1
|
Husain I, Shah H, Jordan CZ, Natesh NR, Fay OK, Chen Y, Privratsky JR, Kitai H, Souma T, Varghese S, Howell DN, Thorp EB, Luo X. Targeting allograft inflammatory factor 1 reprograms kidney macrophages to enhance repair. J Clin Invest 2025; 135:e185146. [PMID: 39836477 PMCID: PMC11870741 DOI: 10.1172/jci185146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 01/08/2025] [Indexed: 01/23/2025] Open
Abstract
The role of macrophages (MΦs) remains incompletely understood in kidney injury and repair. The plasticity of MΦs offers an opportunity to polarize them toward mediating injury resolution in both native and transplanted kidneys undergoing ischemia and/or rejection. Here, we show that infiltrating kidney MΦs augmented their own allograft inflammatory factor 1 (AIF-1) expression after injury. Aif1 genetic deletion led to MΦ polarization toward a reparative phenotype while halting the development of kidney fibrosis. The enhanced repair was mediated by higher levels of antiinflammatory and proregenerative markers, leading to a reduction in cell death and an increase in proliferation of kidney tubular epithelial cells after ischemia followed by reperfusion injury (I/RI). Adoptive transfer of Aif1-/- MΦs into Aif1+/+ mice conferred protection against I/RI. Conversely, depletion of MΦs reversed the tissue-reparative effects in Aif1-/- mice. We further demonstrated increased expression of AIF-1 in human kidney biopsies from native kidneys with acute kidney injury or chronic kidney disease, as well as in biopsies from kidney allografts undergoing acute or chronic rejection. We conclude that AIF-1 is a MΦ marker of renal inflammation, and its targeting uncouples MΦ reparative functions from profibrotic functions. Thus, therapies inhibiting AIF-1 when ischemic injury is inevitable have the potential to reduce the global burden of kidney disease.
Collapse
Affiliation(s)
- Irma Husain
- Division of Nephrology, Department of Medicine, and
- Duke Transplant Center, Duke University School of Medicine, Durham, North Carolina, USA
| | - Holly Shah
- Division of Nephrology, Department of Medicine, and
| | | | - Naveen R. Natesh
- Department of Biomedical Engineering, Duke University Pratt School of Engineering, Durham, North Carolina, USA
| | | | | | | | - Hiroki Kitai
- Division of Nephrology, Department of Medicine, and
| | | | - Shyni Varghese
- Department of Biomedical Engineering, Duke University Pratt School of Engineering, Durham, North Carolina, USA
- Department of Mechanical Engineering and Materials Science, and
- Department of Orthopaedic Surgery, Duke University, Durham, North Carolina, USA
| | | | - Edward B. Thorp
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Xunrong Luo
- Division of Nephrology, Department of Medicine, and
- Duke Transplant Center, Duke University School of Medicine, Durham, North Carolina, USA
| |
Collapse
|
2
|
Svetláková BB, Líšková VP, Barančík M. Wnt Signaling Inhibitors as Therapeutic Approach in Ischemic Heart Disease. Molecules 2024; 29:5958. [PMID: 39770047 PMCID: PMC11677181 DOI: 10.3390/molecules29245958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 12/05/2024] [Accepted: 12/16/2024] [Indexed: 01/11/2025] Open
Abstract
Wnt (wingless-type MMTV integration site family) signaling is an evolutionary conserved system highly active during embryogenesis, but in adult hearts has low activities under normal conditions. It is essential for a variety of physiological processes including stem cell regeneration, proliferation, migration, cell polarity, and morphogenesis, thereby ensuring homeostasis and regeneration of cardiac tissue. Its dysregulation and excessive activation during pathological conditions leads to morphological and functional changes in the heart resulting in impaired myocardial regeneration under pathological conditions such as myocardial infarction, heart failure, and coronary artery disease. Several groups of Wnt inhibitors have demonstrated the ability to modulate the Wnt pathway and thereby significantly reduce fibrosis and improve cardiac function after myocardial ischemia. Their inhibitory effect can be realized at multiple levels, which include the inhibition of Wnt ligands, the inhibition of Frizzled receptors, the stabilization of the β-catenin destruction complex, and the disruption of nuclear β-catenin interactions. In this review, we overview the function of Wnt signaling in responses of cardiac cells to pathological conditions, especially ischemic heart disease, with an emphasis on the use of inhibitors of this signaling as a therapeutic approach. Finally, we summarize the current knowledge about the potential of the targeting of Wnt signaling in therapeutic applications.
Collapse
Affiliation(s)
| | | | - Miroslav Barančík
- Centre of Experimental Medicine, Slovak Academy of Sciences, 841 04 Bratislava, Slovakia; (B.B.S.); (V.P.L.)
| |
Collapse
|
3
|
Yuan X, Pan L, Zhang C, Zhu Q, Huang Z, Qin Y, Zhang G, Feng Z, Yang C, Hou N. Empagliflozin improves pressure-overload-induced cardiac hypertrophy by inhibiting the canonical Wnt/β-catenin signaling pathway. Front Pharmacol 2024; 15:1499542. [PMID: 39664517 PMCID: PMC11631586 DOI: 10.3389/fphar.2024.1499542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Accepted: 10/30/2024] [Indexed: 12/13/2024] Open
Abstract
Background Empagliflozin (EMPA) is an SGLT-2 inhibitor that can control hyperglycemia. Clinical trials have indicated its cardio-protective effects against cardiac remodeling in diabetes or non-diabetes patients. However, the underlying molecular mechanisms of EMPA's cardio-protective effects remain elusive. Methods We evaluated whether the EMPA attenuated the pressure-overload-induced cardiac hypertrophy by inhibiting the Wnt/β-catenin pathway. Furthermore, the effects of the EMPA on a mouse model of transverse aortic constriction (TAC) induced cardiac hypertrophy was also evaluated. Mice were administrated with 0.5% CMC-Na as a vehicle or EMPA (10 mg/kg/day, daily, throughout the study) by intragastric gavage. Results The in vivo echocardiography and histologic morphological analyses revealed that EMPA attenuated TAC-induced cardiac hypertrophy. Moreover, it also ameliorated TAC-induced cardiac fibrosis and decreased the cell size of the cardiomyocytes in isolated adult cardiomyocytes. Molecular mechanism analysis revealed that the EMPA reduced the TAC-induced enhanced expression of the Wnt/β-catenin pathway in vivo. For in vitro assessments, isolated neonatal rat cardiomyocytes (NRCMs) were treated with Angiotensin II (AngII) and EMPA; the results showed that in the absence of EMPA, the expression of the Wnt/β-catenin pathway was enhanced. In the trans-genetic heterozygous β-catenin deletion mice, EMPA attenuated TAC-induced cardiac remodeling by reducing the Wnt/β-catenin pathway. In addition, molecular docking analysis indicated that EMPA interacts with FZD4 to inhibit the TAC and AngII induced Wnt/β-catenin pathway in cardiomyocytes. Conclusion Our study illustrated that EMPA might directly interact with FZD4 to inhibit the TAC and AngII-induced activation of the Wnt/β-catenin pathway to attenuate the adverse cardiac remodeling.
Collapse
Affiliation(s)
- Xun Yuan
- The Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, China
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou Medical University, NMPA Key Laboratory for Clinical Research and Evaluation of Drug for Thoracic Diseases, Guangzhou Medical University, Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology, Guangzhou Medical University, Guangzhou, China
| | - Li Pan
- Department of Physiology, School of Basic Medicine Sciences, Guangzhou Health Science College, Guangzhou, China
| | - Chi Zhang
- School of Stomatology, Guangzhou Medical University, Guangzhou, China
| | - Qiulian Zhu
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou Medical University, NMPA Key Laboratory for Clinical Research and Evaluation of Drug for Thoracic Diseases, Guangzhou Medical University, Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology, Guangzhou Medical University, Guangzhou, China
| | - Zexin Huang
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou Medical University, NMPA Key Laboratory for Clinical Research and Evaluation of Drug for Thoracic Diseases, Guangzhou Medical University, Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology, Guangzhou Medical University, Guangzhou, China
| | - Yuan Qin
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou Medical University, NMPA Key Laboratory for Clinical Research and Evaluation of Drug for Thoracic Diseases, Guangzhou Medical University, Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology, Guangzhou Medical University, Guangzhou, China
| | - Guiping Zhang
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou Medical University, NMPA Key Laboratory for Clinical Research and Evaluation of Drug for Thoracic Diseases, Guangzhou Medical University, Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology, Guangzhou Medical University, Guangzhou, China
| | - Zhimei Feng
- The Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, China
| | - Caixian Yang
- The Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, China
| | - Ning Hou
- The Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, China
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou Medical University, NMPA Key Laboratory for Clinical Research and Evaluation of Drug for Thoracic Diseases, Guangzhou Medical University, Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
4
|
Yin W, Chen Y, Wang W, Guo M, Tong L, Zhang M, Wang Z, Yuan H. Macrophage-mediated heart repair and remodeling: A promising therapeutic target for post-myocardial infarction heart failure. J Cell Physiol 2024; 239:e31372. [PMID: 39014935 DOI: 10.1002/jcp.31372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 06/06/2024] [Accepted: 06/25/2024] [Indexed: 07/18/2024]
Abstract
Heart failure (HF) remains prevalent in patients who survived myocardial infarction (MI). Despite the accessibility of the primary percutaneous coronary intervention and medications that alleviate ventricular remodeling with functional improvement, there is an urgent need for clinicians and basic scientists to further reveal the mechanisms behind post-MI HF as well as investigate earlier and more efficient treatment after MI. Growing numbers of studies have highlighted the crucial role of macrophages in cardiac repair and remodeling following MI, and timely intervention targeting the immune response via macrophages may represent a promising therapeutic avenue. Recently, technology such as single-cell sequencing has provided us with an updated and in-depth understanding of the role of macrophages in MI. Meanwhile, the development of biomaterials has made it possible for macrophage-targeted therapy. Thus, an overall and thorough understanding of the role of macrophages in post-MI HF and the current development status of macrophage-based therapy will assist in the further study and development of macrophage-targeted treatment for post-infarction cardiac remodeling. This review synthesizes the spatiotemporal dynamics, function, mechanism and signaling of macrophages in the process of HF after MI, as well as discusses the emerging bio-materials and possible therapeutic agents targeting macrophages for post-MI HF.
Collapse
Affiliation(s)
- Wenchao Yin
- Department of Cardiology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
| | - Yong Chen
- Department of Emergency, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Wenjun Wang
- Department of Intensive Care Unit, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Mengqi Guo
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Lingjun Tong
- Medical Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Mingxiang Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Department of Cardiology, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital, Shandong University, Jinan, Shandong, China
| | - Zhaoyang Wang
- Department of Cardiology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Haitao Yuan
- Department of Cardiology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
5
|
Huang J, Shi Z, Huang Z, Lai S. Identification and Verification of Potential Markers Related to Myocardial Fibrosis by Bioinformatics Analysis. Biochem Genet 2024:10.1007/s10528-024-10937-9. [PMID: 39387979 DOI: 10.1007/s10528-024-10937-9] [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: 10/07/2023] [Accepted: 10/01/2024] [Indexed: 10/15/2024]
Abstract
Mounting evidence indicates that myocardial fibrosis (MF) is frequently intertwined with immune and metabolic disorders. This comprehensive review aims to delve deeply into the crucial role of immune-related signature genes in the pathogenesis and progression of MF. This exploration holds significant importance as understanding the underlying mechanisms of MF is essential for developing effective diagnostic and therapeutic strategies. The dataset GSE9735 about myocardial fibrosis and non-fibrosis was downloaded from GEO database. Differentially expressed genes (DEGs) were identified by 'limma' package in R software. Then, the biological function of DEG was determined by gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis. XCell was used to estimate the composition pattern of matrix and immune cells. Protein-protein interaction (PPI) network was constructed based on STRING analysis software, and Hub genes were screened and functional modules were analyzed. The correlation between hub genes and immune cell subtypes was analyzed. Hub genes with |correlation coefficient|> 0.45 and p-value < 0.05 were used as characteristic biomarkers. Finally, the logistic regression model is used to verify the three markers in the training set and verification set (GSE97358 and GSE225336). A total of 635 DEGs were identified. Functional enrichment analysis shows that inflammation and immune response, extracellular matrix and structural remodeling play an important role in the pathological mechanism of MF. Immune cell infiltration analysis showed that immune cells (Plasma cells, Eosinophils, Chondrocytes and Th2 cells) significantly changed in MF pathological conditions. In PPI network analysis, IL1β, TTN, PTPRC, IGF1, ALDH1A1, CYP26A1, ALDH1A3, MYH11, CSF1R and CD80 were identified as hub genes, among which IL1β, CYP26A1 and GNG2 were regarded as immune-related characteristic markers. The AUC scores of the three biomarkers are all above 0.65, which proves that they have a good discrimination effect in MF. In this study, three immune-related genes were identified as diagnostic biomarkers of MF, which provided a new perspective for exploring the molecular mechanism of MF. This study takes a comprehensive approach to understanding the intricate relationship between myocardial fibrosis and immune metabolism. By identifying key immune-related biomarkers, this study not only reveals the molecular basis of myocardial fibrosis but also paves the way for the development of novel diagnostic tools and therapeutic strategies. These findings are critical for improving patient prognosis and may have broader implications for studying and treating other cardiovascular diseases associated with immune dysregulation.
Collapse
Affiliation(s)
- Jiazhuo Huang
- Department of Cardiology, The First People's Hospital of Zhaoqing City, No.9 Donggang East Road, Zhaoqing, 526040, Guangdong, China
| | - Zhentao Shi
- Department of Cardiology, The First People's Hospital of Zhaoqing City, No.9 Donggang East Road, Zhaoqing, 526040, Guangdong, China
| | - Zhifeng Huang
- Department of Cardiology, The First People's Hospital of Zhaoqing City, No.9 Donggang East Road, Zhaoqing, 526040, Guangdong, China
| | - Shaobin Lai
- Department of Cardiology, The First People's Hospital of Zhaoqing City, No.9 Donggang East Road, Zhaoqing, 526040, Guangdong, China.
| |
Collapse
|
6
|
Mohamad HE, Askar ME, Shaheen MA, Baraka NM, Mahmoud YK. Sacubitril/valsartan alleviates sunitinib-induced cardiac fibrosis and oxidative stress via improving TXNIP/TRX system and downregulation of NF-ĸB/Wnt/β-catenin/SOX9 signaling. Int Immunopharmacol 2024; 132:111963. [PMID: 38560962 DOI: 10.1016/j.intimp.2024.111963] [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: 12/21/2023] [Revised: 03/12/2024] [Accepted: 03/26/2024] [Indexed: 04/04/2024]
Abstract
We aimed in this study to investigate the possible cardioprotective effects of sacubitril/valsartan against sunitinib-induced cardiac fibrosis (CF) and oxidative stress via targeting thioredoxin-interacting protein/thioredoxin (TXNIP/TRX) system and nuclear factor-kappa B (NF-κB)/Wingless-related MMTV integration site (Wnt)/β-catenin/Sex-determining region Y box 9 (SOX9) signaling. CF was induced in male Wistar albino rats by cumulative dose of sunitinib (300 mg/kg, given over 4 weeks as: 25 mg/kg orally, three times a week), which were co-treated with sacubitril/valsartan (68 mg/kg/day, orally) for four weeks. Significant elevation in blood pressure, cardiac inflammatory and fibrotic markers besides cardiac dysfunction were observed. These alterations were associated with disruption of TXNIP/TRX system, upregulation of NF-κB/Wnt/β-catenin/SOX9 pathway along with marked increase in lysyl oxidase (LOX) and matrix metalloproteinase-1 (MMP-1) expressions and extensive deposition of collagen fibers in cardiac tissues. Luckily, sacubitril/valsartan was able to reverse all of the aforementioned detrimental effects in sunitinib-administered rats. These findings illustrate a potential role of sacubitril/valsartan in alleviating CF and oxidative stress induced by sunitinib via antioxidant, anti-inflammatory and antifibrotic properties. These remarkable effects of sacubitril/valsartan were mediated by its ability to improve TXNIP/TRX system and downregulate NF-κB/Wnt/β-catenin/SOX9 signaling in addition to decreasing LOX and MMP-1 expressions in cardiac tissues. In summary, this study highlights sacubitril/valsartan as a potential therapeutic agent in mitigating CF and oxidative stress especially in cancer cases treated with sunitinib.
