1
|
Xia T, Yu J, Du M, Chen X, Wang C, Li R. Vascular endothelial cell injury: causes, molecular mechanisms, and treatments. MedComm (Beijing) 2025; 6:e70057. [PMID: 39931738 PMCID: PMC11809559 DOI: 10.1002/mco2.70057] [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: 06/12/2024] [Revised: 12/16/2024] [Accepted: 12/17/2024] [Indexed: 02/13/2025] Open
Abstract
Vascular endothelial cells form a single layer of flat cells that line the inner surface of blood vessels, extending from large vessels to the microvasculature of various organs. These cells are crucial metabolic and endocrine components of the body, playing vital roles in maintaining circulatory stability, regulating vascular tone, and preventing coagulation and thrombosis. Endothelial cell injury is regarded as a pivotal initiating factor in the pathogenesis of various diseases, triggered by multiple factors, including infection, inflammation, and hemodynamic changes, which significantly compromise vascular integrity and function. This review examines the causes, underlying molecular mechanisms, and potential therapeutic approaches for endothelial cell injury, focusing specifically on endothelial damage in cardiac ischemia/reperfusion (I/R) injury, sepsis, and diabetes. It delves into the intricate signaling pathways involved in endothelial cell injury, emphasizing the roles of oxidative stress, mitochondrial dysfunction, inflammatory mediators, and barrier damage. Current treatment strategies-ranging from pharmacological interventions to regenerative approaches and lifestyle modifications-face ongoing challenges and limitations. Overall, this review highlights the importance of understanding endothelial cell injury within the context of various diseases and the necessity for innovative therapeutic methods to improve patient outcomes.
Collapse
Affiliation(s)
- Tian Xia
- Department of Laboratory MedicineThe First Medical Center of Chinese PLA General HospitalBeijingChina
- Department of Laboratory MedicineMedical School of Chinese PLABeijingChina
| | - Jiachi Yu
- Department of Laboratory MedicineThe First Medical Center of Chinese PLA General HospitalBeijingChina
- Department of Laboratory MedicineMedical School of Chinese PLABeijingChina
| | - Meng Du
- Department of Laboratory MedicineThe First Medical Center of Chinese PLA General HospitalBeijingChina
- Department of Clinical LaboratoryHuaian Hospital of Huaian CityHuaianJiangsuChina
| | - Ximeng Chen
- Department of Laboratory MedicineThe First Medical Center of Chinese PLA General HospitalBeijingChina
- Department of Laboratory MedicineMedical School of Chinese PLABeijingChina
| | - Chengbin Wang
- Department of Laboratory MedicineThe First Medical Center of Chinese PLA General HospitalBeijingChina
- Department of Laboratory MedicineMedical School of Chinese PLABeijingChina
| | - Ruibing Li
- Department of Laboratory MedicineThe First Medical Center of Chinese PLA General HospitalBeijingChina
- Department of Laboratory MedicineMedical School of Chinese PLABeijingChina
| |
Collapse
|
2
|
Huang RL, Li Q, Ma JX, Atala A, Zhang Y. Body fluid-derived stem cells - an untapped stem cell source in genitourinary regeneration. Nat Rev Urol 2023; 20:739-761. [PMID: 37414959 PMCID: PMC11639537 DOI: 10.1038/s41585-023-00787-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/30/2023] [Indexed: 07/08/2023]
Abstract
Somatic stem cells have been obtained from solid organs and tissues, including the bone marrow, placenta, corneal stroma, periosteum, adipose tissue, dental pulp and skeletal muscle. These solid tissue-derived stem cells are often used for tissue repair, disease modelling and new drug development. In the past two decades, stem cells have also been identified in various body fluids, including urine, peripheral blood, umbilical cord blood, amniotic fluid, synovial fluid, breastmilk and menstrual blood. These body fluid-derived stem cells (BFSCs) have stemness properties comparable to those of other adult stem cells and, similarly to tissue-derived stem cells, show cell surface markers, multi-differentiation potential and immunomodulatory effects. However, BFSCs are more easily accessible through non-invasive or minimally invasive approaches than solid tissue-derived stem cells and can be isolated without enzymatic tissue digestion. Additionally, BFSCs have shown good versatility in repairing genitourinary abnormalities in preclinical models through direct differentiation or paracrine mechanisms such as pro-angiogenic, anti-apoptotic, antifibrotic, anti-oxidant and anti-inflammatory effects. However, optimization of protocols is needed to improve the efficacy and safety of BFSC therapy before therapeutic translation.
Collapse
Affiliation(s)
- Ru-Lin Huang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qingfeng Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jian-Xing Ma
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Anthony Atala
- Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Yuanyuan Zhang
- Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA.
