrated for the first time that KLK8 promoted EMT and CRC progression, and this effect might be, at least partly mediated by PAR1-dependent pathway.
Collapse
Affiliation(s)
- Qing Hua
- Department of Anesthesiology, Shanghai Cancer Center, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, No. 270 Dong an Road, 200032, Shanghai, China
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, 200032, Shanghai, China
| | - Zhirong Sun
- Department of Anesthesiology, Shanghai Cancer Center, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, No. 270 Dong an Road, 200032, Shanghai, China
| | - Yi Liu
- Department of Anesthesiology, Shanghai Cancer Center, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, No. 270 Dong an Road, 200032, Shanghai, China
| | - Xuefang Shen
- Department of Anesthesiology, Shanghai Cancer Center, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, No. 270 Dong an Road, 200032, Shanghai, China
| | - Weiwei Zhao
- Department of Integrated Therapy, Fudan University Shanghai Cancer Centre, Shanghai, China
| | - Xiaoyan Zhu
- Department of Physiology, Navy Medical University, 800 Xiangyin Road, 200433, Shanghai, China.
| | - Pingbo Xu
- Department of Anesthesiology, Shanghai Cancer Center, Fudan University, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, No. 270 Dong an Road, 200032, Shanghai, China.
| |
Collapse
|
36
|
Hua Q, Li T, Liu Y, Shen X, Zhu X, Xu P. Upregulation of KLK8 Predicts Poor Prognosis in Pancreatic Cancer. Front Oncol 2021; 11:624837. [PMID: 34395235 PMCID: PMC8362328 DOI: 10.3389/fonc.2021.624837] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Accepted: 06/08/2021] [Indexed: 12/24/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a growing cause of cancer-related mortality worldwide. Kallikrein-related peptidase 8 (KLK8) has potential clinical values in many cancers. However, the clinicopathological significances of KLK8 in PDAC remain unknown. We explored the relationship of KLK8 to clinicopathological features of PDAC based on public databases. KLK8 expression was examined in human PDAC tissues. Cell proliferation and apoptosis were evaluated in KLK8-overexpressed human pancreatic cancer cell lines Mia-paca-2 and Panc-1. The related signaling pathways of KLK8 involved in pancreatic cancer progression were analyzed by gene set enrichment analysis (GSEA) and further verified in in vitro studies. We found that KLK8 was up-regulated in tumor tissues in the TCGA-PAAD cohort, and was an independent prognostic factor for both overall survival and disease-free survival of PDAC. KLK8 mRNA and protein expressions were increased in PDAC tissues compared with para-cancerous pancreas. KLK8 overexpression exerted pro-proliferation and anti-apoptotic functions in Mia-paca-2 and Panc-1 cells. GSEA analysis showed that KLK8 was positively associated with PI3K-Akt-mTOR and Notch pathways. KLK8-induced pro-proliferation and anti-apoptotic effects in Mia-paca-2 and Panc-1 cells were attenuated by inhibitors for PI3K, Akt, and mTOR, but not by inhibitor for Notch. Furthermore, overexpression of KLK8 in Mia-paca-2 and Panc-1 cells significantly increased epidermal growth factor (EGF) levels in the culture media. EGF receptor (EGFR) inhibitor could block KLK8-induced activation of PI3K/Akt/mTOR pathway and attenuate pro-proliferation and anti-apoptotic of KLK8 in Mia-paca-2 and Panc-1 cells. In conclusion, KLK8 overexpression exerts pro-proliferation and anti-apoptotic functions in pancreatic cancer cells via EGF signaling-dependent activation of PI3K/Akt/mTOR pathway. Upregulated KLK8 in PDAC predicts poor prognosis and may be a potential therapeutic target for PDAC.
Collapse
Affiliation(s)
- Qing Hua
- Department of Anesthesiology, Shanghai Cancer Centre, Fudan University, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Tianjiao Li
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Department of Pancreatic Surgery, Shanghai Cancer Centre, Fudan University, Shanghai, China.,Shanghai Pancreatic Cancer Institute, Fudan University Shanghai, Shanghai, China.,Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Yixuan Liu
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Department of Clinical Laboratory, Shanghai Cancer Centre, Fudan University, Shanghai, China
| | - Xuefang Shen
- Department of Anesthesiology, Shanghai Cancer Centre, Fudan University, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiaoyan Zhu
- Department of Physiology, Navy Medical University, Shanghai, China
| | - Pingbo Xu
- Department of Anesthesiology, Shanghai Cancer Centre, Fudan University, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|