Collapse
Affiliation(s)
- Hoda E Mohamad
- Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
| | - Mervat E Askar
- Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt.
| | - Mohamed A Shaheen
- Department of Histology & Cell Biology, Faculty of Medicine, Zagazig University, Zagazig 44519, Egypt
| | - Nourhan M Baraka
- Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
| | - Yasmin K Mahmoud
- Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
| |
Collapse
|
7
|
Horitani K, Shiojima I. Wnt signaling in cardiac development and heart diseases. In Vitro Cell Dev Biol Anim 2024; 60:482-488. [PMID: 38709417 PMCID: PMC11126472 DOI: 10.1007/s11626-024-00917-z] [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: 01/03/2024] [Accepted: 04/22/2024] [Indexed: 05/07/2024]
Abstract
The Wnt signaling pathway is a fundamental cellular communication system with extensive implications in various organs including the heart. In cardiac homeostasis, it governs essential processes like cellular proliferation, differentiation, and apoptosis, ensuring the heart's structural and functional integrity from embryonic stages and throughout life. Both canonical and non-canonical Wnt signaling pathways play a critical role during embryonic heart development in a region- and stage-specific manner. Canonical Wnt signaling also plays a significant role in heart diseases such as myocardial infarction and heart failure. However, the role of non-canonical Wnt signaling in heart diseases has not been fully elucidated. Wnt5a is a major ligand that activates non-canonical Wnt pathway, and recent studies start to clarify the role of the Wnt5a signaling axis in cardiac health and disease. In this review, we will briefly summarize the previous findings on the role of Wnt signaling pathways in heart development and diseases, and then focus on the role of Wnt5a signaling in heart failure progression. The multifaceted roles of the Wnt signaling pathway highlight its therapeutic potential for various types of heart diseases.
Collapse
Affiliation(s)
- Keita Horitani
- Department of Medicine II, Kansai Medical University, 2-5-1, Shin-Machi, Hirakata, Osaka, 573-1010, Japan
| | - Ichiro Shiojima
- Department of Medicine II, Kansai Medical University, 2-5-1, Shin-Machi, Hirakata, Osaka, 573-1010, Japan.
| |
Collapse
|
8
|
Zelikson N, Ben S, Caspi M, Tarabe R, Shaleve Y, Pri-Paz Basson Y, Tayer-Shifman O, Goldberg E, Kivity S, Rosin-Arbesfeld R. Wnt signaling regulates chemokine production and cell migration of circulating human monocytes. Cell Commun Signal 2024; 22:229. [PMID: 38622714 PMCID: PMC11020454 DOI: 10.1186/s12964-024-01608-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Accepted: 04/09/2024] [Indexed: 04/17/2024] Open
Abstract
The β-catenin dependent canonical Wnt signaling pathway plays a crucial role in maintaining normal homeostasis. However, when dysregulated, Wnt signaling is closely associated with various pathological conditions, including inflammation and different types of cancer.Here, we show a new connection between the leukocyte inflammatory response and the Wnt signaling pathway. Specifically, we demonstrate that circulating human primary monocytes express distinct Wnt signaling components and are susceptible to stimulation by the classical Wnt ligand-Wnt-3a. Although this stimulation increased the levels of β-catenin protein, the expression of the classical Wnt-target genes was not affected. Intriguingly, treating circulating human monocytes with Wnt-3a induces the secretion of cytokines and chemokines, enhancing monocyte migration. Mechanistically, the enhanced monocyte migration in response to Wnt stimuli is mediated through CCL2, a strong monocyte-chemoattractant.To further explore the physiological relevance of these findings, we conducted ex-vivo experiments using blood samples of patients with rheumatic joint diseases (RJD) - conditions where monocytes are known to be dysfunctional. Wnt-3a generated a unique cytokine expression profile, which was significantly distinct from that observed in monocytes obtained from healthy donors.Thus, our results provide the first evidence that Wnt-3a may serve as a potent stimulator of monocyte-driven immune processes. These findings contribute to our understanding of inflammatory diseases and, more importantly, shed light on the role of a core signaling pathway in the circulation.
Collapse
Affiliation(s)
- Natalie Zelikson
- Department of Clinical Microbiology and Immunology, Faculty of Medical & Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Shaina Ben
- Department of Clinical Microbiology and Immunology, Faculty of Medical & Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Michal Caspi
- Department of Clinical Microbiology and Immunology, Faculty of Medical & Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Raneen Tarabe
- Department of Clinical Microbiology and Immunology, Faculty of Medical & Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Yonatan Shaleve
- Department of Medicine F, Rabin Medical Center, Beilinson Hospital, Petah Tikva, Israel
- Faculty of Medical & Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Yael Pri-Paz Basson
- Rheumatology Unit, Meir Medical Center, Kfar Saba, Israel
- Faculty of Medical & Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Oshrat Tayer-Shifman
- Rheumatology Unit, Meir Medical Center, Kfar Saba, Israel
- Faculty of Medical & Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Elad Goldberg
- Department of Medicine F, Rabin Medical Center, Beilinson Hospital, Petah Tikva, Israel
- Faculty of Medical & Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Shaye Kivity
- Rheumatology Unit, Meir Medical Center, Kfar Saba, Israel
- Faculty of Medical & Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Rina Rosin-Arbesfeld
- Department of Clinical Microbiology and Immunology, Faculty of Medical & Health Sciences, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
9
|
Liu Y, Hu G, Jia Y, Qin L, Xu L, Chang Y, Li B, Li H. Wnt10b knockdown regulates the relative balance of adipose tissue-resident T cells and inhibits white fat deposition. Mol Biol Rep 2024; 51:272. [PMID: 38302806 DOI: 10.1007/s11033-024-09249-3] [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: 09/21/2023] [Accepted: 01/12/2024] [Indexed: 02/03/2024]
Abstract
BACKGROUND Wnt10b is one of critical Wnt family members that being involved in networks controlling stemness, pluripotency and cell fate decisions. However, its role in adipose-resident T lymphocytes and further in fat metabolism yet remains largely unknown. METHODS AND RESULTS In the present study, we demonstrated a distinctive effect for Wnt10b on the relative balance of T lymphocytes in adipose tissue by using a Wnt10b knockdown mouse model. Wnt10b knockdown led to a reduction of adipose-resident CD4+ T cells and an elevation of Foxp3+/CD4+ Treg cells. Wnt10b-knockdown mice fed with standard diet showed less white fat deposition owing to the suppressed adipogenic process. Moreover, under high fat diet conditions, Wnt10b knockdown resulted in an alleviated obesity symptoms, as well as an improvement of glucose homeostasis and hepatic steatosis. CONCLUSIONS Collectively, we reveal an unexpected and novel function for Wnt10b in mediating the frequency of adipose-resident T cell subsets, that when knockdown skewing toward a Treg-dominated phenotype and further improving fat metabolism.
Collapse
Affiliation(s)
- Yan Liu
- College of Life Sciences, Shandong Agricultural University, Tai'an, 271018, China
| | - Geng Hu
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an, 271018, China
| | - Yanxin Jia
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an, 271018, China
| | - Lining Qin
- College of Life Sciences, Shandong Agricultural University, Tai'an, 271018, China
| | - Longfei Xu
- College of Life Sciences, Shandong Agricultural University, Tai'an, 271018, China
| | - Yaxin Chang
- College of Life Sciences, Shandong Agricultural University, Tai'an, 271018, China
| | - Bin Li
- College of Life Sciences, Shandong Agricultural University, Tai'an, 271018, China
| | - Haifang Li
- College of Life Sciences, Shandong Agricultural University, Tai'an, 271018, China.
| |
Collapse
|
10
|
Li Y, Li X, Guo D, Meng L, Feng X, Zhang Y, Pan S. Immune dysregulation and macrophage polarization in peri-implantitis. Front Bioeng Biotechnol 2024; 12:1291880. [PMID: 38347915 PMCID: PMC10859439 DOI: 10.3389/fbioe.2024.1291880] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 01/09/2024] [Indexed: 02/15/2024] Open
Abstract
The term "peri-implantitis" (peri-implantitis) refers to an inflammatory lesion of the mucosa surrounding an endosseous implant and a progressive loss of the peri-implant bone that supports the implant. Recently, it has been suggested that the increased sensitivity of implants to infection and the quick elimination of supporting tissue after infection may be caused by a dysregulated peri-implant mucosal immune response. Macrophages are polarized in response to environmental signals and play multiple roles in peri-implantitis. In peri-implantitis lesion samples, recent investigations have discovered a considerable increase in M1 type macrophages, with M1 type macrophages contributing to the pro-inflammatory response brought on by bacteria, whereas M2 type macrophages contribute to inflammation remission and tissue repair. In an effort to better understand the pathogenesis of peri-implantitis and suggest potential immunomodulatory treatments for peri-implantitis in the direction of macrophage polarization patterns, this review summarizes the research findings related to macrophage polarization in peri-implantitis and compares them with periodontitis.
Collapse
Affiliation(s)
- Yue Li
- Department of Prosthodontics, Peking University School and Hospital of Stomatology and National Center for Stomatology and National Clinical Research Center for Oral Diseases and National Engineering Research Center of Oral Biomaterials and Digital Medical Devices and Beijing Key Laboratory of Digital Stomatology and Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health and NMPA Key Laboratory for Dental Materials, Beijing, China
| | - Xue Li
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Radiation Medicine, Beijing, China
| | - Danni Guo
- Department of Prosthodontics, Peking University School and Hospital of Stomatology and National Center for Stomatology and National Clinical Research Center for Oral Diseases and National Engineering Research Center of Oral Biomaterials and Digital Medical Devices and Beijing Key Laboratory of Digital Stomatology and Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health and NMPA Key Laboratory for Dental Materials, Beijing, China
| | - Lingwei Meng
- Department of Prosthodontics, Peking University School and Hospital of Stomatology and National Center for Stomatology and National Clinical Research Center for Oral Diseases and National Engineering Research Center of Oral Biomaterials and Digital Medical Devices and Beijing Key Laboratory of Digital Stomatology and Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health and NMPA Key Laboratory for Dental Materials, Beijing, China
| | - Xianghui Feng
- Department of Periodontology, Peking University School and Hospital of Stomatology and National Center for Stomatology and National Clinical Research Center for Oral Diseases and National Engineering Research Center of Oral Biomaterials and Digital Medical Devices and Beijing Key Laboratory of Digital Stomatology and Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health and NMPA Key Laboratory for Dental Materials, Beijing, China
| | - Yi Zhang
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Radiation Medicine, Beijing, China
| | - Shaoxia Pan
- Department of Prosthodontics, Peking University School and Hospital of Stomatology and National Center for Stomatology and National Clinical Research Center for Oral Diseases and National Engineering Research Center of Oral Biomaterials and Digital Medical Devices and Beijing Key Laboratory of Digital Stomatology and Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health and NMPA Key Laboratory for Dental Materials, Beijing, China
| |
Collapse
|
11
|
Abstract
Despite significant advances in the field of transplantation in the past two decades, current clinically available therapeutic options for immunomodulation remain fairly limited. The advent of calcineurin inhibitor-based immunosuppression has led to significant success in improving short-term graft survival; however, improvements in long-term graft survival have stalled. Solid organ transplantation provides a unique opportunity for immunomodulation of both the donor organ prior to implantation and the recipient post transplantation. Furthermore, therapies beyond targeting the adaptive immune system have the potential to ameliorate ischemic injury to the allograft and halt its aging process, augment its repair, and promote recipient immune tolerance. Other recent advances include expanding the donor pool by reducing organ discard, and bioengineering and genetically modifying organs from other species to generate transplantable organs. Therapies discussed here will likely be most impactful if individualized on the basis of specific donor and recipient considerations.
Collapse
Affiliation(s)
- Irma Husain
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA;
| | - Xunrong Luo
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA;
- Duke Transplant Center, Duke University School of Medicine, Durham, North Carolina, USA
| |
Collapse
|
12
|
Zhang M, Liu Q, Meng H, Duan H, Liu X, Wu J, Gao F, Wang S, Tan R, Yuan J. Ischemia-reperfusion injury: molecular mechanisms and therapeutic targets. Signal Transduct Target Ther 2024; 9:12. [PMID: 38185705 PMCID: PMC10772178 DOI: 10.1038/s41392-023-01688-x] [Citation(s) in RCA: 151] [Impact Index Per Article: 151.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 08/29/2023] [Accepted: 10/18/2023] [Indexed: 01/09/2024] Open
Abstract
Ischemia-reperfusion (I/R) injury paradoxically occurs during reperfusion following ischemia, exacerbating the initial tissue damage. The limited understanding of the intricate mechanisms underlying I/R injury hinders the development of effective therapeutic interventions. The Wnt signaling pathway exhibits extensive crosstalk with various other pathways, forming a network system of signaling pathways involved in I/R injury. This review article elucidates the underlying mechanisms involved in Wnt signaling, as well as the complex interplay between Wnt and other pathways, including Notch, phosphatidylinositol 3-kinase/protein kinase B, transforming growth factor-β, nuclear factor kappa, bone morphogenetic protein, N-methyl-D-aspartic acid receptor-Ca2+-Activin A, Hippo-Yes-associated protein, toll-like receptor 4/toll-interleukine-1 receptor domain-containing adapter-inducing interferon-β, and hepatocyte growth factor/mesenchymal-epithelial transition factor. In particular, we delve into their respective contributions to key pathological processes, including apoptosis, the inflammatory response, oxidative stress, extracellular matrix remodeling, angiogenesis, cell hypertrophy, fibrosis, ferroptosis, neurogenesis, and blood-brain barrier damage during I/R injury. Our comprehensive analysis of the mechanisms involved in Wnt signaling during I/R reveals that activation of the canonical Wnt pathway promotes organ recovery, while activation of the non-canonical Wnt pathways exacerbates injury. Moreover, we explore novel therapeutic approaches based on these mechanistic findings, incorporating evidence from animal experiments, current standards, and clinical trials. The objective of this review is to provide deeper insights into the roles of Wnt and its crosstalk signaling pathways in I/R-mediated processes and organ dysfunction, to facilitate the development of innovative therapeutic agents for I/R injury.
Collapse
Affiliation(s)
- Meng Zhang
- The Collaborative Innovation Center, Jining Medical University, Jining, Shandong, 272067, China
| | - Qian Liu
- Clinical Medical College, Jining Medical University, Jining, Shandong, 272067, China
| | - Hui Meng
- Clinical Medical College, Jining Medical University, Jining, Shandong, 272067, China
| | - Hongxia Duan
- Clinical Medical College, Jining Medical University, Jining, Shandong, 272067, China
| | - Xin Liu
- Second Clinical Medical College, Jining Medical University, Jining, Shandong, 272067, China
| | - Jian Wu
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Fei Gao
- The Collaborative Innovation Center, Jining Medical University, Jining, Shandong, 272067, China
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Shijun Wang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China.
| | - Rubin Tan
- Department of Physiology, Basic medical school, Xuzhou Medical University, Xuzhou, 221004, China.
| | - Jinxiang Yuan
- The Collaborative Innovation Center, Jining Medical University, Jining, Shandong, 272067, China.
| |
Collapse
|
13
|
Song Z, Cheng Y, Chen M, Xie X. Macrophage polarization in bone implant repair: A review. Tissue Cell 2023; 82:102112. [PMID: 37257287 DOI: 10.1016/j.tice.2023.102112] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 04/10/2023] [Accepted: 05/15/2023] [Indexed: 06/02/2023]
Abstract
Macrophages (MΦ) are highly adaptable and functionally polarized cells that play a crucial role in various physiological and pathological processes. Typically, MΦ differentiate into two distinct subsets: the proinflammatory (M1) and anti-inflammatory (M2) phenotypes. Due to their potent immunomodulatory and anti-inflammatory properties, MΦ have garnered significant attention in recent decades. In the context of bone implant repair, the immunomodulatory function of MΦ is of paramount importance. Depending on their polarization phenotype, MΦ can exert varying effects on osteogenesis, angiogenesis, and the inflammatory response around the implant. This paper provides an overview of the immunomodulatory and inflammatory effects of MΦ polarization in the repair of bone implants.