| |
Collapse
|
3
|
Dinc R. A review of the current state in neointimal hyperplasia development following endovascular intervention and minor emphasis on new horizons in immunotherapy. Transl Clin Pharmacol 2023; 31:191-201. [PMID: 38196998 PMCID: PMC10772059 DOI: 10.12793/tcp.2023.31.e18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 10/20/2023] [Accepted: 10/24/2023] [Indexed: 01/11/2024] Open
Abstract
Endovascular strategies play a vital role in the treatment of peripheral arterial disease (PAD). However, luminal loss or restenosis after endovascular intervention remains a significant challenge. The main underlying mechanisms are negative vascular remodeling and elastic recoil in balloon angioplasty. During stenting, the main reason for this complex is neointimal proliferation. Endothelial cell injury due to endovascular intervention initiates a series of molecular events, such as overexpression of growth factors, cytokine secretion, and adhesion molecules. These induce platelet activation and inflammatory processes, which trigger the proliferation and migration of vascular smooth muscle cells into the intima, resulting in neointimal hyperplasia. During this process, PAD progression is mainly caused by chronic inflammation, in which macrophages play a central role. Of the current strategies, drug release interventions aim to suppress restenosis using antiproliferative drugs, such as sirolimus and paclitaxel, during drug release. These drugs inhibit vascular reendothelialization and reduce late in-stent restenosis. For this reason, immunotherapy can be considered an important alternative. Interventions that polarize macrophages to the M2 subtype are particularly important, as they shape the immune response in an anti-inflammatory direction and contribute to tissue repair. However, there are several challenges to overcome, such as localizing antiproliferative or polarizing agents only to areas of vascular injury. This review discusses, based on the early study observations, immunotherapeutic approaches to prevent restenosis after endovascular intervention for the treatment of PAD.
Collapse
Affiliation(s)
- Rasit Dinc
- INVAMED Medical Innovation Institute, Ankara 06810, Turkey
| |
Collapse
|
4
|
Wang X, Zhou Y, Sun Q, Zhang Q, Zhou H, Zhang J, Du Y, Wang Y, Yuan K, Xu L, Zhang M, Yan D, Zeng L, Xu K, Sang W. Thymosin β4 exerts cytoprotective function and attenuates liver injury in murine hepatic sinusoidal obstruction syndrome after hematopoietic stem cell transplantation. Transplant Cell Ther 2023:S2666-6367(23)01292-7. [PMID: 37192732 DOI: 10.1016/j.jtct.2023.05.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 05/11/2023] [Accepted: 05/11/2023] [Indexed: 05/18/2023]
Abstract
Hepatic sinusoidal obstruction syndrome (HSOS) is one of the life-threatening complications that may occur after hematopoietic stem cell transplantation (HSCT). Hepatic sinusoidal endothelial cells (HSECs) injury and liver fibrosis are key mechanisms of HSOS. Thymosin β4 (Tβ4) is an active polypeptide that functions in a variety of pathological and physiological states such as inflammation regulation, anti-apoptosis and anti-fibrosis. In this study, we found that Tβ4 can stimulate HSECs proliferation, migration and tube formation in vitro via activation of pro-survival signaling AKT (protein kinase B). In addition, Tβ4 resisted γ irradiation-induced HSECs growth arrest and apoptosis in parallel with upregulation of anti-apoptotic protein B-cell lymphoma-extra-large (Bcl-xL) and B-cell lymphoma-2 (Bcl-2), which may be associated with activation of AKT. More importantly, Tβ4 significantly inhibited irradiation-induced proinflammatory cytokines in parallel with negative regulation of TLR4/MyD88/NF-κB and MAPK p38. Meanwhile, Tβ4 reduced intracellular reactive oxygen species production and upregulated antioxidants in HSECs. Additionally, Tβ4 inhibited irradiation-induced activation of hepatic stellate cells via downregulation expression of fibrogenic markers α-SMA, PAI-1 and TGF-β. In a murine HSOS model, levels of circulating alanine aminotransferase, aspartate aminotransferase, total bilirubin, and pro-inflammatory cytokines IL-6, IL-1β and TNF-α were significantly reduced after administration of Tβ4 peptide; further, Tβ4 treatment successfully ameliorated HSECs injury, inflammatory damage and fibrosis of murine liver. Taken together, Tβ4 stimulates proliferation and angiogenesis of HSECs, exerts cytoprotective effect and attenuates liver injury in murine HSOS model, which could be a potential strategy to prevent and treat HSOS after HSCT.
Collapse
Affiliation(s)
- Xiangmin Wang
- Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China; Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, China
| | - Yi Zhou
- Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China; Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, China
| | - Qian Sun
- Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China; Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, China
| | - Qing Zhang
- Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China; Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, China
| | - Hongyuan Zhou
- Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China; Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, China
| | - Jiaoli Zhang
- Department of Rehabilitation, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Yuwei Du
- Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China; Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, China
| | - Yuhan Wang
- Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China; Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, China
| | - Ke Yuan
- Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China; Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, China
| | - Linyan Xu
- Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China; Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, China
| | - Meng Zhang
- Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China; Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, China
| | - Dongmei Yan
- Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China; Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, China
| | - Lingyu Zeng
- Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China; Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, China.
| | - Kailin Xu
- Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China; Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, China.
| | - Wei Sang
- Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China; Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, China.
| |
Collapse
|
5
|
Wang W, Ye Y, Du Y, Xu Z, Yuan K, Wang Y, Adzraku SY, Li Y, Xu K, Qiao J, Ju W, Zeng L. EPC infusion ameliorates acute graft-versus-host disease-related endothelial injury after allogeneic bone marrow transplantation. Front Immunol 2022; 13:1019657. [PMID: 36591312 PMCID: PMC9795844 DOI: 10.3389/fimmu.2022.1019657] [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: 08/15/2022] [Accepted: 11/21/2022] [Indexed: 12/23/2022] Open
Abstract
Introduction Graft-versus-host disease (GVHD) damages vascular endothelium. Endothelial progenitor cell (EPC) can differentiate to endothelial cell and promote angiogenesis, but its role in endothelial damage in GVHD is unclear. Methods In this study, we intend to assess whether EPC infusion promotes the repair of endothelial injury in GVHD mouse model. Male BALB/c mice were randomly divided into 5 groups: control group, total body irradiation group (TBI group), allogeneic bone marrow transplantation group (Allo-BMT group), acute graft versus host disease group (GVHD group), EPC infusion group (GVHD+EPC group) followed by analysis of mice survival, acute GVHD (aGVHD) score, T cell infiltration by immunofluorescence, as well as continuity of vascular endothelium in liver. Results Compared with Allo-BMT group, the clinical and pathological score of aGVHD mice were higher. On day 21 after transplantation, a large number of mononuclear cell infiltrations were seen in the target tissues of aGVHD mice and mice died within 30 days. In addition, aGVHD group also presented increased subendothelial infiltration of CD3+ T cells in the liver, decreased VE-cadherin expression and elevated major histocompatibility complex (MHC) II molecule expression in the endothelium. Moreover, expression of MHC-II molecule increased in endothelial cell after irradiation injury and LPS stimulation, indicating abnormally activated endothelial cell with antigen-presenting function. Interestingly, infusion of EPC reduced the clinical and pathological score of aGVHD, decreased infiltration of mononuclear cells, improved survival as well as upregulated VE-cadherin and downregulated MHC-II molecule. Discussion EPC infusion can mobilize to affected endothelium to decrease the infiltration of T cells and pathological endothelial activation contributing to ameliorating the damage of endothelium. EPC infusion combined with bone marrow transplantation might be a perspective strategy for the prevention and treatment of aGVHD.
Collapse
Affiliation(s)
- Weiwei Wang
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
- Jiangsu Key Laboratory of Bone Marrow Stem Cells, Xuzhou, Jiangsu, China
| | - Yali Ye
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
- Jiangsu Key Laboratory of Bone Marrow Stem Cells, Xuzhou, Jiangsu, China
| | - Yuwei Du
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
- Jiangsu Key Laboratory of Bone Marrow Stem Cells, Xuzhou, Jiangsu, China
| | - Zhengqing Xu
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
- Jiangsu Key Laboratory of Bone Marrow Stem Cells, Xuzhou, Jiangsu, China
| | - Ke Yuan
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
- Jiangsu Key Laboratory of Bone Marrow Stem Cells, Xuzhou, Jiangsu, China
| | - Yizhou Wang
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
- Jiangsu Key Laboratory of Bone Marrow Stem Cells, Xuzhou, Jiangsu, China
| | - Seyram Yao Adzraku
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
- Jiangsu Key Laboratory of Bone Marrow Stem Cells, Xuzhou, Jiangsu, China
| | - Yue Li
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
- Jiangsu Key Laboratory of Bone Marrow Stem Cells, Xuzhou, Jiangsu, China
| | - Kailin Xu
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
- Jiangsu Key Laboratory of Bone Marrow Stem Cells, Xuzhou, Jiangsu, China
| | - Jianlin Qiao
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
- Jiangsu Key Laboratory of Bone Marrow Stem Cells, Xuzhou, Jiangsu, China
| | - Wen Ju
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
- Jiangsu Key Laboratory of Bone Marrow Stem Cells, Xuzhou, Jiangsu, China
| | - Lingyu Zeng
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
- Jiangsu Key Laboratory of Bone Marrow Stem Cells, Xuzhou, Jiangsu, China
| |
Collapse
|
6
|
Huang Y, Song C, He J, Li M. Research progress in endothelial cell injury and repair. Front Pharmacol 2022; 13:997272. [PMID: 36176426 PMCID: PMC9513221 DOI: 10.3389/fphar.2022.997272] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 08/24/2022] [Indexed: 11/30/2022] Open
Abstract
Endothelial cells, which are important metabolic and endocrine cells, play an important role in regulating vascular function. The occurrence and development of various cardiovascular and cerebrovascular diseases are associated with endothelial dysfunction. However, the underlying mechanism of vascular endothelial injury is not fully understood. It has been reported that the mechanism of endothelial injury mainly involves inflammation and oxidative stress. Moreover, endothelial progenitor cells are regarded as important contributors in repairing damaged endothelium. Multiple interventions (including chemical drugs and traditional Chinese medicines) exert endothelial protection by decreasing the release of inducing factors, suppressing inflammation and oxidative stress, and preventing endothelial cell senescence.