Collapse
Affiliation(s)
- Zhengzheng Song
- Central South University Xiangya Stomatological Hospital, Central South University, Changsha 410078, Hunan, China; Xiangya School of Stomatology, Central South University, Changsha 410008, Hunan, China
| | - Yuxi Cheng
- Central South University Xiangya Stomatological Hospital, Central South University, Changsha 410078, Hunan, China; Xiangya School of Stomatology, Central South University, Changsha 410008, Hunan, China
| | - Minmin Chen
- Central South University Xiangya Stomatological Hospital, Central South University, Changsha 410078, Hunan, China.
| | - Xiaoli Xie
- Central South University Xiangya Stomatological Hospital, Central South University, Changsha 410078, Hunan, China; Hunan Key Laboratory of Oral Health Research, Changsha 410008, Hunan, China.
| |
Collapse
|
14
|
Schary Y, Rotem I, Caller T, Lewis N, Shaihov-Teper O, Brzezinski RY, Lendengolts D, Raanani E, Sternik L, Naftali-Shani N, Leor J. CRISPR-Cas9 editing of TLR4 to improve the outcome of cardiac cell therapy. Sci Rep 2023; 13:4481. [PMID: 36934130 PMCID: PMC10024743 DOI: 10.1038/s41598-023-31286-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 03/09/2023] [Indexed: 03/20/2023] Open
Abstract
Inflammation and fibrosis limit the reparative properties of human mesenchymal stromal cells (hMSCs). We hypothesized that disrupting the toll-like receptor 4 (TLR4) gene would switch hMSCs toward a reparative phenotype and improve the outcome of cell therapy for infarct repair. We developed and optimized an improved electroporation protocol for CRISPR-Cas9 gene editing. This protocol achieved a 68% success rate when applied to isolated hMSCs from the heart and epicardial fat of patients with ischemic heart disease. While cell editing lowered TLR4 expression in hMSCs, it did not affect classical markers of hMSCs, proliferation, and migration rate. Protein mass spectrometry analysis revealed that edited cells secreted fewer proteins involved in inflammation. Analysis of biological processes revealed that TLR4 editing reduced processes linked to inflammation and extracellular organization. Furthermore, edited cells expressed less NF-ƙB and secreted lower amounts of extracellular vesicles and pro-inflammatory and pro-fibrotic cytokines than unedited hMSCs. Cell therapy with both edited and unedited hMSCs improved survival, left ventricular remodeling, and cardiac function after myocardial infarction (MI) in mice. Postmortem histologic analysis revealed clusters of edited cells that survived in the scar tissue 28 days after MI. Morphometric analysis showed that implantation of edited cells increased the area of myocardial islands in the scar tissue, reduced the occurrence of transmural scar, increased scar thickness, and decreased expansion index. We show, for the first time, that CRISPR-Cas9-based disruption of the TLR4-gene reduces pro-inflammatory polarization of hMSCs and improves infarct healing and remodeling in mice. Our results provide a new approach to improving the outcomes of cell therapy for cardiovascular diseases.
Collapse
Affiliation(s)
- Yeshai Schary
- Neufeld and Tamman Cardiovascular Research Institutes, Sheba Medical Center, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
- Heart Center, Sheba Medical Center, 52621, Tel-Hashomer, Israel
| | - Itai Rotem
- Neufeld and Tamman Cardiovascular Research Institutes, Sheba Medical Center, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
- Heart Center, Sheba Medical Center, 52621, Tel-Hashomer, Israel
| | - Tal Caller
- Neufeld and Tamman Cardiovascular Research Institutes, Sheba Medical Center, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
- Heart Center, Sheba Medical Center, 52621, Tel-Hashomer, Israel
| | - Nir Lewis
- Neufeld and Tamman Cardiovascular Research Institutes, Sheba Medical Center, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
- Heart Center, Sheba Medical Center, 52621, Tel-Hashomer, Israel
| | - Olga Shaihov-Teper
- Neufeld and Tamman Cardiovascular Research Institutes, Sheba Medical Center, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
- Heart Center, Sheba Medical Center, 52621, Tel-Hashomer, Israel
| | - Rafael Y Brzezinski
- Neufeld and Tamman Cardiovascular Research Institutes, Sheba Medical Center, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
- Heart Center, Sheba Medical Center, 52621, Tel-Hashomer, Israel
| | - Daria Lendengolts
- Neufeld and Tamman Cardiovascular Research Institutes, Sheba Medical Center, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
- Heart Center, Sheba Medical Center, 52621, Tel-Hashomer, Israel
| | - Ehud Raanani
- Heart Center, Sheba Medical Center, 52621, Tel-Hashomer, Israel
- Department of Cardiac Surgery, Leviev Cardiothoracic and Vascular Center, Sheba Medical Center, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Leonid Sternik
- Heart Center, Sheba Medical Center, 52621, Tel-Hashomer, Israel
- Department of Cardiac Surgery, Leviev Cardiothoracic and Vascular Center, Sheba Medical Center, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Nili Naftali-Shani
- Neufeld and Tamman Cardiovascular Research Institutes, Sheba Medical Center, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
- Heart Center, Sheba Medical Center, 52621, Tel-Hashomer, Israel
| | - Jonathan Leor
- Neufeld and Tamman Cardiovascular Research Institutes, Sheba Medical Center, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.
- Heart Center, Sheba Medical Center, 52621, Tel-Hashomer, Israel.
| |
Collapse
|
15
|
Abstract
Intercellular communication by Wnt proteins governs many essential processes during development, tissue homeostasis and disease in all metazoans. Many context-dependent effects are initiated in the Wnt-producing cells and depend on the export of lipidated Wnt proteins. Although much focus has been on understanding intracellular Wnt signal transduction, the cellular machinery responsible for Wnt secretion became better understood only recently. After lipid modification by the acyl-transferase Porcupine, Wnt proteins bind their dedicated cargo protein Evi/Wntless for transport and secretion. Evi/Wntless and Porcupine are conserved transmembrane proteins, and their 3D structures were recently determined. In this Review, we summarise studies and structural data highlighting how Wnts are transported from the ER to the plasma membrane, and the role of SNX3-retromer during the recycling of its cargo receptor Evi/Wntless. We also describe the regulation of Wnt export through a post-translational mechanism and review the importance of Wnt secretion for organ development and cancer, and as a future biomarker.
Collapse
Affiliation(s)
- Lucie Wolf
- German Cancer Research Center (DKFZ), Division of Signalling and Functional Genomics and Heidelberg University, BioQuant and Department of Cell and Molecular Biology, 69120 Heidelberg, Germany
| | - Michael Boutros
- German Cancer Research Center (DKFZ), Division of Signalling and Functional Genomics and Heidelberg University, BioQuant and Department of Cell and Molecular Biology, 69120 Heidelberg, Germany
| |
Collapse
|
16
|
Saeedi-Boroujeni A, Purrahman D, Shojaeian A, Poniatowski ŁA, Rafiee F, Mahmoudian-Sani MR. Progranulin (PGRN) as a regulator of inflammation and a critical factor in the immunopathogenesis of cardiovascular diseases. J Inflamm (Lond) 2023; 20:1. [PMID: 36658641 PMCID: PMC9851114 DOI: 10.1186/s12950-023-00327-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 01/15/2023] [Indexed: 01/20/2023] Open
Abstract
Immune dysregulation has been identified as a critical cause of the most common types of cardiovascular diseases (CVDs). Notably, the innate and adaptive immune responses under physiological conditions are typically regulated with high sensitivity to avoid the exacerbation of inflammation, but any dysregulation can probably be associated with CVDs. In this respect, progranulin (PGRN) serves as one of the main components of the regulation of inflammatory processes, which significantly contributes to the immunopathogenesis of such disorders. PGRN has been introduced among the secreted growth factors as one related to wound healing, inflammation, and human embryonic development, as well as a wide variety of autoimmune diseases. The relationship between the serum PGRN and TNF-α ratio with the spontaneous bacterial peritonitis constitute one of the independent predictors of these conditions. The full-length PGRN can thus effectively reduce the calcification of valve interstitial cells, and the granulin precursor (GRN), among the degradation products of PGRN, can be beneficial. Moreover, it was observed that, PGRN protects the heart against ischemia-reperfusion injury. Above all, PGRN also provides protection in the initial phase following myocardial ischemia-reperfusion injury. The protective impact of PGRN on this may be associated with the early activation of the PI3K/Akt signaling pathway. PGRN also acts as a protective factor in hyperhomocysteinemia, probably by down-regulating the wingless-related integration site Wnt/β-catenin signaling pathway. Many studies have further demonstrated that SARS-CoV-2 (COVID-19) has dramatically increased the risks of CVDs due to inflammation, so PGRN has drawn much more attention among scholars. Lysosomes play a pivotal role in the inflammation process, and PGRN is one of the key regulators in their functioning, which contributes to the immunomodulatory mechanism in the pathogenesis of CVDs. Therefore, investigation of PGRN actions can help find new prospects in the treatment of CVDs. This review aims to summarize the role of PGRN in the immunopathogenesis of CVD, with an emphasis on its treatment.
Collapse
Affiliation(s)
- Ali Saeedi-Boroujeni
- Department of Microbiology, School of Medicine, Abadan University of Medical Sciences, Abadan, Iran
| | - Daryush Purrahman
- grid.411230.50000 0000 9296 6873Thalassemia and Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Ali Shojaeian
- grid.411950.80000 0004 0611 9280Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Łukasz A. Poniatowski
- grid.491786.50000 0001 0211 9062Department of Neurosurgery, Dietrich-Bonhoeffer-Klinikum, Neubrandenburg, Germany
| | - Fatemeh Rafiee
- grid.469309.10000 0004 0612 8427Zanjan Metabolic Diseases Research Center, Zanjan University of Medical Science, Zanjan, Iran
| | - Mohammad-Reza Mahmoudian-Sani
- grid.411230.50000 0000 9296 6873Thalassemia and Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran ,grid.411230.50000 0000 9296 6873Clinical Research Development Unit, Golestan Hospital, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
17
|
Tian CX, Li MY, Shuai XX, Jiang F, Dong YL, Gui Y, Zhang ZL, Qin RJ, Kang ZY, Lin L, Sarapultsev A, Wu B, Luo SS, Hu DS. Berberine plays a cardioprotective role by inhibiting macrophage Wnt5a/β-catenin pathway in the myocardium of mice after myocardial infarction. Phytother Res 2023; 37:50-61. [PMID: 36218220 PMCID: PMC10092487 DOI: 10.1002/ptr.7592] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 07/16/2022] [Accepted: 08/07/2022] [Indexed: 01/19/2023]
Abstract
Myocardial infarction (MI) is one of the diseases with high fatality rate. Berberine (BBR) is a monomer compound with various biological functions. And some studies have confirmed that BBR plays an important role in alleviating cardiomyocyte injury after MI. However, the specific mechanism is unclear. In this study, we induced a model of MI by ligation of the left anterior descending coronary artery and we surprisingly found that BBR significantly improved ventricular remodeling, with a minor inflammatory and oxidative stress injury, and stronger angiogenesis. Moreover, BBR inhibited the secretion of Wnt5a/β-catenin pathway in macrophages after MI, thus promoting the differentiation of macrophages into M2 type. In summary, BBR effectively improved cardiac function of mice after MI, and the potential protective mechanism was associated with the regulation of inflammatory responses and the inhibition of macrophage Wnt5a/β-catenin pathway in the infarcted heart tissues. Importantly, these findings supported BBR as an effective cardioprotective drug after MI.
Collapse
Affiliation(s)
- Chun-Xia Tian
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Cardiology, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, Hubei, China
| | - Ming-Yue Li
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Gastroenterology, Zhongda Hospital, Southeast University, Nanjing, China
| | - Xin-Xin Shuai
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Feng Jiang
- Department of International Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Ya-Lan Dong
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yang Gui
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zi-Li Zhang
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ren-Jie Qin
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhen-Yu Kang
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lan Lin
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Alexey Sarapultsev
- Institute of Immunology and Physiology, Ural Branch of the Russian Academy of Science, Ekaterinburg, Russia
| | - Bin Wu
- Department of Cardiology, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, Hubei, China
| | - Shan-Shan Luo
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - De-Sheng Hu
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Biological Targeted Therapy, The Ministry of Education, Wuhan, China
| |
Collapse
|
18
|
Macrophages and Wnts in Tissue Injury and Repair. Cells 2022; 11:cells11223592. [PMID: 36429021 PMCID: PMC9688352 DOI: 10.3390/cells11223592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 11/07/2022] [Accepted: 11/07/2022] [Indexed: 11/16/2022] Open
Abstract
Macrophages are important players in the immune system that sense various tissue challenges and trigger inflammation. Tissue injuries are followed by inflammation, which is tightly coordinated with tissue repair processes. Dysregulation of these processes leads to chronic inflammation or tissue fibrosis. Wnt ligands are present both in homeostatic and pathological conditions. However, their roles and mechanisms regulating inflammation and tissue repair are being investigated. Here we aim to provide an overview of overarching themes regarding Wnt and macrophages by reviewing the previous literature. We aim to gain future insights into how tissue inflammation, repair, regeneration, and fibrosis events are regulated by macrophages.
Collapse
|
19
|
Li Z, Liu X, Zhang X, Zhang W, Gong M, Qin X, Luo J, Fang Y, Liu B, Wei Y. TRIM21 aggravates cardiac injury after myocardial infarction by promoting M1 macrophage polarization. Front Immunol 2022; 13:1053171. [PMID: 36439111 PMCID: PMC9684192 DOI: 10.3389/fimmu.2022.1053171] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Accepted: 10/25/2022] [Indexed: 08/27/2024] Open
Abstract
Macrophage polarization followed by myocardial infarction (MI) is essential for wound healing. Tripartite motif-containing protein 21 (TRIM21), a member of E3 ubiquitin ligases, is emerging as a mediator in cardiac injury and heart failure. However, its function in modulating post-MI macrophage polarization remains elusive. Here, we detected that the levels of TRIM21 significantly increased in macrophages of wild-type (WT) mice after MI. In contrast, MI was ameliorated in TRIM21 knockout (TRIM21-/-) mice with improved cardiac remodeling, characterized by a marked decrease in mortality, decreased infarct size, and improved cardiac function compared with WT-MI mice. Notably, TRIM21 deficiency impeded the post-MI apoptosis and DNA damage in the hearts of mice. Consistently, the accumulation of M1 phenotype macrophages in the infarcted tissues was significantly reduced with TRIM21 deletion. Mechanistically, the deletion of TRIM21 orchestrated the process of M1 macrophage polarization at least partly via a PI3K/Akt signaling pathway. Overall, we identify TRIM21 drives the inflammatory response and cardiac remodeling by stimulating M1 macrophage polarization through a PI3K/Akt signaling pathway post-MI.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Baoxin Liu
- Department of Cardiology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yidong Wei
- Department of Cardiology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
20
|
Meyer IS, Li X, Meyer C, Voloshanenko O, Pohl S, Boutros M, Katus HA, Frey N, Leuschner F. Blockade of Wnt Secretion Attenuates Myocardial Ischemia-Reperfusion Injury by Modulating the Inflammatory Response. Int J Mol Sci 2022; 23:ijms232012252. [PMID: 36293109 PMCID: PMC9602582 DOI: 10.3390/ijms232012252] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/06/2022] [Accepted: 10/07/2022] [Indexed: 11/16/2022] Open
Abstract
Wnt (a portmanteau of Wingless and Int-1) signaling in the adult heart is largely quiescent. However, there is accumulating evidence that it gets reactivated during the healing process after myocardial infarction (MI). We here tested the therapeutic potential of the Wnt secretion inhibitor LGK-974 on MI healing. Ischemia/reperfusion (I/R) injury was induced in mice and Wnt signaling was inhibited by oral administration of the porcupine inhibitor LGK-974. The transcriptome was analyzed from infarcted tissue by using RNA sequencing analysis. The inflammatory response after I/R was evaluated by flow cytometry. Heart function was assessed by echocardiography and fibrosis by Masson's trichrome staining. Transcriptome and gene set enrichment analysis revealed a modulation of the inflammatory response upon administration of the Wnt secretion inhibitor LGK-974 following I/R. In addition, LGK-974-treated animals showed an attenuated inflammatory response and improved heart function. In an in vitro model of hypoxic cardiomyocyte and monocyte/macrophage interaction, LGK974 inhibited the activation of Wnt signaling in monocytes/macrophages and reduced their pro-inflammatory phenotype. We here show that Wnt signaling affects inflammatory processes after MI. The Wnt secretion inhibitor LGK-974 appears to be a promising compound for future immunomodulatory approaches to improve cardiac remodeling after MI.