Collapse
Affiliation(s)
- Yongpan Huang
- Medicine School, Changsha Social Work College, Changsha, Hunan, China
| | - Chong Song
- Medicine School, Changsha Social Work College, Changsha, Hunan, China
| | - Jianbin He
- Department of Respiratory and Critical Care Medicine, The First People’s Hospital of Huaihua, Affiliated to University of South China, Huaihua, Hunan, China
- *Correspondence: Jianbin He, ; Min Li,
| | - Min Li
- Medicine School, Changsha Social Work College, Changsha, Hunan, China
- *Correspondence: Jianbin He, ; Min Li,
| |
Collapse
|
7
|
Virovic-Jukic L, Ljubas D, Stojsavljevic-Shapeski S, Ljubičić N, Filipec Kanizaj T, Mikolasevic I, Grgurevic I. Liver regeneration as treatment target for severe alcoholic hepatitis. World J Gastroenterol 2022; 28:4557-4573. [PMID: 36157937 PMCID: PMC9476880 DOI: 10.3748/wjg.v28.i32.4557] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 05/30/2022] [Accepted: 06/16/2022] [Indexed: 02/06/2023] Open
Abstract
Severe alcoholic hepatitis (AH) is a distinct entity in the spectrum of alcohol-related liver disease, with limited treatment options and high mortality. Supportive medical care with corticosteroids in selected patients is the only currently available treatment option, often with poor outcomes. Based on the insights into the pathogenetic mechanisms of AH, which are mostly obtained from animal studies, several new treatment options are being explored. Studies have implicated impaired and deranged liver regeneration processes as one of the culprit mechanisms and a potential therapeutic target. Acknowledging evidence for the beneficial effects of granulocyte colony-stimulating factor (G-CSF) on liver regeneration and immunomodulation in animal models, several human studies investigated its role in the treatment of advanced alcohol-related liver disease and AH. Contrary to the previously published studies suggesting benefits of G-CSF in the outcomes of patients with severe AH, these effects were not confirmed by a recently published multicenter randomized trial, suggesting that other options should rather be pursued. Stem cell transplantation represents another option for improving liver regeneration, but evidence for its efficacy in patients with severe AH and advanced alcohol-related liver disease is still very scarce and unconvincing, with established lack of efficacy in patients with compensated cirrhosis. In this review, we summarize the current knowledge on the pathogenesis and experimental therapies targeting liver regeneration. The lack of high-quality studies and evidence is a major obstacle in further treatment development. New insights into the pathogenesis of not only liver injury, but also liver regeneration processes are mandatory for the development of new treatment options. A reliable experimental model of the pathogenesis of AH and processes involved in liver recovery is still missing, and data obtained from animal studies are essential for future research.
Collapse
Affiliation(s)
- Lucija Virovic-Jukic
- Department of Gastroenterology and Hepatology, Sisters of Charity University Hospital Center, Zagreb 10000, Croatia
- Department of Internal Medicine, University of Zagreb School of Medicine, Zagreb 10000, Croatia
| | - Dominik Ljubas
- Department of Internal Medicine, University of Zagreb School of Medicine, Zagreb 10000, Croatia
| | - Sanja Stojsavljevic-Shapeski
- Department of Gastroenterology and Hepatology, Sisters of Charity University Hospital Center, Zagreb 10000, Croatia
| | - Neven Ljubičić
- Department of Gastroenterology and Hepatology, Sisters of Charity University Hospital Center, Zagreb 10000, Croatia
- Department of Internal Medicine, University of Zagreb School of Medicine, Zagreb 10000, Croatia
- Department of Internal Medicine, University of Zagreb School of Dental Medicine, Zagreb 10000, Croatia
| | - Tajana Filipec Kanizaj
- Department of Internal Medicine, University of Zagreb School of Medicine, Zagreb 10000, Croatia
- Department of Gastroenterology, Merkur University Hospital, Zagreb 10000, Croatia
| | - Ivana Mikolasevic
- Department of Gastroenterology, Rijeka University Hospital Center, Rijeka 51000, Croatia
- Department of Internal Medicine, University of Rijeka School of Medicine, Rijeka 10000, Croatia
| | - Ivica Grgurevic
- Department of Internal Medicine, University of Zagreb School of Medicine, Zagreb 10000, Croatia
- Department of Gastroenterology, Hepatology and Clinical Nutrition, Dubrava University Hospital, Zagreb 10000, Croatia
| |
Collapse
|
8
|
Yin F, Mao LC, Cai QQ, Jiang WH. Effect of Hepatocyte Growth Factor-Transfected Human Umbilical Cord Mesenchymal Stem Cells on Hepatic Stellate Cells by Regulating Transforming Growth Factor-β1/Smads Signaling Pathway. Stem Cells Dev 2021; 30:1070-1081. [PMID: 34514810 DOI: 10.1089/scd.2021.0136] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Studies have shown that human umbilical cord mesenchymal stem cells (hUCMSCs) could ameliorate liver fibrosis (LF) through inhibiting the activation of hepatic stellate cells (HSCs). However, the specific mechanisms have not been studied clearly. The purpose of this study was to explore the possible mechanism of hepatocyte growth factor (HGF)-transfected hUCMSCs in inhibiting the proliferation and activation of HSCs-T6. The upper and lower double-cell coculture system was established among HGF-hUCMSCs, LV5-NC-hUCMSCs, hUCMSCs, and HSCs-T6 in experimental groups; HSCs-T6 were cultured alone as control group. After coculturing for 1, 2, and 3 days, results showed that HGF-transfected hUCMSCs could decrease cell viability of HSCs-T6 and promote apoptosis; inhibit their activation and reduce the expression of Collagen I, Collagen III, TGF-β1, Smad2 and Smad3, which may be related to inhibiting the activation of TGF-β1/Smads signaling pathway. These findings suggested that HGF-transfected hUCMSCs may be used as an alternative and novel therapeutic approach for the treatment of LF.
Collapse
Affiliation(s)
- Fei Yin
- Department of Histology and Embryology, Basic Medical College of Jilin University, Changchun, Jilin Province, China
| | - Li-Cui Mao
- Department of Histology and Embryology, Basic Medical College of Jilin University, Changchun, Jilin Province, China
| | - Qi-Qi Cai
- Department of Histology and Embryology, Basic Medical College of Jilin University, Changchun, Jilin Province, China
| | - Wen-Hua Jiang
- Department of Histology and Embryology, Basic Medical College of Jilin University, Changchun, Jilin Province, China
| |
Collapse
|
9
|
Gao J, Pan X, Li G, Chatterjee E, Xiao J. Physical Exercise Protects Against Endothelial Dysfunction in Cardiovascular and Metabolic Diseases. J Cardiovasc Transl Res 2021; 15:604-620. [PMID: 34533746 PMCID: PMC8447895 DOI: 10.1007/s12265-021-10171-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 09/02/2021] [Indexed: 12/16/2022]
Abstract
Increasing evidence shows that endothelial cells play critical roles in maintaining vascular homeostasis, regulating vascular tone, inhibiting inflammatory response, suppressing lipid leakage, and preventing thrombosis. The damage or injury of endothelial cells induced by physical, chemical, and biological risk factors is a leading contributor to the development of mortal cardiovascular and cerebrovascular diseases. However, the underlying mechanism of endothelial injury remains to be elucidated. Notably, no drugs effectively targeting and mending injured vascular endothelial cells have been approved for clinical practice. There is an urgent need to understand pathways important for repairing injured vasculature that can be targeted with novel therapies. Exercise training-induced protection to endothelial injury has been well documented in clinical trials, and the underlying mechanism has been explored in animal models. This review mainly summarizes the protective effects of exercise on vascular endothelium and the recently identified potential therapeutic targets for endothelial dysfunction.
Collapse
Affiliation(s)
- Juan Gao
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong, 226011, China.,Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, 333 Nan Chen Road, Shanghai, 200444, China
| | - Xue Pan
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong, 226011, China.,Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, 333 Nan Chen Road, Shanghai, 200444, China
| | - Guoping Li
- Cardiovascular Division of the Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Emeli Chatterjee
- Cardiovascular Division of the Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Junjie Xiao
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong, 226011, China. .,Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, 333 Nan Chen Road, Shanghai, 200444, China.
| |
Collapse
|
10
|
Chen L, Zhang J, Liu X, Yao M, Zhang H. The protective effects of umbilical cord-derived endothelial colony forming cells on hepatic veno-occlusive disease. Cell Biol Int 2020; 44:2541-2552. [PMID: 32876991 DOI: 10.1002/cbin.11461] [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: 03/02/2020] [Revised: 08/28/2020] [Accepted: 08/30/2020] [Indexed: 11/08/2022]
Abstract
Hepatic veno-occlusive disease (HVOD) characterized by endothelial cell dysfunction is one of the serious complications after hematopoietic stem-cell transplantation or chemotherapeutic drug application. The mortality of HVOD patients with multiorgan dysfunction is as high as 80%. The primary aim of this study was to evaluate whether the infusion of human umbilical cord-derived endothelial colony forming cells (hUC-ECFCs) could mitigate HVOD injury and investigate the underlying mechanism. We found that the expression of chemokine C-X-C chemokine ligand 12 (CXCL12) was markedly increased in the livers of HVOD mice. Meanwhile, hUC-ECFCs infusion could significantly ameliorate liver injury in HVOD mice, which was accompanied by hUC-ECFCs recruitment in the liver, reduced liver pathological alterations, and decreased serum alanine aminotransferase and aspartate aminotransferase activity. Besides, CXCL12-induced migration in hUC-ECFCs was partly impeded by chemokine receptor type 7 (CXCR7) silence or CXCR4 blockage. In conclusion, our results demonstrated that hUC-ECFCs could mitigate HVOD through homing to the injured liver via the CXCL12-CXCR4/CXCR7 signaling pathway.
Collapse
Affiliation(s)
- Lulu Chen
- Department of Clinical Medicine, Graduate School, Jining Medical University, Jining, Shandong, China.,Department of Hematology, Affiliated Hospital of Jining Medical University, Jining, Shandong, China.,Institute of Blood and Marrow Transplantation, Jining Medical University, Jining, Shandong, China
| | - Jingjing Zhang
- Department of Hematology, Affiliated Hospital of Jining Medical University, Jining, Shandong, China.,Institute of Blood and Marrow Transplantation, Jining Medical University, Jining, Shandong, China
| | - Xin Liu
- Department of Clinical Medicine, Graduate School, Jining Medical University, Jining, Shandong, China.,Department of Hematology, Affiliated Hospital of Jining Medical University, Jining, Shandong, China.,Institute of Blood and Marrow Transplantation, Jining Medical University, Jining, Shandong, China
| | - Mingkang Yao
- Department of Clinical Medicine, Graduate School, Jining Medical University, Jining, Shandong, China.,Department of Hematology, Affiliated Hospital of Jining Medical University, Jining, Shandong, China.,Institute of Blood and Marrow Transplantation, Jining Medical University, Jining, Shandong, China
| | - Hao Zhang
- Department of Clinical Medicine, Graduate School, Jining Medical University, Jining, Shandong, China.,Department of Hematology, Affiliated Hospital of Jining Medical University, Jining, Shandong, China.,Institute of Blood and Marrow Transplantation, Jining Medical University, Jining, Shandong, China
| |
Collapse
|
11
|
Auvinen K, Lokka E, Mokkala E, Jäppinen N, Tyystjärvi S, Saine H, Peurla M, Shetty S, Elima K, Rantakari P, Salmi M. Fenestral diaphragms and PLVAP associations in liver sinusoidal endothelial cells are developmentally regulated. Sci Rep 2019; 9:15698. [PMID: 31666588 PMCID: PMC6821839 DOI: 10.1038/s41598-019-52068-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 10/11/2019] [Indexed: 12/30/2022] Open
Abstract
Endothelial cells contain several nanoscale domains such as caveolae, fenestrations and transendothelial channels, which regulate signaling and transendothelial permeability. These structures can be covered by filter-like diaphragms. A transmembrane PLVAP (plasmalemma vesicle associated protein) protein has been shown to be necessary for the formation of diaphragms. The expression, subcellular localization and fenestra-forming role of PLVAP in liver sinusoidal endothelial cells (LSEC) have remained controversial. Here we show that fenestrations in LSEC contain PLVAP-diaphragms during the fetal angiogenesis, but they lose the diaphragms at birth. Although it is thought that PLVAP only localizes to diaphragms, we found luminal localization of PLVAP in adult LSEC using several imaging techniques. Plvap-deficient mice revealed that the absence of PLVAP and diaphragms did not affect the morphology, the number of fenestrations or the overall vascular architecture in the liver sinusoids. Nevertheless, PLVAP in fetal LSEC (fenestrations with diaphragms) associated with LYVE-1 (lymphatic vessel endothelial hyaluronan receptor 1), neuropilin-1 and VEGFR2 (vascular endothelial growth factor receptor 2), whereas in the adult LSEC (fenestrations without diaphragms) these complexes disappeared. Collectively, our data show that PLVAP can be expressed on endothelial cells without diaphragms, contradict the prevailing concept that biogenesis of fenestrae would be PLVAP-dependent, and reveal previously unknown PLVAP-dependent molecular complexes in LSEC during angiogenesis.