Collapse
Affiliation(s)
- Ingmar Sören Meyer
- Internal Medicine III, University Hospital Heidelberg, 69120 Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg-Mannheim, 69120 Heidelberg, Germany
| | - Xue Li
- Internal Medicine III, University Hospital Heidelberg, 69120 Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg-Mannheim, 69120 Heidelberg, Germany
| | - Carina Meyer
- Internal Medicine III, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | | | - Susann Pohl
- Internal Medicine III, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Michael Boutros
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Hugo Albert Katus
- Internal Medicine III, University Hospital Heidelberg, 69120 Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg-Mannheim, 69120 Heidelberg, Germany
| | - Norbert Frey
- Internal Medicine III, University Hospital Heidelberg, 69120 Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg-Mannheim, 69120 Heidelberg, Germany
| | - Florian Leuschner
- Internal Medicine III, University Hospital Heidelberg, 69120 Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg-Mannheim, 69120 Heidelberg, Germany
- Correspondence:
| |
Collapse
|
21
|
Abstract
Understanding how macrophages promote myocardial repair can help create new therapies for infarct repair. We aimed to determine what mechanisms underlie the reparative properties of macrophages. Cytokine arrays revealed that neonatal cardiac macrophages from the injured neonatal heart secreted high amounts of osteopontin (OPN). In vitro, recombinant OPN stimulated cardiac cell outgrowth, cardiomyocyte (CM) cell-cycle re-entry, and CM migration. In addition, OPN induced nuclear translocation of the cytoplasmatic yes-associated protein 1 (YAP1) and upregulated transcriptional factors and cell-cycle genes. Significantly, by blocking the OPN receptor CD44, we eliminated the effects of OPN on CMs. OPN also activated the proliferation and migration of non-CM cells: endothelial cells and cardiac mesenchymal stromal cells in vitro. Notably, the significant role of OPN in myocardial healing was demonstrated by impaired healing in OPN-deficient neonatal hearts. Finally, in the adult mice, a single injection of OPN into the border of the ischemic zone induced CM cell-cycle re-entry, improved scar formation, local and global cardiac function, and LV remodelling 30 days after MI. In summary, we have shown, for the first time, that recombinant OPN activates cell-cycle re-entry in CMs. In addition, recombinant OPN stimulates multiple cardiac cells and improves scar formation, LV remodelling, and regional and global function after MI. Therefore, we propose OPN as a new cell-free therapy to optimize infarct repair.
Collapse
|
22
|
Wnt is coming: A role for serum Wnt2 and 4 in fibrotic remodeling following acute myocardial infarction. EBioMedicine 2022; 77:103877. [PMID: 35168106 PMCID: PMC8850681 DOI: 10.1016/j.ebiom.2022.103877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 01/25/2022] [Indexed: 12/01/2022] Open
|
23
|
Picchio V, Bordin A, Floris E, Cozzolino C, Dhori X, Peruzzi M, Frati G, De Falco E, Pagano F, Chimenti I. The dynamic facets of the cardiac stroma: from classical markers to omics and translational perspectives. Am J Transl Res 2022; 14:1172-1187. [PMID: 35273721 PMCID: PMC8902528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 11/29/2021] [Indexed: 06/14/2023]
Abstract
Cardiac stromal cells have been long underestimated in their functions in homeostasis and repair. Recent evidence has changed this perspective in that many more players and facets than just "cardiac fibroblasts" have entered the field. Single cell transcriptomic studies on cardiac interstitial cells have shed light on the phenotypic plasticity of the stroma, whose transcriptional profile is dynamically regulated in homeostatic conditions and in response to external stimuli. Different populations and/or functional states that appear in homeostasis and pathology have been described, particularly increasing the complexity of studying the cardiac response to injury. In this review, we outline current phenotypical and molecular markers, and the approaches developed for identifying and classifying cardiac stromal cells. Significant advances in our understanding of cardiac stromal populations will provide a deeper knowledge on myocardial functional cellular components, as well as a platform for future developments of novel therapeutic strategies to counteract cardiac fibrosis and adverse cardiac remodeling.
Collapse
Affiliation(s)
- Vittorio Picchio
- Department of Medical Surgical Sciences and Biotechnologies, Sapienza University of RomeItaly
| | - Antonella Bordin
- Department of Medical Surgical Sciences and Biotechnologies, Sapienza University of RomeItaly
| | - Erica Floris
- Department of Medical Surgical Sciences and Biotechnologies, Sapienza University of RomeItaly
| | - Claudia Cozzolino
- Department of Medical Surgical Sciences and Biotechnologies, Sapienza University of RomeItaly
| | - Xhulio Dhori
- Department of Medical Surgical Sciences and Biotechnologies, Sapienza University of RomeItaly
| | - Mariangela Peruzzi
- Mediterranea CardiocentroNapoli, Italy
- Department of Clinical, Internal Medicine, Anaesthesiology and Cardiovascular Sciences, Sapienza University of RomeItaly
| | - Giacomo Frati
- Department of Medical Surgical Sciences and Biotechnologies, Sapienza University of RomeItaly
- IRCCS NeuromedPozzilli, Italy
| | - Elena De Falco
- Department of Medical Surgical Sciences and Biotechnologies, Sapienza University of RomeItaly
- Mediterranea CardiocentroNapoli, Italy
| | - Francesca Pagano
- Biochemistry and Cellular Biology Institute, CNRMonterotondo, Italy
| | - Isotta Chimenti
- Department of Medical Surgical Sciences and Biotechnologies, Sapienza University of RomeItaly
- Mediterranea CardiocentroNapoli, Italy
| |
Collapse
|
24
|
Schoger E, Lelek S, Panáková D, Zelarayán LC. Tailoring Cardiac Synthetic Transcriptional Modulation Towards Precision Medicine. Front Cardiovasc Med 2022; 8:783072. [PMID: 35097003 PMCID: PMC8795974 DOI: 10.3389/fcvm.2021.783072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Accepted: 12/07/2021] [Indexed: 11/13/2022] Open
Abstract
Molecular and genetic differences between individual cells within tissues underlie cellular heterogeneities defining organ physiology and function in homeostasis as well as in disease states. Transcriptional control of endogenous gene expression has been intensively studied for decades. Thanks to a fast-developing field of single cell genomics, we are facing an unprecedented leap in information available pertaining organ biology offering a comprehensive overview. The single-cell technologies that arose aided in resolving the precise cellular composition of many organ systems in the past years. Importantly, when applied to diseased tissues, the novel approaches have been immensely improving our understanding of the underlying pathophysiology of common human diseases. With this information, precise prediction of regulatory elements controlling gene expression upon perturbations in a given cell type or a specific context will be realistic. Simultaneously, the technological advances in CRISPR-mediated regulation of gene transcription as well as their application in the context of epigenome modulation, have opened up novel avenues for targeted therapy and personalized medicine. Here, we discuss the fast-paced advancements during the recent years and the applications thereof in the context of cardiac biology and common cardiac disease. The combination of single cell technologies and the deep knowledge of fundamental biology of the diseased heart together with the CRISPR-mediated modulation of gene regulatory networks will be instrumental in tailoring the right strategies for personalized and precision medicine in the near future. In this review, we provide a brief overview of how single cell transcriptomics has advanced our knowledge and paved the way for emerging CRISPR/Cas9-technologies in clinical applications in cardiac biomedicine.
Collapse
Affiliation(s)
- Eric Schoger
- Institute of Pharmacology and Toxicology, University Medical Center Goettingen, Goettingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Goettingen, Goettingen, Germany
- Cluster of Excellence “Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells”, University of Goettingen, Goettingen, Germany
| | - Sara Lelek
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Daniela Panáková
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Daniela Panáková
| | - Laura Cecilia Zelarayán
- Institute of Pharmacology and Toxicology, University Medical Center Goettingen, Goettingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Goettingen, Goettingen, Germany
- Cluster of Excellence “Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells”, University of Goettingen, Goettingen, Germany
- *Correspondence: Laura Cecilia Zelarayán
| |
Collapse
|
25
|
Liu J, Zheng X, Zhang C, Zhang C, Bu P. Lcz696 Alleviates Myocardial Fibrosis After Myocardial Infarction Through the sFRP-1/Wnt/β-Catenin Signaling Pathway. Front Pharmacol 2021; 12:724147. [PMID: 34539406 PMCID: PMC8443774 DOI: 10.3389/fphar.2021.724147] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Accepted: 08/20/2021] [Indexed: 12/11/2022] Open
Abstract
Background: Lcz696 (ARNI, angiotensin receptor–neprilysin inhibitor; sacubitril/valsartan) shows an inhibitory effect on fibrosis after myocardial infarction (MI). However, the underlying signaling mechanisms are poorly understood. The Wnt/β-catenin signaling pathway is activated after MI and participates in the process of myocardial fibrosis. Here, we aimed to assess the efficacy of ARNI for alleviating myocardial fibrosis after MI and hypothesized that ARNI alleviates myocardial fibrosis by inhibiting the Wnt/β-catenin signaling pathway and overexpressing sFRP-1, an inhibitor of the Wnt/β-catenin signaling pathway. Methods: Mice randomized at 1 week post-MI were administered lcz696 (60 mg/kg, n = 21), valsartan (30 mg/kg, n = 19), or corn oil (n = 13) orally for 4 weeks, while the sham-operated group received vehicle (corn oil, n = 19). Cardiac function and extent of myocardial fibrosis were measured. Western blotting and quantitative real-time polymerase chain reaction were used to detect the expression of Wnt/β-catenin pathway-related proteins. Furthermore, primary myocardial fibroblasts were stimulated with angiotensin II (Ang II) and cultured with lcz696 and the sFRP-1 inhibitor way316606 to detect the expression of Wnt/β-catenin pathway proteins. Results: Both lcz696 and valsartan alleviated myocardial fibrosis and improved cardiac function, but lcz696 had superior efficiency compared to valsartan. Furthermore, β-catenin expression was inhibited and sFRP-1 was overexpressed after drug treatment, which could be significantly improved by lcz696 in mice. In addition, lcz696 inhibited β-catenin expression in AngII-stimulated myocardial fibroblasts, and β-catenin expression increased after the inhibition of sFRP-1. Conclusion: ARNI alleviated cardiac fibrosis and cardiac remodeling by inhibiting the Wnt/β-catenin signaling pathway. In addition, ARNI can lead to overexpression of sFRP-1, which is an inhibitor of the Wnt/β-catenin signaling pathway. These results indicate a new therapeutic target of ARNI to improve myocardial fibrosis and prevent myocardial remodeling.
Collapse
Affiliation(s)
- Jing Liu
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Cardiology, Heze Municipal Hospital, Heze, China
| | - Xuehui Zheng
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Chen Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Chunmei Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Peili Bu
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
26
|
Wang X, Chen J, Xu J, Xie J, Harris DCH, Zheng G. The Role of Macrophages in Kidney Fibrosis. Front Physiol 2021; 12:705838. [PMID: 34421643 PMCID: PMC8378534 DOI: 10.3389/fphys.2021.705838] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 06/07/2021] [Indexed: 12/27/2022] Open
Abstract
The phenotypic heterogeneity and functional diversity of macrophages confer on them complexed roles in the development and progression of kidney diseases. After kidney injury, bone marrow-derived monocytes are rapidly recruited to the glomerulus and tubulointerstitium. They are activated and differentiated on site into pro-inflammatory M1 macrophages, which initiate Th1-type adaptive immune responses and damage normal tissues. In contrast, anti-inflammatory M2 macrophages induce Th2-type immune responses, secrete large amounts of TGF-β and anti-inflammatory cytokines, transform into αSMA+ myofibroblasts in injured kidney, inhibit immune responses, and promote wound healing and tissue fibrosis. Previous studies on the role of macrophages in kidney fibrosis were mainly focused on inflammation-associated injury and injury repair. Apart from macrophage-secreted profibrotic cytokines, such as TGF-β, evidence for a direct contribution of macrophages to kidney fibrosis is lacking. However, under inflammatory conditions, Wnt ligands are derived mainly from macrophages and Wnt signaling is central in the network of multiple profibrotic pathways. Largely underinvestigated are the direct contribution of macrophages to profibrotic signaling pathways, macrophage phenotypic heterogeneity and functional diversity in relation to kidney fibrosis, and on their cross-talk with other cells in profibrotic signaling networks that cause fibrosis. Here we aim to provide an overview on the roles of macrophage phenotypic and functional diversity in their contribution to pro-fibrotic signaling pathways, and on the therapeutic potential of targeting macrophages for the treatment of kidney fibrosis.
Collapse
Affiliation(s)
- Xiaoling Wang
- Shanxi Key Laboratory of Birth Defect and Cell Regeneration, Shanxi Medical University, Taiyuan, China
- Clinical Laboratory, Shanxi Academy of Traditional Chinese Medicine, Taiyuan, China
| | - Jianwei Chen
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, NSW, Australia
| | - Jun Xu
- Department of General Surgery, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Jun Xie
- Shanxi Key Laboratory of Birth Defect and Cell Regeneration, Shanxi Medical University, Taiyuan, China
| | - David C. H. Harris
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, NSW, Australia
| | - Guoping Zheng
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
27
|
Siman-Tov R, Zelikson N, Caspi M, Levi Y, Perry C, Khair F, Stauber H, Sznitman J, Rosin-Arbesfeld R. Circulating Wnt Ligands Activate the Wnt Signaling Pathway in Mature Erythrocytes. Arterioscler Thromb Vasc Biol 2021; 41:e243-e264. [PMID: 33626913 DOI: 10.1161/atvbaha.120.315413] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Ronen Siman-Tov
- Department of Clinical Microbiology and Immunology, Sackler School of Medicine, Tel Aviv University, Israel (R.S.-T., N.Z., M.C., Y.L., C.P., F.K., R.R.-A.)
| | - Natalie Zelikson
- Department of Clinical Microbiology and Immunology, Sackler School of Medicine, Tel Aviv University, Israel (R.S.-T., N.Z., M.C., Y.L., C.P., F.K., R.R.-A.)
| | - Michal Caspi
- Department of Clinical Microbiology and Immunology, Sackler School of Medicine, Tel Aviv University, Israel (R.S.-T., N.Z., M.C., Y.L., C.P., F.K., R.R.-A.)
| | - Yakir Levi
- Department of Clinical Microbiology and Immunology, Sackler School of Medicine, Tel Aviv University, Israel (R.S.-T., N.Z., M.C., Y.L., C.P., F.K., R.R.-A.)
| | - Chava Perry
- Department of Clinical Microbiology and Immunology, Sackler School of Medicine, Tel Aviv University, Israel (R.S.-T., N.Z., M.C., Y.L., C.P., F.K., R.R.-A.)