Collapse
Affiliation(s)
- Kaisa Auvinen
- MediCity Research Laboratory, University of Turku, Turku, Finland.,Institute of Biomedicine, University of Turku, Turku, Finland
| | - Emmi Lokka
- MediCity Research Laboratory, University of Turku, Turku, Finland.,Institute of Biomedicine, University of Turku, Turku, Finland
| | - Elias Mokkala
- MediCity Research Laboratory, University of Turku, Turku, Finland.,Institute of Biomedicine, University of Turku, Turku, Finland
| | - Norma Jäppinen
- MediCity Research Laboratory, University of Turku, Turku, Finland.,Institute of Biomedicine, University of Turku, Turku, Finland
| | - Sofia Tyystjärvi
- MediCity Research Laboratory, University of Turku, Turku, Finland.,Institute of Biomedicine, University of Turku, Turku, Finland
| | - Heikki Saine
- MediCity Research Laboratory, University of Turku, Turku, Finland.,Institute of Biomedicine, University of Turku, Turku, Finland
| | - Markus Peurla
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - Shishir Shetty
- Centre for Liver Research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Kati Elima
- MediCity Research Laboratory, University of Turku, Turku, Finland.,Institute of Biomedicine, University of Turku, Turku, Finland
| | - Pia Rantakari
- MediCity Research Laboratory, University of Turku, Turku, Finland.,Institute of Biomedicine, University of Turku, Turku, Finland
| | - Marko Salmi
- MediCity Research Laboratory, University of Turku, Turku, Finland. .,Institute of Biomedicine, University of Turku, Turku, Finland.
| |
Collapse
|
12
|
Li M, Chen Y, Shi J, Ju W, Qi K, Fu C, Li Z, Zhang X, Qiao J, Xu K, Zeng L. NLRP6 deficiency aggravates liver injury after allogeneic hematopoietic stem cell transplantation. Int Immunopharmacol 2019; 74:105740. [PMID: 31301646 DOI: 10.1016/j.intimp.2019.105740] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Revised: 07/03/2019] [Accepted: 07/03/2019] [Indexed: 12/15/2022]
Abstract
This study aims to observe the expression and role of NLRP6 in liver injury after allogeneic hematopoietic stem cell transplantation (Allo-HSCT). Allo-HSCT model was established through infusion of 5 × 106 bone marrow mononuclear cells into whole body irradiated mice. On days 7, 14, 21 and 28 after transplantation, the peripheral blood was collected to detect liver function. The liver of the mice was obtained to assess the pathological changes of liver tissues after allo-HSCT by H&E staining and Mason staining. Meanwhile, expression of NLRP6, phosphorylated p38-MAPK and IκBα, caspase-1 and NLRP3 in liver were detected by Western blot. ELISA was used for detection of the level of interleukin (IL)-1β, IL-18, tumor necrosis factor (TNF)-α, IL-6, myeloperoxidase (MPO) and tumor growth factor (TGF)-β1. Increased expression of NLRP6, phosphorylated Iκbα, phosphorylated p38-MAPK, pro-caspase-1, and p20, in liver tissue with injury and fibrosis in mice after allo-HSCT were observed. Meanwhile, the level of IL-1β, IL-18, IL-6 and TNF-α was also increased. However, NLRP6-/- mice showed more severe liver damage and liver fibrosis after transplantation together with higher level of phosphorylated Iκbα, phosphorylated p38-MAPK, Pro-caspase-1, p20 expression as well as IL-1β, IL-18, IL-6, and TNF-α secretion compared with wide-type. Interestingly, the expression of NLRP3 in the liver of NLRP6-/- mice was significantly higher than that of wild-type. In conclusion, the expression of NLRP6 in host's liver is associated with liver injury after allo-HSCT. NLRP6 deficiency in host's liver leads to more severe liver damage, indicating a protective role of NLRP6 in host's liver to liver damage after allo-HSCT.
Collapse
Affiliation(s)
- Mingfeng Li
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou 221002, China; Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China; Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou 221002, China
| | - Yuting Chen
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou 221002, China; Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China; Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou 221002, China
| | - Jinrui Shi
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou 221002, China; Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China; Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou 221002, China
| | - Wen Ju
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou 221002, China; Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China; Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou 221002, China
| | - Kungming Qi
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China
| | - Chunling Fu
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou 221002, China; Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China; Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou 221002, China
| | - Zhenyu Li
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China
| | - Xi Zhang
- Department of Hematology, Xinqiao Hospital of Third Military Medical University, Chongqing 400037, China
| | - Jianlin Qiao
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou 221002, China; Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China; Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou 221002, China.
| | - Kailin Xu
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou 221002, China; Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China; Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou 221002, China.
| | - Lingyu Zeng
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou 221002, China; Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China; School of Medical Technology, Xuzhou Medical University, 221004, China.
| |
Collapse
|
13
|
Kumar A, Palek R, Liska V. A Critical Analysis of Experimental Animal Models of Sinusoidal Obstruction Syndrome. J Clin Exp Hepatol 2019; 9:345-353. [PMID: 31360027 PMCID: PMC6637067 DOI: 10.1016/j.jceh.2018.07.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 07/07/2018] [Indexed: 02/08/2023] Open
Abstract
Given the high mortality rate and clinical impact associated with sinusoidal obstruction syndrome (SOS), many studies have attempted to better characterize the disease and potential treatment strategies. However, the unpredictability of SOS onset represents a major obstacle when developing reproducible and controlled clinical trials in humans. Similarly, although in vitro studies have elucidated many of the molecular and cellular mechanisms of SOS, they often lack clinical relevance and translatability, highlighting the importance of experimental in vivo research. Animal models have greatly varied in the approach used to induce SOS in accordance with the numerous causes of human disease. Thus far, the most common and prevalent model is the monocrotaline-induced model in rats, which has served as the basis for both new diagnostic and treatment studies and has been revised over the last 20 years to optimize its use. Furthermore, radiotherapy, oxaliplatin-based chemotherapy, and even hematopoietic stem cell transplantation have been recently used to better replicate human SOS in animals. Nevertheless, because of the novelty of such research, further studies should be conducted to better understand the reproducibility and applicability of these newer models. Thus, this review seeks to summarize the methods and results of experimental in vivo models of SOS and compare the efficacy of these various adaptations.
Collapse
Key Words
- BM SPC, Bone Marrow Endothelial Progenitor Cell
- CRLM, Colorectal Liver Metastases
- CV, Central Vein
- HSCT, Hematopoietic Stem Cell Transplantation
- HVOD, Hepatic Veno-Occlusive Disease
- MCT, Monocrotaline
- MMP-9, Matrix Metalloproteinase-9
- NO, Nitric Oxide
- PA, Pyrrolizidine Alkaloid
- RILD, Radiation-Induced Liver Disease
- SEC, Sinusoidal Endothelial Cell
- SOS, Sinusoidal Obstruction Syndrome
- blue liver disease
- in vivo
- monocrotaline
- oxaliplatin
- veno-occlusive disease
Collapse
Affiliation(s)
- Arvind Kumar
- Biomedical Center, Charles University, Faculty of Medicine in Pilsen, Pilsen, Czech Republic,Department of Surgery, Charles University, Faculty of Medicine in Pilsen, Teaching Hospital Pilsen, Pilsen, Czech Republic
| | - Richard Palek
- Biomedical Center, Charles University, Faculty of Medicine in Pilsen, Pilsen, Czech Republic,Department of Surgery, Charles University, Faculty of Medicine in Pilsen, Teaching Hospital Pilsen, Pilsen, Czech Republic
| | - Vaclav Liska
- Biomedical Center, Charles University, Faculty of Medicine in Pilsen, Pilsen, Czech Republic,Department of Surgery, Charles University, Faculty of Medicine in Pilsen, Teaching Hospital Pilsen, Pilsen, Czech Republic,Address for correspondence: Vaclav Liska, Biomedical Center, Charles University, Faculty of Medicine in Pilsen, Alej Svobody 1655/76 323 00, Pilsen, Czech Republic.
| |
Collapse
|
14
|
Bian N, Du G, Ip MF, Ding J, Chang Q, Li Z. Pituitary adenylate cyclase-activating polypeptide attenuates tumor necrosis factor-α-induced apoptosis in endothelial colony-forming cells. Biomed Rep 2017; 7:11-16. [PMID: 28685053 PMCID: PMC5492523 DOI: 10.3892/br.2017.917] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 12/20/2016] [Indexed: 12/13/2022] Open
Abstract
Endothelial colony-forming cells (ECFCs) are important in angiogenesis and vascular proliferation. Tumor necrosis factor (TNF)-α is a significant risk factor for the development of atherosclerosis and a key proinflammatory cytokine known to induce apoptosis in endothelial cells. Pituitary adenylate cyclase-activating polypeptide (PACAP) is one of the members of the vasoactive intestinal peptide/secretin/growth hormone-releasing hormone/glucagon superfamily and exists in two biological active forms, PACAP 38 and PACAP 27. PACAP has been reported to help prevent endothelial apoptosis via an anti-inflammatory mechanism. However, to the best of our knowledge, the anti-apoptotic potential of PACAP has not been investigated in ECFCs. The aim of the present study was to demonstrate the efficacy of PACAP for decreasing TNF-α-induced apoptosis in ECFCs. The results indicated that PACAP exerts a cytoprotective effect on ECFCs exposed to TNF-α. Furthermore, PACAP partially rescues the proliferation potential of ECFCs inhibited by prolonged TNF-α exposure. These findings support an anti-inflammatory role for PACAP in circulation diseases.