- BMT Unit, Institute of Hematology, Tel-Aviv Sourasky Medical Center, Israel (C.P.)
| | - Fayhaa Khair
- Department of Clinical Microbiology and Immunology, Sackler School of Medicine, Tel Aviv University, Israel (R.S.-T., N.Z., M.C., Y.L., C.P., F.K., R.R.-A.)
| | - Hagit Stauber
- Department of Biomedical Engineering, Technion-Israel Institute of Technology, Haifa (H.S., J.S.)
| | - Josué Sznitman
- Department of Biomedical Engineering, Technion-Israel Institute of Technology, Haifa (H.S., J.S.)
| | - Rina Rosin-Arbesfeld
- Department of Clinical Microbiology and Immunology, Sackler School of Medicine, Tel Aviv University, Israel (R.S.-T., N.Z., M.C., Y.L., C.P., F.K., R.R.-A.)
| |
Collapse
|
28
|
Shaihov-Teper O, Ram E, Ballan N, Brzezinski RY, Naftali-Shani N, Masoud R, Ziv T, Lewis N, Schary Y, Levin-Kotler LP, Volvovitch D, Zuroff EM, Amunts S, Regev-Rudzki N, Sternik L, Raanani E, Gepstein L, Leor J. Extracellular Vesicles From Epicardial Fat Facilitate Atrial Fibrillation. Circulation 2021; 143:2475-2493. [PMID: 33793321 DOI: 10.1161/circulationaha.120.052009] [Citation(s) in RCA: 124] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND The role of epicardial fat (eFat)-derived extracellular vesicles (EVs) in the pathogenesis of atrial fibrillation (AF) has never been studied. We tested the hypothesis that eFat-EVs transmit proinflammatory, profibrotic, and proarrhythmic molecules that induce atrial myopathy and fibrillation. METHODS We collected eFat specimens from patients with (n=32) and without AF (n=30) during elective heart surgery. eFat samples were grown as organ cultures, and the culture medium was collected every 2 days. We then isolated and purified eFat-EVs from the culture medium, and analyzed the EV number, size, morphology, specific markers, encapsulated cytokines, proteome, and microRNAs. Next, we evaluated the biological effects of unpurified and purified EVs on atrial mesenchymal stromal cells and endothelial cells in vitro. To establish a causal association between eFat-EVs and vulnerability to AF, we modeled AF in vitro using induced pluripotent stem cell-derived cardiomyocytes. RESULTS Microscopic examination revealed excessive inflammation, fibrosis, and apoptosis in fresh and cultured eFat tissues. Cultured explants from patients with AF secreted more EVs and harbored greater amounts of proinflammatory and profibrotic cytokines, and profibrotic microRNA, as well, than those without AF. The proteomic analysis confirmed the distinctive profile of purified eFat-EVs from patients with AF. In vitro, purified and unpurified eFat-EVs from patients with AF had a greater effect on proliferation and migration of human mesenchymal stromal cells and endothelial cells, compared with eFat-EVs from patients without AF. Last, whereas eFat-EVs from patients with and without AF shortened the action potential duration of induced pluripotent stem cell-derived cardiomyocytes, only eFat-EVs from patients with AF induced sustained reentry (rotor) in induced pluripotent stem cell-derived cardiomyocytes. CONCLUSIONS We show, for the first time, a distinctive proinflammatory, profibrotic, and proarrhythmic signature of eFat-EVs from patients with AF. Our findings uncover another pathway by which eFat promotes the development of atrial myopathy and fibrillation.
Collapse
Affiliation(s)
- Olga Shaihov-Teper
- Neufeld and Tamman Cardiovascular Research Institutes (O.S.-T., R.Y.B., N.N.-S., N.L., Y.S., L.-P.L.-K., J.L.), Sheba Medical Center, Sackler School of Medicine, Tel Aviv University, Israel.,Heart Center, Sheba Medical Center, Tel Hashomer, Israel (O.S.-T., E. Ram, R.Y.B., N.N.-S., N.L., Y.S., L.-P.L.-K., D.V., E.M.Z., S.A., L.S., E. Raanani, J.L.)
| | - Eilon Ram
- Department of Cardiac Surgery, Leviev Cardiothoracic and Vascular Center (E. Ram, E.M.Z., S.A., L.S., E. Raanani), Sheba Medical Center, Sackler School of Medicine, Tel Aviv University, Israel.,Heart Center, Sheba Medical Center, Tel Hashomer, Israel (O.S.-T., E. Ram, R.Y.B., N.N.-S., N.L., Y.S., L.-P.L.-K., D.V., E.M.Z., S.A., L.S., E. Raanani, J.L.)
| | - Nimer Ballan
- The Sohnis Family Laboratory for Cardiac Electrophysiology and Regenerative Medicine, Rappaport Faculty of Medicine, Technion Institute of Technology, Israel (N.B., L.G.)
| | - Rafael Y Brzezinski
- Neufeld and Tamman Cardiovascular Research Institutes (O.S.-T., R.Y.B., N.N.-S., N.L., Y.S., L.-P.L.-K., J.L.), Sheba Medical Center, Sackler School of Medicine, Tel Aviv University, Israel.,Heart Center, Sheba Medical Center, Tel Hashomer, Israel (O.S.-T., E. Ram, R.Y.B., N.N.-S., N.L., Y.S., L.-P.L.-K., D.V., E.M.Z., S.A., L.S., E. Raanani, J.L.)
| | - Nili Naftali-Shani
- Neufeld and Tamman Cardiovascular Research Institutes (O.S.-T., R.Y.B., N.N.-S., N.L., Y.S., L.-P.L.-K., J.L.), Sheba Medical Center, Sackler School of Medicine, Tel Aviv University, Israel.,Heart Center, Sheba Medical Center, Tel Hashomer, Israel (O.S.-T., E. Ram, R.Y.B., N.N.-S., N.L., Y.S., L.-P.L.-K., D.V., E.M.Z., S.A., L.S., E. Raanani, J.L.)
| | - Rula Masoud
- Cancer Research Center, Chaim Sheba Medical Center, Tel Hashomer, Israel (R.M.)
| | - Tamar Ziv
- Smoler Proteomics Center, Faculty of Biology, Technion-Israel Institute of Technology, Haifa, Israel (T.Z.)
| | - Nir Lewis
- Neufeld and Tamman Cardiovascular Research Institutes (O.S.-T., R.Y.B., N.N.-S., N.L., Y.S., L.-P.L.-K., J.L.), Sheba Medical Center, Sackler School of Medicine, Tel Aviv University, Israel.,Heart Center, Sheba Medical Center, Tel Hashomer, Israel (O.S.-T., E. Ram, R.Y.B., N.N.-S., N.L., Y.S., L.-P.L.-K., D.V., E.M.Z., S.A., L.S., E. Raanani, J.L.)
| | - Yeshai Schary
- Neufeld and Tamman Cardiovascular Research Institutes (O.S.-T., R.Y.B., N.N.-S., N.L., Y.S., L.-P.L.-K., J.L.), Sheba Medical Center, Sackler School of Medicine, Tel Aviv University, Israel.,Heart Center, Sheba Medical Center, Tel Hashomer, Israel (O.S.-T., E. Ram, R.Y.B., N.N.-S., N.L., Y.S., L.-P.L.-K., D.V., E.M.Z., S.A., L.S., E. Raanani, J.L.)
| | - La-Paz Levin-Kotler
- Neufeld and Tamman Cardiovascular Research Institutes (O.S.-T., R.Y.B., N.N.-S., N.L., Y.S., L.-P.L.-K., J.L.), Sheba Medical Center, Sackler School of Medicine, Tel Aviv University, Israel.,Heart Center, Sheba Medical Center, Tel Hashomer, Israel (O.S.-T., E. Ram, R.Y.B., N.N.-S., N.L., Y.S., L.-P.L.-K., D.V., E.M.Z., S.A., L.S., E. Raanani, J.L.)
| | - David Volvovitch
- Heart Center, Sheba Medical Center, Tel Hashomer, Israel (O.S.-T., E. Ram, R.Y.B., N.N.-S., N.L., Y.S., L.-P.L.-K., D.V., E.M.Z., S.A., L.S., E. Raanani, J.L.)
| | - Elchanan M Zuroff
- Department of Cardiac Surgery, Leviev Cardiothoracic and Vascular Center (E. Ram, E.M.Z., S.A., L.S., E. Raanani), Sheba Medical Center, Sackler School of Medicine, Tel Aviv University, Israel.,Heart Center, Sheba Medical Center, Tel Hashomer, Israel (O.S.-T., E. Ram, R.Y.B., N.N.-S., N.L., Y.S., L.-P.L.-K., D.V., E.M.Z., S.A., L.S., E. Raanani, J.L.)
| | - Sergei Amunts
- Department of Cardiac Surgery, Leviev Cardiothoracic and Vascular Center (E. Ram, E.M.Z., S.A., L.S., E. Raanani), Sheba Medical Center, Sackler School of Medicine, Tel Aviv University, Israel.,Heart Center, Sheba Medical Center, Tel Hashomer, Israel (O.S.-T., E. Ram, R.Y.B., N.N.-S., N.L., Y.S., L.-P.L.-K., D.V., E.M.Z., S.A., L.S., E. Raanani, J.L.)
| | - Neta Regev-Rudzki
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel (N.R.-R.)
| | - Leonid Sternik
- Department of Cardiac Surgery, Leviev Cardiothoracic and Vascular Center (E. Ram, E.M.Z., S.A., L.S., E. Raanani), Sheba Medical Center, Sackler School of Medicine, Tel Aviv University, Israel.,Heart Center, Sheba Medical Center, Tel Hashomer, Israel (O.S.-T., E. Ram, R.Y.B., N.N.-S., N.L., Y.S., L.-P.L.-K., D.V., E.M.Z., S.A., L.S., E. Raanani, J.L.)
| | - Ehud Raanani
- Department of Cardiac Surgery, Leviev Cardiothoracic and Vascular Center (E. Ram, E.M.Z., S.A., L.S., E. Raanani), Sheba Medical Center, Sackler School of Medicine, Tel Aviv University, Israel.,Heart Center, Sheba Medical Center, Tel Hashomer, Israel (O.S.-T., E. Ram, R.Y.B., N.N.-S., N.L., Y.S., L.-P.L.-K., D.V., E.M.Z., S.A., L.S., E. Raanani, J.L.)
| | - Lior Gepstein
- The Sohnis Family Laboratory for Cardiac Electrophysiology and Regenerative Medicine, Rappaport Faculty of Medicine, Technion Institute of Technology, Israel (N.B., L.G.)
| | - Jonathan Leor
- Neufeld and Tamman Cardiovascular Research Institutes (O.S.-T., R.Y.B., N.N.-S., N.L., Y.S., L.-P.L.-K., J.L.), Sheba Medical Center, Sackler School of Medicine, Tel Aviv University, Israel.,Heart Center, Sheba Medical Center, Tel Hashomer, Israel (O.S.-T., E. Ram, R.Y.B., N.N.-S., N.L., Y.S., L.-P.L.-K., D.V., E.M.Z., S.A., L.S., E. Raanani, J.L.)
| |
Collapse
|
29
|
Translational insights from single-cell technologies across the cardiovascular disease continuum. Trends Cardiovasc Med 2021; 32:127-135. [PMID: 33667644 DOI: 10.1016/j.tcm.2021.02.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 02/22/2021] [Accepted: 02/23/2021] [Indexed: 12/12/2022]
Abstract
Cardiovascular disease is the leading cause of death worldwide. The societal health burden it represents can be reduced by taking preventive measures and developing more effective therapies. Reaching these goals, however, requires a better understanding of the pathophysiological processes leading to and occurring in the diseased heart. In the last 5 years, several biological advances applying single-cell technologies have enabled researchers to study cardiovascular diseases with unprecedented resolution. This has produced many new insights into how specific cell types change their gene expression level, activation status and potential cellular interactions with the development of cardiovascular disease, but a comprehensive overview of the clinical implications of these findings is lacking. In this review, we summarize and discuss these recent advances and the promise of single-cell technologies from a translational perspective across the cardiovascular disease continuum, covering both animal and human studies, and explore the future directions of the field.
Collapse
|
30
|
Li L, Qiu X, Zhang N, Sun Y, Wang Y, Wang L. Crosstalk between adipocytes and M2 macrophages compensates for osteopenic phenotype in the Lrp5-deficient mice. Exp Biol Med (Maywood) 2021; 246:572-583. [PMID: 33197324 PMCID: PMC7934148 DOI: 10.1177/1535370220972320] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 10/19/2020] [Indexed: 02/06/2023] Open
Abstract
A loss-of-function mutation in the Lrp5 gene in mice leads to a low bone mass disorder due to the inhibition of the canonical Wnt signaling pathway; however, the role of bone marrow microenvironment in mice with this mutation remains unclear. In this study, we evaluated proliferation and osteogenic potential of mouse osteoblasts using the MTT assay and Alizarin red staining. The levels of alkaline phosphatase, tartrate-resistant acid phosphatase, and adiponectin in culture supernatants were measured using the enzyme-linked immunosorbent assay. Osteoclast bone resorbing activity was evaluated by toluidine staining and the number and area of bone resorption pits were determined. We observed increased osteogenesis in osteoblasts co-cultured with the BM-derived myeloid cells compared to the osteoblasts cultured alone. Mice with global Lrp5 deletion had a relatively higher bone density compared to the mice carrying osteoblast/osteocyte-specific Lrp5 deletion. An increased frequency of M2 macrophages and reduced expression of inflammatory cytokines were detected in the myeloid cells derived from the bone marrow of mice with global Lrp5 deletion. Higher adipogenic potential and elevated levels of adiponectin in the global Lrp5 deletion mice contributed to the preferential M2 macrophage polarization. Here, we identified a novel systemic regulatory mechanism of bone formation and degradation in mice with global Lrp5 deletion. This mechanism depends on a crosstalk between the adipocytes and M2 macrophages in the bone marrow and is responsible for partly rescuing osteopenia developed as a result of decreased Wnt signaling.
Collapse
Affiliation(s)
- Lisha Li
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, China
- The Academy of Integrative Medicine of Fudan University, Shanghai 200011, China
- Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Shanghai 200011, China
| | - Xuemin Qiu
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, China
- The Academy of Integrative Medicine of Fudan University, Shanghai 200011, China
- Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Shanghai 200011, China
| | - Na Zhang
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, China
- The Academy of Integrative Medicine of Fudan University, Shanghai 200011, China
- Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Shanghai 200011, China
| | - Yan Sun
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, China
- The Academy of Integrative Medicine of Fudan University, Shanghai 200011, China
- Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Shanghai 200011, China
| | - Yan Wang
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, China
| | - Ling Wang
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, China
- The Academy of Integrative Medicine of Fudan University, Shanghai 200011, China
- Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Shanghai 200011, China
| |
Collapse
|
31
|
Hsueh YC, Hodgkinson CP, Gomez JA. The role of Sfrp and DKK proteins in cardiomyocyte development. Physiol Rep 2021; 9:e14678. [PMID: 33587322 PMCID: PMC7883806 DOI: 10.14814/phy2.14678] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 11/11/2020] [Accepted: 11/18/2020] [Indexed: 12/12/2022] Open
Abstract
In this review, we summarize the role of Wnt proteins in cardiomyogenesis. More specifically, we focus on how the development of cardiomyocytes from precursor cells involves a complex interplay between Wnt canonical β-catenin signaling pathways and Wnt noncanonical signaling pathways involving PCP and JNK. We also describe recent literature which suggests that endogenous Wnt inhibitors such as the Sfrp and DKK proteins play important roles in regulating the cardiomyocyte differentiation.
Collapse
Affiliation(s)
- Ying-Chang Hsueh
- Mandel Center for Heart and Vascular Research, and the Duke Cardiovascular Research Center, Duke University Medical Center, Durham, NC, USA
| | - Conrad P Hodgkinson
- Mandel Center for Heart and Vascular Research, and the Duke Cardiovascular Research Center, Duke University Medical Center, Durham, NC, USA
| | - Jose A Gomez
- Department of Medicine, Clinical Pharmacology Division, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
32
|
Effect of Interventions in WNT Signaling on Healing of Cardiac Injury: A Systematic Review. Cells 2021; 10:cells10020207. [PMID: 33494313 PMCID: PMC7912185 DOI: 10.3390/cells10020207] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 01/08/2021] [Accepted: 01/14/2021] [Indexed: 12/12/2022] Open
Abstract
The wound healing that follows myocardial infarction is a complex process involving multiple mechanisms, such as inflammation, angiogenesis and fibrosis. In the last two decades, the involvement of WNT signaling has been extensively studied and effects on virtually all aspects of this wound healing have been reported. However, as often is the case in a newly emerging field, inconsistent and sometimes even contradictory findings have been reported. The aim of this systematic review is to provide a comprehensive overview of studies in which the effect of interventions in WNT signaling were investigated in in vivo models of cardiac injury. To this end, we used different search engines to perform a systematic search of the literature using the key words "WNT and myocardial and infarction". We categorized the interventions according to their place in the WNT signaling pathway (ligand, receptor, destruction complex or nuclear level). The most consistent improvements of the wound healing response were observed in studies in which the acylation of WNT proteins was inhibited by administering porcupine inhibitors, by inhibiting of the downstream glycogen synthase kinase-3β (GSK3β) and by intervening in the β-catenin-mediated gene transcription. Interestingly, in several of these studies, evidence was presented for activation of cardiomyocyte proliferation around the infarct area. These findings indicate that inhibition of WNT signaling can play a valuable role in the repair of cardiac injury, thereby improving cardiac function and preventing the development of heart failure.
Collapse
|
33
|
Stafford N, Zi M, Baudoin F, Mohamed TMA, Prehar S, De Giorgio D, Cartwright EJ, Latini R, Neyses L, Oceandy D. PMCA4 inhibition does not affect cardiac remodelling following myocardial infarction, but may reduce susceptibility to arrhythmia. Sci Rep 2021; 11:1518. [PMID: 33452399 PMCID: PMC7810749 DOI: 10.1038/s41598-021-81170-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 01/04/2021] [Indexed: 12/03/2022] Open
Abstract
Ischaemic heart disease is the world's leading cause of mortality. Survival rates from acute myocardial infarction (MI) have improved in recent years; however, this has led to an increase in the prevalence of heart failure (HF) due to chronic remodelling of the infarcted myocardium, for which treatment options remain poor. We have previously shown that inhibition of isoform 4 of the plasma membrane calcium ATPase (PMCA4) prevents chronic remodelling and HF development during pressure overload, through fibroblast mediated Wnt signalling modulation. Given that Wnt signalling also plays a prominent role during remodelling of the infarcted heart, this study investigated the effect of genetic and functional loss of PMCA4 on cardiac outcomes following MI. Neither genetic deletion nor pharmacological inhibition of PMCA4 affected chronic remodelling of the post-MI myocardium. This was the case when PMCA4 was deleted globally, or specifically from cardiomyocytes or fibroblasts. PMCA4-ablated hearts were however less prone to acute arrhythmic events, which may offer a slight survival benefit. Overall, this study demonstrates that PMCA4 inhibition does not affect chronic outcomes following MI.
Collapse
Affiliation(s)
- Nicholas Stafford
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| | - Min Zi
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| | - Florence Baudoin
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| | - Tamer M A Mohamed
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
- Department of Medicine, Institute of Molecular Cardiology, University of Louisville, Louisville, KY, USA
- Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Sukhpal Prehar
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| | - Daria De Giorgio
- Department of Cardiovascular Medicine, Mario Negri Institute for Pharmacological Research, Milan, Italy
| | - Elizabeth J Cartwright
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| | - Roberto Latini
- Department of Cardiovascular Medicine, Mario Negri Institute for Pharmacological Research, Milan, Italy
| | - Ludwig Neyses
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
- Simply Uni, Sète, France
| | - Delvac Oceandy
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK.
| |
Collapse
|
34
|
Liu W, Liu Y, Pan Z, Zhang X, Qin Y, Chen X, Li M, Chen X, Zheng Q, Liu X, Li D. Systematic Analysis of tRNA-Derived Small RNAs Discloses New Therapeutic Targets of Caloric Restriction in Myocardial Ischemic Rats. Front Cell Dev Biol 2020; 8:568116. [PMID: 33224944 PMCID: PMC7670042 DOI: 10.3389/fcell.2020.568116] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 10/01/2020] [Indexed: 12/23/2022] Open
Abstract
Caloric restriction (CR) is a novel dietary therapy that has a protective effect on myocardial ischemia. However, the mechanisms underlying the therapeutic effect of CR remain unclear. Transfer RNA-derived small RNAs (tsRNAs) are a novel type of short non-coding RNAs that have potential regulatory functions in various physiological and pathological processes. In this study, we explored new therapeutic targets of CR through tsRNA sequencing. Rats were randomly divided into three groups: a normal control group (norm group), isoproterenol (ISO)-induced myocardial ischemic group (MI group), and CR pretreatment plus ISO-induced myocardial ischemic group (CR + MI group). Triphenyl tetrazolium chloride staining, terminal deoxynucleotidyl transferase dUTP nick-end labeling staining, serum creatine kinase (CK) and lactic acid dehydrogenase activity detection kits, and creatine kinase isoenzyme 1 levels were used to measure the degree of myocardial ischemic injury. These indicators of myocardial ischemia were significantly improved in the CR + MI group compared with those in the MI group. In the ischemic myocardial tissue of the MI group, a total of 708 precisely matched tsRNAs were identified, and 302 tsRNAs (fold change >1.5, P < 0.05) were significantly changed when compared with those in the norm group. Furthermore, 55 tsRNAs were significantly regulated by CR pretreatment, among which five tsRNAs (tiRNA-His-GTG-004, tRF-Gly-TCC-018, tRF-Cys-GCA-022, tRF-Lys-CTT-026, tRF-Met-CAT-008) were randomly selected and verified by quantitative real-time polymerase chain reaction. In addition, predictions of target genes and bioinformatics analysis indicated that these tsRNAs may play a therapeutic role through the regulation of macromolecular metabolism. In conclusion, our findings reveal that tsRNAs are potential therapeutic targets for CR pre-pretreatment to improve myocardial ischemic injury. This study provides new ideas for future research on elucidating the mechanisms of CR pretreatment in ameliorating myocardial ischemic injury.
Collapse
Affiliation(s)
- Wenjing Liu
- Yantai Key Laboratory of Pharmacology of Traditional Chinese Medicine in Tumor Metabolism, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, China
| | - Yang Liu
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Zhaohai Pan
- Yantai Key Laboratory of Pharmacology of Traditional Chinese Medicine in Tumor Metabolism, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, China
| | - Xin Zhang
- Yantai Key Laboratory of Pharmacology of Traditional Chinese Medicine in Tumor Metabolism, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, China
| | - Yao Qin
- Yantai Key Laboratory of Pharmacology of Traditional Chinese Medicine in Tumor Metabolism, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, China
| | - Xiaojie Chen
- Yantai Key Laboratory of Pharmacology of Traditional Chinese Medicine in Tumor Metabolism, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, China
| | - Minjing Li
- Yantai Key Laboratory of Pharmacology of Traditional Chinese Medicine in Tumor Metabolism, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, China
| | - Xiaoyu Chen
- Yantai Key Laboratory of Pharmacology of Traditional Chinese Medicine in Tumor Metabolism, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, China
| | - Qiusheng Zheng
- Yantai Key Laboratory of Pharmacology of Traditional Chinese Medicine in Tumor Metabolism, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, China.,Key Laboratory of Xinjiang Endemic Phytomedicine Resources, Ministry of Education, School of Pharmacy, Shihezi University, Shihezi, China
| | - Xiaona Liu
- Yantai Key Laboratory of Pharmacology of Traditional Chinese Medicine in Tumor Metabolism, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, China
| | - Defang Li
- Yantai Key Laboratory of Pharmacology of Traditional Chinese Medicine in Tumor Metabolism, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, China
| |
Collapse
|
35
|
Wu X, Reboll MR, Korf-Klingebiel M, Wollert KC. Angiogenesis after acute myocardial infarction. Cardiovasc Res 2020; 117:1257-1273. [PMID: 33063086 DOI: 10.1093/cvr/cvaa287] [Citation(s) in RCA: 203] [Impact Index Per Article: 40.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 07/09/2020] [Accepted: 09/30/2020] [Indexed: 12/16/2022] Open
Abstract
Acute myocardial infarction (MI) inflicts massive injury to the coronary microcirculation leading to vascular disintegration and capillary rarefication in the infarct region. Tissue repair after MI involves a robust angiogenic response that commences in the infarct border zone and extends into the necrotic infarct core. Technological advances in several areas have provided novel mechanistic understanding of postinfarction angiogenesis and how it may be targeted to improve heart function after MI. Cell lineage tracing studies indicate that new capillary structures arise by sprouting angiogenesis from pre-existing endothelial cells (ECs) in the infarct border zone with no meaningful contribution from non-EC sources. Single-cell RNA sequencing shows that ECs in infarcted hearts may be grouped into clusters with distinct gene expression signatures, likely reflecting functionally distinct cell populations. EC-specific multicolour lineage tracing reveals that EC subsets clonally expand after MI. Expanding EC clones may arise from tissue-resident ECs with stem cell characteristics that have been identified in multiple organs including the heart. Tissue repair after MI involves interactions among multiple cell types which occur, to a large extent, through secreted proteins and their cognate receptors. While we are only beginning to understand the full complexity of this intercellular communication, macrophage and fibroblast populations have emerged as major drivers of the angiogenic response after MI. Animal data support the view that the endogenous angiogenic response after MI can be boosted to reduce scarring and adverse left ventricular remodelling. The improved mechanistic understanding of infarct angiogenesis therefore creates multiple therapeutic opportunities. During preclinical development, all proangiogenic strategies should be tested in animal models that replicate both cardiovascular risk factor(s) and the pharmacotherapy typically prescribed to patients with acute MI. Considering that the majority of patients nowadays do well after MI, clinical translation will require careful selection of patients in need of proangiogenic therapies.
Collapse
Affiliation(s)
- Xuekun Wu
- Division of Molecular and Translational Cardiology, Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg-Str. 1, Hannover 30625, Germany
| | - Marc R Reboll
- Division of Molecular and Translational Cardiology, Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg-Str. 1, Hannover 30625, Germany
| | - Mortimer Korf-Klingebiel
- Division of Molecular and Translational Cardiology, Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg-Str. 1, Hannover 30625, Germany
| | - Kai C Wollert
- Division of Molecular and Translational Cardiology, Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg-Str. 1, Hannover 30625, Germany
| |
Collapse
|
36
|
Huang CK, Dai D, Xie H, Zhu Z, Hu J, Su M, Liu M, Lu L, Shen W, Ning G, Wang J, Zhang R, Yan X. Lgr4 Governs a Pro-Inflammatory Program in Macrophages to Antagonize Post-Infarction Cardiac Repair. Circ Res 2020; 127:953-973. [PMID: 32600176 DOI: 10.1161/circresaha.119.315807] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
RATIONALE Macrophages are critically involved in wound healing following myocardial infarction (MI). Lgr4, a member of LGR (leucine-rich repeat-containing G protein-coupled receptor) family, is emerging as a regulator of macrophage-associated immune responses. However, the contribution of Lgr4 to macrophage phenotype and function in the context of MI remains unclear. OBJECTIVE To determine the role of macrophage Lgr4 in MI and to dissect the underlying mechanisms. METHODS AND RESULTS During early inflammatory phase of MI, infarct macrophages rather than neutrophils expressed high level of Lgr4. Macrophage-specific Lgr4 knockout mice had no baseline cardiovascular defects but manifested improved heart function, modestly reduced infarct size, decreased early mortality due to cardiac rupture, and ameliorated adverse remodeling after MI. Improved outcomes in macrophage-specific Lgr4 knockout mice subjected to MI were associated with mitigated ischemic injury and optimal infarct healing, as determined by reduction of cardiac apoptosis in the peri-infarct zone, attenuation of local myocardial inflammatory response, decrease of matrix metalloproteinase expression in the infarct, enhancement of angiogenesis, myofibroblast proliferation, and collagen I deposition in reparative granulation tissue as well as formation of collagen-rich scar. More importantly, macrophage-specific Lgr4 knockout infarcts had reduced numbers of infiltrating leukocytes and inflammatory macrophages but harbored abundant reparative macrophage subsets. Lgr4-null infarct macrophages exhibited a less inflammatory transcriptional signature. These findings were further supported by transcriptomic profiling data showing repression of multiple pathways and broad-spectrum genes associated with proinflammatory responses in macrophage-specific Lgr4 knockout infarcts. Notably, we discovered that Lgr4-mediated functional phenotype programing in infarct macrophages was at least partly attributed to regulation of AP (activator protein)-1 activity. We further demonstrated that the synergistic effects of Lgr4 on AP-1 activation in inflammatory macrophages occurred via enhancing CREB (cAMP response element-binding protein)-mediated c-Fos, Fosl1, and Fosb transactivation. CONCLUSIONS Together, our data highlight the significance of Lgr4 in governing proinflammatory phenotype of infarct macrophages and postinfarction repair.
Collapse
Affiliation(s)
- Chun-Kai Huang
- From the Department of Cardiology (C.-K.H., D.D., H.X., Z.Z., J.H., L.L., W.S., R.Z., X.Y.), Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
- Ruijin Hospital, Institute of Cardiovascular Diseases (C.-K.H., D.D., H.X., Z.Z., J.H., L.L., W.S., R.Z., X.Y.), Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Daopeng Dai
- From the Department of Cardiology (C.-K.H., D.D., H.X., Z.Z., J.H., L.L., W.S., R.Z., X.Y.), Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
- Ruijin Hospital, Institute of Cardiovascular Diseases (C.-K.H., D.D., H.X., Z.Z., J.H., L.L., W.S., R.Z., X.Y.), Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Hongyang Xie
- From the Department of Cardiology (C.-K.H., D.D., H.X., Z.Z., J.H., L.L., W.S., R.Z., X.Y.), Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
- Ruijin Hospital, Institute of Cardiovascular Diseases (C.-K.H., D.D., H.X., Z.Z., J.H., L.L., W.S., R.Z., X.Y.), Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Zhengbin Zhu
- From the Department of Cardiology (C.-K.H., D.D., H.X., Z.Z., J.H., L.L., W.S., R.Z., X.Y.), Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
- Ruijin Hospital, Institute of Cardiovascular Diseases (C.-K.H., D.D., H.X., Z.Z., J.H., L.L., W.S., R.Z., X.Y.), Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Jian Hu
- From the Department of Cardiology (C.-K.H., D.D., H.X., Z.Z., J.H., L.L., W.S., R.Z., X.Y.), Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
- Ruijin Hospital, Institute of Cardiovascular Diseases (C.-K.H., D.D., H.X., Z.Z., J.H., L.L., W.S., R.Z., X.Y.), Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Min Su
- Department of Pathology, Institute of Clinical Pathology, Shantou University Medical College, Guangdong, PR China (M.S.)
| | - Mingyao Liu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, PR China (M.L.)
| | - Lin Lu
- From the Department of Cardiology (C.-K.H., D.D., H.X., Z.Z., J.H., L.L., W.S., R.Z., X.Y.), Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Weifeng Shen
- From the Department of Cardiology (C.-K.H., D.D., H.X., Z.Z., J.H., L.L., W.S., R.Z., X.Y.), Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
- Ruijin Hospital, Institute of Cardiovascular Diseases (C.-K.H., D.D., H.X., Z.Z., J.H., L.L., W.S., R.Z., X.Y.), Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Guang Ning
- Department of Endocrinology and Metabolism (G.N., J.W.), Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Jiqiu Wang
- Department of Endocrinology and Metabolism (G.N., J.W.), Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Ruiyan Zhang
- From the Department of Cardiology (C.-K.H., D.D., H.X., Z.Z., J.H., L.L., W.S., R.Z., X.Y.), Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
- Ruijin Hospital, Institute of Cardiovascular Diseases (C.-K.H., D.D., H.X., Z.Z., J.H., L.L., W.S., R.Z., X.Y.), Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Xiaoxiang Yan
- From the Department of Cardiology (C.-K.H., D.D., H.X., Z.Z., J.H., L.L., W.S., R.Z., X.Y.), Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
- Ruijin Hospital, Institute of Cardiovascular Diseases (C.-K.H., D.D., H.X., Z.Z., J.H., L.L., W.S., R.Z., X.Y.), Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| |
Collapse
|
37
|
Sasaki T, Shimazawa M, Kanamori H, Yamada Y, Nishinaka A, Kuse Y, Suzuki G, Masuda T, Nakamura S, Hosokawa M, Minatoguchi S, Hara H. Effects of progranulin on the pathological conditions in experimental myocardial infarction model. Sci Rep 2020; 10:11842. [PMID: 32678228 PMCID: PMC7367277 DOI: 10.1038/s41598-020-68804-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 05/28/2020] [Indexed: 12/13/2022] Open
Abstract
Progranulin is a secreted growth factor associated with multiple physiological functions in ischemic pathophysiology. However, it is still not fully understood how progranulin is involved in ischemic lesion and cardiac remodeling after myocardial infarction (MI). In this study, we investigated the effects of progranulin on myocardial ischemia and reperfusion injury. We investigated progranulin expression using Western blotting and immunostaining after permanent left coronary artery (LCA) occlusion in mice. Infarct size and the number of infiltrating neutrophils were measured 24 h after permanent LCA occlusion. Recombinant mouse progranulin was administered before LCA occlusion. In addition, we evaluated cardiac function using cardiac catheterization and echocardiography, and fibrosis size by Masson's trichrome staining after myocardial ischemia/reperfusion in rabbits. Recombinant human progranulin was administered immediately after induction of reperfusion. Progranulin expression increased in the myocardial ischemic area 1, 3, and 5 days after permanent LCA occlusion in mice. The administration of recombinant mouse progranulin significantly attenuated infarct size and infiltrating neutrophils 24 h after permanent LCA occlusion in mice. We also found that administration of recombinant human progranulin ameliorated the deterioration of cardiac dysfunction and fibrosis after myocardial ischemia/reperfusion in rabbits. These findings suggest that progranulin may protect myocardial ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Takahiro Sasaki
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu, 501-1196, Japan
| | - Masamitsu Shimazawa
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu, 501-1196, Japan
| | - Hiromitsu Kanamori
- Department of Cardiology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Yoshihisa Yamada
- Department of Cardiology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Anri Nishinaka
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu, 501-1196, Japan
| | - Yoshiki Kuse
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu, 501-1196, Japan
| | - Genjiro Suzuki
- Dementia Research Project, Department of Dementia and Higher Brain Function, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Tomomi Masuda
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu, 501-1196, Japan
| | - Shinsuke Nakamura
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu, 501-1196, Japan
| | - Masato Hosokawa
- Dementia Research Project, Department of Dementia and Higher Brain Function, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Shinya Minatoguchi
- Department of Circulatory and Respiratory Advanced Medicine, Gifu University Graduate School of Medicine, Gifu, Japan
- Heart Failure Center, Gifu Municipal Hospital, Gifu, Japan
| | - Hideaki Hara
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu, 501-1196, Japan.
| |
Collapse
|
38
|
Abaricia JO, Shah AH, Chaubal M, Hotchkiss KM, Olivares-Navarrete R. Wnt signaling modulates macrophage polarization and is regulated by biomaterial surface properties. Biomaterials 2020; 243:119920. [PMID: 32179303 PMCID: PMC7191325 DOI: 10.1016/j.biomaterials.2020.119920] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 02/15/2020] [Accepted: 02/25/2020] [Indexed: 12/28/2022]
Abstract
Macrophages are among the first cells to interact with biomaterials and ultimately determine their integrative fate. Biomaterial surface characteristics like roughness and hydrophilicity can activate macrophages to an anti-inflammatory phenotype. Wnt signaling, a key cell proliferation and differentiation pathway, has been associated with dysregulated macrophage activity in disease. However, the role Wnt signaling plays in macrophage activation and response to biomaterials is unknown. The aim of this study was to characterize the regulation of Wnt signaling in macrophages during classical pro- and anti-inflammatory polarization and in their response to smooth, rough, and rough-hydrophilic titanium (Ti) surfaces. Peri-implant Wnt signaling in macrophage-ablated (MaFIA) mice instrumented with intramedullary Ti rods was significantly attenuated compared to untreated controls. Wnt ligand mRNA were upregulated in a surface modification-dependent manner in macrophages isolated from the surface of Ti implanted in C57Bl/6 mice. In vitro, Wnt mRNAs were regulated in primary murine bone-marrow-derived macrophages cultured on Ti in a surface modification-dependent manner. When macrophageal Wnt secretion was inhibited, macrophage sensitivity to both physical and biological stimuli was abrogated. Loss of macrophage-derived Wnts also impaired recruitment of mesenchymal stem cells and T-cells to Ti implants in vivo. Finally, inhibition of integrin signaling decreased surface-dependent upregulation of Wnt genes. These results suggest that Wnt signaling regulates macrophage response to biomaterials and that macrophages are an important source of Wnt ligands during inflammation and healing.
Collapse
Affiliation(s)
- Jefferson O Abaricia
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA, United States
| | - Arth H Shah
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA, United States
| | - Manotri Chaubal
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA, United States
| | - Kelly M Hotchkiss
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA, United States
| | - Rene Olivares-Navarrete
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA, United States.
| |
Collapse
|
39
|
Blankesteijn WM. Interventions in WNT Signaling to Induce Cardiomyocyte Proliferation: Crosstalk with Other Pathways. Mol Pharmacol 2020; 97:90-101. [PMID: 31757861 DOI: 10.1124/mol.119.118018] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 11/06/2019] [Indexed: 12/26/2022] Open
Abstract
Myocardial infarction is a frequent cardiovascular event and a major cause for cardiomyocyte loss. In adult mammals, cardiomyocytes are traditionally considered to be terminally differentiated cells, unable to proliferate. Therefore, the wound-healing response in the infarct area typically yields scar tissue rather than newly formed cardiomyocytes. In the last decade, several lines of evidence have challenged the lack of proliferative capacity of the differentiated cardiomyocyte: studies in zebrafish and neonatal mammals have convincingly demonstrated the regenerative capacity of cardiomyocytes. Moreover, multiple signaling pathways have been identified in these models that-when activated in adult mammalian cardiomyocytes-can reactivate the cell cycle in these cells. However, cardiomyocytes frequently exit the cell cycle before symmetric division into daughter cells, leading to polyploidy and multinucleation. Now that there is more insight into the reactivation of the cell cycle machinery, other prerequisites for successful symmetric division of cardiomyocytes, such as the control of sarcomere disassembly to allow cytokinesis, require more investigation. This review aims to discuss the signaling pathways involved in cardiomyocyte proliferation, with a specific focus on wingless/int-1 protein signaling. Comparing the conflicting results from in vitro and in vivo studies on this pathway illustrates that the interaction with other cells and structures around the infarct is likely to be essential to determine the outcome of these interventions. The extensive crosstalk with other pathways implicated in cardiomyocyte proliferation calls for the identification of nodal points in the cell signaling before cardiomyocyte proliferation can be moved forward toward clinical application as a cure of cardiac disease. SIGNIFICANCE STATEMENT: Evidence is mounting that proliferation of pre-existing cardiomyocytes can be stimulated to repair injury of the heart. In this review article, an overview is provided of the different signaling pathways implicated in cardiomyocyte proliferation with emphasis on wingless/int-1 protein signaling, crosstalk between the pathways, and controversial results obtained in vitro and in vivo.
Collapse
Affiliation(s)
- W Matthijs Blankesteijn
- Department of Pharmacology and Toxicology, Cardiovascular Research Institute Maastricht, Maastricht University, The Netherlands
| |
Collapse
|
40
|
Fretwurst T, Garaicoa‐Pazmino C, Nelson K, Giannobile WV, Squarize CH, Larsson L, Castilho RM. Characterization of macrophages infiltrating peri‐implantitis lesions. Clin Oral Implants Res 2020; 31:274-281. [DOI: 10.1111/clr.13568] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 12/09/2019] [Accepted: 12/17/2019] [Indexed: 12/25/2022]
Affiliation(s)
- Tobias Fretwurst
- Department of Periodontics and Oral Medicine University of Michigan School of Dentistry Ann Arbor MI USA
- Department of Oral‐ and Maxillofacial Surgery Medical Center – University of Freiburg Faculty of Medicine University of Freiburg Germany
- Laboratory of Epithelial Biology Department of Periodontics and Oral Medicine University of Michigan School Ann Arbor MI USA
| | - Carlos Garaicoa‐Pazmino
- Department of Periodontology School of Dentistry Oregon Health & Science University Portland OR USA
- Laboratory of Epithelial Biology Department of Periodontics and Oral Medicine University of Michigan School Ann Arbor MI USA
| | - Katja Nelson
- Department of Oral‐ and Maxillofacial Surgery Medical Center – University of Freiburg Faculty of Medicine University of Freiburg Germany
| | - William V. Giannobile
- Department of Periodontics and Oral Medicine University of Michigan School of Dentistry Ann Arbor MI USA
| | - Cristiane H. Squarize
- Department of Periodontics and Oral Medicine University of Michigan School of Dentistry Ann Arbor MI USA
- Laboratory of Epithelial Biology Department of Periodontics and Oral Medicine University of Michigan School Ann Arbor MI USA
| | - Lena Larsson
- Department of Periodontics and Oral Medicine University of Michigan School of Dentistry Ann Arbor MI USA
- Department of Periodontology Institute of Odontology University of Gothenburg Gothenburg Sweden
| | - Rogerio M. Castilho
- Department of Periodontics and Oral Medicine University of Michigan School of Dentistry Ann Arbor MI USA
- Laboratory of Epithelial Biology Department of Periodontics and Oral Medicine University of Michigan School Ann Arbor MI USA
| |
Collapse
|
41
|
Overexpression of miR-375 Protects Cardiomyocyte Injury following Hypoxic-Reoxygenation Injury. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:7164069. [PMID: 31976033 PMCID: PMC6961604 DOI: 10.1155/2020/7164069] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Revised: 12/05/2019] [Accepted: 12/17/2019] [Indexed: 12/11/2022]
Abstract
The aim of the study was to evaluate the clinical significance of microRNA-375 in acute myocardial infarction patients and its mimic action in hypoxia/reoxygenation- (H/R-) induced ventricular cardiomyocyte H9c2 injury. In the current study, 90 ST-elevated acute MI patients (STEMI), 75 non-ST-elevated acute MI patients (NSTEMI), 90 healthy subjects, 14 weeks old mice, and ventricular cardiomyocyte H9c2 were included. The expressions of plasma microRNA-375 in patients with STEMI and NSTEMI and AMI mouse models were remarkably decreased than in controls (P < 0.001). The areas under the curve (AUC) of plasma microRNA-375 were revealed 0.939 in STEMI and 0.935 in NSTEMI subjects. Moreover, microRNA-375 levels in H/R-exposed cardiac H9c2 cells were evidently downregulated and significantly increased apoptosis rate and caspase-3 activity levels, while overexpression of miR-375 remarkably reduced apoptosis percentage and caspase-3 levels as compared with normal cells. Furthermore, this study also demonstrated that Nemo-like kinase (NLK), NLK mRNA, and protein expression levels were significantly downregulated in H/R-injured H9c2 cells, on the contrary, H9c2 cells transfected with mimic-miR-375 greatly upregulated NLK mRNA and protein expression. Plasma microRNA-375 may serve as an essential clinical biomarker for diagnosis of early-stage AMI. Mimic expression of miR-375 significantly prevented H/R-induced cardiomyocyte injury by decreasing caspase-3 activity through upregulation of the NLK gene, recommended as a new therapeutic option for AMI patient.
Collapse
|
42
|
Ljungberg JK, Kling JC, Tran TT, Blumenthal A. Functions of the WNT Signaling Network in Shaping Host Responses to Infection. Front Immunol 2019; 10:2521. [PMID: 31781093 PMCID: PMC6857519 DOI: 10.3389/fimmu.2019.02521] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 10/10/2019] [Indexed: 12/15/2022] Open
Abstract
It is well-established that aberrant WNT expression and signaling is associated with developmental defects, malignant transformation and carcinogenesis. More recently, WNT ligands have emerged as integral components of host responses to infection but their functions in the context of immune responses are incompletely understood. Roles in the modulation of inflammatory cytokine production, host cell intrinsic innate defense mechanisms, as well as the bridging of innate and adaptive immunity have been described. To what degree WNT responses are defined by the nature of the invading pathogen or are specific for subsets of host cells is currently not well-understood. Here we provide an overview of WNT responses during infection with phylogenetically diverse pathogens and highlight functions of WNT ligands in the host defense against infection. Detailed understanding of how the WNT network orchestrates immune cell functions will not only improve our understanding of the fundamental principles underlying complex immune response, but also help identify therapeutic opportunities or potential risks associated with the pharmacological targeting of the WNT network, as currently pursued for novel therapeutics in cancer and bone disorders.
Collapse
Affiliation(s)
- Johanna K Ljungberg
- The University of Queensland Diamantina Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Jessica C Kling
- The University of Queensland Diamantina Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Thao Thanh Tran
- The University of Queensland Diamantina Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Antje Blumenthal
- The University of Queensland Diamantina Institute, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
43
|
Wnt Signaling in the Regulation of Immune Cell and Cancer Therapeutics. Cells 2019; 8:cells8111380. [PMID: 31684152 PMCID: PMC6912555 DOI: 10.3390/cells8111380] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Revised: 10/21/2019] [Accepted: 11/01/2019] [Indexed: 12/17/2022] Open
Abstract
Wnt signaling is one of the important pathways to play a major role in various biological processes, such as embryonic stem-cell development, tissue regeneration, cell differentiation, and immune cell regulation. Recent studies suggest that Wnt signaling performs an essential function in immune cell modulation and counteracts various disorders. Nonetheless, the emerging role and mechanism of action of this signaling cascade in immune cell regulation, as well as its involvement in various cancers, remain debatable. The Wnt signaling in immune cells is very diverse, e.g., the tolerogenic role of dendritic cells, the development of natural killer cells, thymopoiesis of T cells, B-cell-driven initiation of T-cells, and macrophage actions in tissue repair, regeneration, and fibrosis. The purpose of this review is to highlight the current therapeutic targets in (and the prospects of) Wnt signaling, as well as the potential suitability of available modulators for the development of cancer immunotherapies. Although there are several Wnt inhibitors relevant to cancer, it would be worthwhile to extend this approach to immune cells.
Collapse
|
44
|
Cosin-Roger J, Ortiz-Masià MD, Barrachina MD. Macrophages as an Emerging Source of Wnt Ligands: Relevance in Mucosal Integrity. Front Immunol 2019; 10:2297. [PMID: 31608072 PMCID: PMC6769121 DOI: 10.3389/fimmu.2019.02297] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 09/11/2019] [Indexed: 02/06/2023] Open
Abstract
The Wnt signaling pathway is a conserved pathway involved in important cellular processes such as the control of embryonic development, cellular polarity, cellular migration, and cell proliferation. In addition to playing a central role during embryogenesis, this pathway is also an essential part of adult homeostasis. Indeed, it controls the proliferation of epithelial cells in different organs such as intestine, lung, and kidney, and guarantees the maintenance of the mucosa in physiological conditions. The origin of this molecular pathway is the binding between Wnt ligands (belonging to a family of 19 different homologous secreted glycoproteins) and their specific membrane receptors, from the Frizzled receptor family. This specific interaction triggers the activation of the signaling cascade, which in turn activates or suppresses the expression of different genes in order to change the behavior of the cell. On the other hand, alterations of this pathway have been described in pathological conditions such as inflammation, fibrosis, and cancer. In recent years, macrophages-among other cell types-have emerged as a potential source of Wnt ligands. Due to their high plasticity, macrophages, which are central to the innate immune response, are capable of adopting different phenotypes depending on their microenvironment. In the past, two different phenotypes were described: a proinflammatory phenotype-M1 macrophages-and an anti-inflammatory phenotype-M2 macrophages-and a selective expression of Wnt ligands has been associated with said phenotypes. However, nowadays it is assumed that macrophages in vivo move through a continual spectrum of functional phenotypes. In both physiological and pathological (inflammation, fibrosis and cancer) conditions, the accumulation and polarization of macrophages conditions the future of the tissue, facilitating various scenarios, such as resolution of inflammation, activation of fibrosis, and cancer development due to the modulation of the Wnt signaling pathway, in autocrine and paracrine manner. In this work, we provide an overview of studies that have explored the role of macrophages and how they act as a source of Wnt ligands and as mediators of mucosal integrity.
Collapse
Affiliation(s)
| | - Mª Dolores Ortiz-Masià
- Departamento de Medicina, Facultad de Medicina, Universidad de Valencia, Valencia, Spain
| | - Mª Dolores Barrachina
- Departamento de Farmacología and CIBER, Facultad de Medicina, Universidad de Valencia, Valencia, Spain
| |
Collapse
|
45
|
Farbehi N, Patrick R, Dorison A, Xaymardan M, Janbandhu V, Wystub-Lis K, Ho JW, Nordon RE, Harvey RP. Single-cell expression profiling reveals dynamic flux of cardiac stromal, vascular and immune cells in health and injury. eLife 2019; 8:43882. [PMID: 30912746 PMCID: PMC6459677 DOI: 10.7554/elife.43882] [Citation(s) in RCA: 380] [Impact Index Per Article: 63.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 03/25/2019] [Indexed: 12/11/2022] Open
Abstract
Besides cardiomyocytes (CM), the heart contains numerous interstitial cell types which play key roles in heart repair, regeneration and disease, including fibroblast, vascular and immune cells. However, a comprehensive understanding of this interactive cell community is lacking. We performed single-cell RNA-sequencing of the total non-CM fraction and enriched (Pdgfra-GFP+) fibroblast lineage cells from murine hearts at days 3 and 7 post-sham or myocardial infarction (MI) surgery. Clustering of >30,000 single cells identified >30 populations representing nine cell lineages, including a previously undescribed fibroblast lineage trajectory present in both sham and MI hearts leading to a uniquely activated cell state defined in part by a strong anti-WNT transcriptome signature. We also uncovered novel myofibroblast subtypes expressing either pro-fibrotic or anti-fibrotic signatures. Our data highlight non-linear dynamics in myeloid and fibroblast lineages after cardiac injury, and provide an entry point for deeper analysis of cardiac homeostasis, inflammation, fibrosis, repair and regeneration. In our bodies, heart attacks lead to cell death and inflammation. This is then followed by a healing phase where the organ repairs itself. There are many types of heart cells, from muscle and pacemaker cells that help to create the beating motion, to so-called fibroblasts that act as a supporting network. Yet, it is still unclear how individual cells participate in the heart's response to injury. All cells possess the same genetic information, but they turn on or off different genes depending on the specific tasks that they need to perform. Spotting which genes are activated in individual cells can therefore provide clues about their exact roles in the body. Until recently, technological limitations meant that this information was difficult to access, because it was only possible to capture the global response of a group of cells in a sample. A new method called single-cell RNA sequencing is now allowing researchers to study the activities of many genes in thousands of individual cells at the same time. Here, Farbehi, Patrick et al. performed single-cell RNA sequencing on over 30,000 individual cells from healthy and injured mouse hearts. Computational approaches were then used to cluster cells into groups according to the activities of their genes. The experiments identified over 30 distinct sub-types of cell, including several that were previously unknown. For example, a group of fibroblasts that express a gene called Wif1 was discovered. Previous genetic studies have shown that Wif1 is essential for the heart's response to injury. Further experiments by Farbehi, Patrick et al. indicated that this new sub-type of cells may control the timing of the different aspects of heart repair after damage. Tens of millions of people around the world suffer from heart attacks and other heart diseases. Knowing how different types of heart cells participate in repair mechanisms may help to find new targets for drugs and other treatments.
Collapse
Affiliation(s)
- Nona Farbehi
- Victor Chang Cardiac Research Institute, Darlinghurst, Australia.,Stem Cells Australia, Melbourne Brain Centre, University of Melbourne, Victoria, Australia.,Garvan Weizmann Centre for Cellular Genomics, Garvan Institute of Medical Research, Sydney, Australia.,Graduate School of Biomedical Engineering, UNSW Sydney, Kensington, Australia
| | - Ralph Patrick
- Victor Chang Cardiac Research Institute, Darlinghurst, Australia.,Stem Cells Australia, Melbourne Brain Centre, University of Melbourne, Victoria, Australia.,St. Vincent's Clinical School, UNSW Sydney, Kensington, Australia
| | - Aude Dorison
- Victor Chang Cardiac Research Institute, Darlinghurst, Australia.,Stem Cells Australia, Melbourne Brain Centre, University of Melbourne, Victoria, Australia
| | - Munira Xaymardan
- Victor Chang Cardiac Research Institute, Darlinghurst, Australia.,Stem Cells Australia, Melbourne Brain Centre, University of Melbourne, Victoria, Australia.,School of Dentistry, Faculty of Medicine and Health, University of Sydney, Westmead Hospital, Westmead, Australia
| | - Vaibhao Janbandhu
- Victor Chang Cardiac Research Institute, Darlinghurst, Australia.,Stem Cells Australia, Melbourne Brain Centre, University of Melbourne, Victoria, Australia.,St. Vincent's Clinical School, UNSW Sydney, Kensington, Australia
| | | | - Joshua Wk Ho
- Victor Chang Cardiac Research Institute, Darlinghurst, Australia.,St. Vincent's Clinical School, UNSW Sydney, Kensington, Australia
| | - Robert E Nordon
- Stem Cells Australia, Melbourne Brain Centre, University of Melbourne, Victoria, Australia.,Graduate School of Biomedical Engineering, UNSW Sydney, Kensington, Australia
| | - Richard P Harvey
- Victor Chang Cardiac Research Institute, Darlinghurst, Australia.,Stem Cells Australia, Melbourne Brain Centre, University of Melbourne, Victoria, Australia.,School of Biotechnology and Biomolecular Science, UNSW Sydney, Kensington, Australia
| |
Collapse
|
46
|
Xu JY, Xiong YY, Lu XT, Yang YJ. Regulation of Type 2 Immunity in Myocardial Infarction. Front Immunol 2019; 10:62. [PMID: 30761134 PMCID: PMC6362944 DOI: 10.3389/fimmu.2019.00062] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 01/11/2019] [Indexed: 12/12/2022] Open
Abstract
Type 2 immunity participates in the pathogeneses of helminth infection and allergic diseases. Emerging evidence indicates that the components of type 2 immunity are also involved in maintaining metabolic hemostasis and facilitating the healing process after tissue injury. Numerous preclinical studies have suggested regulation of type 2 immunity-related cytokines, such as interleukin-4, -13, and -33, and cell types, such as M2 macrophages, mast cells, and eosinophils, affects cardiac functions after myocardial infarction (MI), providing new insights into the importance of immune modulation in the infarcted heart. This review provides an overview of the functions of these cytokines and cells in the setting of MI as well as their potential to predict the severity and prognosis of MI.
Collapse
Affiliation(s)
- Jun-Yan Xu
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Yu-Yan Xiong
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Xiao-Tong Lu
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yue-Jin Yang
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| |
Collapse
|
47
|
Jung M, Dodsworth M, Thum T. Inflammatory cells and their non-coding RNAs as targets for treating myocardial infarction. Basic Res Cardiol 2018; 114:4. [PMID: 30523422 PMCID: PMC6290728 DOI: 10.1007/s00395-018-0712-z] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Accepted: 11/29/2018] [Indexed: 12/22/2022]
Abstract
Myocardial infarction triggers infiltration of several types of immune cells that coordinate both innate and adaptive immune responses. These play a dual role in post-infarction cardiac remodeling by initiating and resolving inflammatory processes, which needs to occur in a timely and well-orchestrated way to ensure a reestablishment of normalized cardiac functions. Thus, therapeutic modulation of immune responses might have benefits for infarct patients. While such strategies have shown great potential in treating cancer, applications in the post-infarction context have been disappointing. One challenge has been the complexity and plasticity of immune cells and their functions in cardiac regulation and healing. The types appear in patterns that are temporally and spatially distinct, while influencing each other and the surrounding tissue. A comprehensive understanding of the immune cell repertoire and their regulatory functions following infarction is sorely needed. Processes of cardiac remodeling trigger additional genetic changes that may also play critical roles in the aftermath of cardiovascular disease. Some of these changes involve non-coding RNAs that play crucial roles in the regulation of immune cells and may, therefore, be of therapeutic interest. This review summarizes what is currently known about the functions of immune cells and non-coding RNAs during post-infarction wound healing. We address some of the challenges that remain and describe novel therapeutic approaches under development that are based on regulating immune responses through non-coding RNAs in the aftermath of the disease.
Collapse
Affiliation(s)
- Mira Jung
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Michael Dodsworth
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.
- National Heart and Lung Institute, Imperial College London, London, UK.
| |
Collapse
|
48
|
Meyer IS, Leuschner F. The role of Wnt signaling in the healing myocardium: a focus on cell specificity. Basic Res Cardiol 2018; 113:44. [PMID: 30327885 DOI: 10.1007/s00395-018-0705-y] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 10/01/2018] [Accepted: 10/09/2018] [Indexed: 12/18/2022]
Abstract
Various cell types are involved in the healing process after myocardial infarction (MI). Besides cardiac resident cells (such as cardiomyocytes, fibroblasts and endothelial cells) already present at the lesion site, a massive influx of leukocytes (mainly monocytes and neutrophils) is observed within hours after the ischemic event. So far, little is known about modes of interaction of these cells. Wnt signaling is an evolutionary conserved signaling cassette known to play an important role in cell-cell communication. While the overall reactivation of Wnt signaling upon ischemic injury is well described, the precise expression pattern of Wnt proteins, however, is far from understood. We here describe known Wnt components that partake in MI healing and differentiate cell-specific aspects. The secretion of Wnt proteins and their antagonists in the context of cardiac inflammation after MI appear to be tightly regulated in a spatial-temporal manner. Overall, we aim to stress the importance of elucidating not only Wnt component-specific aspects, but also their sometimes contradicting effects in different target cells. A better understanding of Wnt signaling in MI healing may eventually lead to the development of successful therapeutic approaches in an often considered "un-druggable" pathway.
Collapse
Affiliation(s)
- Ingmar Sören Meyer
- Department of Internal Medicine III, University Hospital Heidelberg, University of Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner site Heidelberg/Mannheim, Heidelberg, Germany
| | - Florian Leuschner
- Department of Internal Medicine III, University Hospital Heidelberg, University of Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany.
- DZHK (German Centre for Cardiovascular Research), Partner site Heidelberg/Mannheim, Heidelberg, Germany.
| |
Collapse
|
49
|
Chae WJ, Bothwell ALM. Canonical and Non-Canonical Wnt Signaling in Immune Cells. Trends Immunol 2018; 39:830-847. [PMID: 30213499 DOI: 10.1016/j.it.2018.08.006] [Citation(s) in RCA: 148] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 08/12/2018] [Accepted: 08/15/2018] [Indexed: 12/18/2022]
Abstract
Cell differentiation, proliferation, and death are vital for immune homeostasis. Wnt signaling plays essential roles in processes across species. The roles of Wnt signaling proteins and Wnt ligands have been studied in the past, but the context-dependent mechanisms and functions of these pathways in immune responses remain unclear. Recent findings regarding the role of Wnt ligands and Wnt signaling in immune cells and their immunomodulatory mechanisms suggest that Wnt ligands and signaling are significant in regulating immune responses. We introduce recent key findings and future perspectives on Wnt ligands and their signaling pathways in immune cells as well as the immunological roles and functions of Wnt antagonists.
Collapse
Affiliation(s)
- Wook-Jin Chae
- Department of Immunobiology, Yale University School of Medicine, 300 Cedar Street, New Haven, CT 06520, USA.
| | - Alfred L M Bothwell
- Department of Immunobiology, Yale University School of Medicine, 300 Cedar Street, New Haven, CT 06520, USA.
| |
Collapse
|
50
|
Meyer IS, Jungmann A, Dieterich C, Zhang M, Lasitschka F, Werkmeister S, Haas J, Müller OJ, Boutros M, Nahrendorf M, Katus HA, Hardt SE, Leuschner F. The cardiac microenvironment uses non-canonical WNT signaling to activate monocytes after myocardial infarction. EMBO Mol Med 2018; 9:1279-1293. [PMID: 28774883 PMCID: PMC5582413 DOI: 10.15252/emmm.201707565] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
A disturbed inflammatory response following myocardial infarction (MI) is associated with poor prognosis and increased tissue damage. Monocytes are key players in healing after MI, but little is known about the role of the cardiac niche in monocyte activation. This study investigated microenvironment‐dependent changes in inflammatory monocytes after MI. RNA sequencing analysis of murine Ly6Chigh monocytes on day 3 after MI revealed differential regulation depending on location. Notably, the local environment strongly impacted components of the WNT signaling cascade. Analysis of WNT modulators revealed a strong upregulation of WNT Inhibitory Factor 1 (WIF1) in cardiomyocytes—but not fibroblasts or endothelial cells—upon hypoxia. Compared to wild‐type (WT) littermates, WIF1 knockout mice showed severe adverse remodeling marked by increased scar size and reduced ejection fraction 4 weeks after MI. While FACS analysis on day 1 after MI revealed no differences in neutrophil numbers, the hearts of WIF1 knockouts contained significantly more inflammatory monocytes than hearts from WT animals. Next, we induced AAV‐mediated cardiomyocyte‐specific WIF1 overexpression, which attenuated the monocyte response and improved cardiac function after MI, as compared to control‐AAV‐treated animals. Finally, WIF1 overexpression in isolated cardiomyocytes limited the activation of non‐canonical WNT signaling and led to reduced IL‐1β and IL‐6 expression in monocytes/macrophages. Taken together, we investigated the cardiac microenvironment's interaction with recruited monocytes after MI and identified a novel mechanism of monocyte activation. The local initiation of non‐canonical WNT signaling shifts the accumulating myeloid cells toward a pro‐inflammatory state and impacts healing after myocardial infarction.
Collapse
Affiliation(s)
- Ingmar Sören Meyer
- Department of Medicine III, University of Heidelberg, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Partnersite, Heidelberg/Mannheim, Germany
| | - Andreas Jungmann
- Department of Medicine III, University of Heidelberg, Heidelberg, Germany
| | - Christoph Dieterich
- Department of Medicine III, University of Heidelberg, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Partnersite, Heidelberg/Mannheim, Germany
| | - Min Zhang
- Department of Medicine III, University of Heidelberg, Heidelberg, Germany
| | - Felix Lasitschka
- Institute of Pathology, University of Heidelberg, Heidelberg, Germany.,Tissue Bank of the National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Susann Werkmeister
- Department of Medicine III, University of Heidelberg, Heidelberg, Germany
| | - Jan Haas
- Department of Medicine III, University of Heidelberg, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Partnersite, Heidelberg/Mannheim, Germany
| | - Oliver J Müller
- Department of Medicine III, University of Heidelberg, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Partnersite, Heidelberg/Mannheim, Germany
| | - Michael Boutros
- DZHK (German Centre for Cardiovascular Research), Partnersite, Heidelberg/Mannheim, Germany.,Division Signaling and Functional Genomics, German Cancer Research Center (DKFZ) and Heidelberg University, Heidelberg, Germany
| | - Matthias Nahrendorf
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Hugo A Katus
- Department of Medicine III, University of Heidelberg, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Partnersite, Heidelberg/Mannheim, Germany
| | - Stefan E Hardt
- Department of Medicine III, University of Heidelberg, Heidelberg, Germany
| | - Florian Leuschner
- Department of Medicine III, University of Heidelberg, Heidelberg, Germany .,DZHK (German Centre for Cardiovascular Research), Partnersite, Heidelberg/Mannheim, Germany
| |
Collapse
|