Collapse
Affiliation(s)
- Ning Bian
- Department of Cardiology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510630, P.R. China
| | - Gang Du
- Department of Cardiology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510630, P.R. China
| | - Man Fai Ip
- Department of Cardiology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510630, P.R. China.,Department of Cardiology, Macau Kernel Earl General Hospital, Macau 820200, P.R. China
| | - Juan Ding
- Department of Cardiology, North Branch, People's Hospital of Xinjiang Uygur Autonomous Region, Ürümqi, Xinjiang Uyghur Autonomous Region 830054, P.R. China
| | - Qing Chang
- Department of Histology and Embryology, Medical College of Jinan University, Guangzhou, Guangdong 510632, P.R. China
| | - Zicheng Li
- Department of Cardiology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510630, P.R. China
| |
Collapse
|
15
|
Number and function of circulating endothelial progenitor cells in patients with primary Budd-Chiari syndrome. Clin Res Hepatol Gastroenterol 2017; 41:139-146. [PMID: 27863925 DOI: 10.1016/j.clinre.2016.10.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2016] [Revised: 09/12/2016] [Accepted: 10/18/2016] [Indexed: 02/04/2023]
Abstract
BACKGROUND AND AIM Primary Budd-Chiari syndrome (BCS) is associated with vascular endothelial injury. Circulating endothelial progenitor cells (EPCs) provide an endogenous mechanism to repair endothelial injury. This study investigated the levels and functionality of EPCs in patients with primary BCS. METHODS EPCs (CD34+/CD133+/KDR+) were quantified in 82 patients with primary BCS (inferior vena cava type: n=19; hepatic vein type: n=22; and mixed type: n=41), 10 cirrhosis controls (CC group) and 10 age-matched healthy controls (HC group), using flow cytometry. EPCs proliferation was detected by MTT assay, adhesion by adhesion activity assay, and migration capacity by Transwell assay. RESULTS EPCs levels were significantly lower in the BCS group (0.020±0.005%) than in the CC and HC groups (0.260±0.201%, 0.038±0.007%; P<0.001 for each). EPCs cultured in vitro from BCS and CC groups had, respectively, lower proliferation activity (0.20±0.04, 0.23±0.06 vs 0.58±0.07, each P<0.001), adhesion activity (15.8±1.7, 18.2±4.3 vs 35.0±2.5 cells/random microscopic field (RMF), each P<0.001) and migration activity (16.1±1.5, 16.7±3.0 vs 23.9±2.0 cells/RMF, each P<0.001) than in the HC group. EPCs functionality did not significantly differ between the BCS and CC groups. The numbers and functions of EPCs did not significantly differ among patients with inferior vena cava type, hepatic vein type and mixed type of BCS. CONCLUSION Patients with primary BCS had lower EPCs levels, with less proliferation, adhesion and migration activities. These findings suggest that lower levels of less functional EPCs may be associated with venous occlusion in primary BCS patients.
Collapse
|
16
|
Xuan J, Feng W, An ZT, Yang J, Xu HB, Li J, Zhao ZF, Wen W. Anti-TGFβ-1 receptor inhibitor mediates the efficacy of the human umbilical cord mesenchymal stem cells against liver fibrosis through TGFβ-1/Smad pathway. Mol Cell Biochem 2017; 429:113-122. [DOI: 10.1007/s11010-017-2940-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 01/16/2017] [Indexed: 12/15/2022]
|
17
|
Guffroy M, Falahatpisheh H, Biddle K, Kreeger J, Obert L, Walters K, Goldstein R, Boucher G, Coskran T, Reagan W, Sullivan D, Huang C, Sokolowski S, Giovanelli R, Gerber HP, Finkelstein M, Khan N. Liver Microvascular Injury and Thrombocytopenia of Antibody-Calicheamicin Conjugates in Cynomolgus Monkeys-Mechanism and Monitoring. Clin Cancer Res 2016; 23:1760-1770. [PMID: 27683177 DOI: 10.1158/1078-0432.ccr-16-0939] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Revised: 08/17/2016] [Accepted: 09/09/2016] [Indexed: 11/16/2022]
Abstract
Purpose: Adverse reactions reported in patients treated with antibody-calicheamicin conjugates such as gemtuzumab ozogamicin (Mylotarg) and inotuzumab ozogamicin include thrombocytopenia and sinusoidal obstruction syndrome (SOS). The objective of this experimental work was to investigate the mechanism for thrombocytopenia, characterize the liver injury, and identify potential safety biomarkers.Experimental Design: Cynomolgus monkeys were dosed intravenously at 6 mg/m2/dose once every 3 weeks with a nonbinding antibody-calicheamicin conjugate (PF-0259) containing the same linker-payload as gemtuzumab ozogamicin and inotuzumab ozogamicin. Monkeys were necropsied 48 hours after the first administration (day 3) or 3 weeks after the third administration (day 63).Results: PF-0259 induced acute thrombocytopenia (up to 86% platelet reduction) with nadirs on days 3 to 4. There was no indication of effects on megakaryocytes in bone marrow or activation of platelets in peripheral blood. Microscopic evaluation of liver from animals necropsied on day 3 demonstrated midzonal degeneration and loss of sinusoidal endothelial cells (SECs) associated with marked platelet accumulation in sinusoids. Liver histopathology on day 63 showed variable endothelial recovery and progression to a combination of sinusoidal capillarization and sinusoidal dilation/hepatocellular atrophy, consistent with early SOS. Among biomarkers evaluated, there were early and sustained increases in serum hyaluronic acid (HA) that correlated well with serum aspartate aminotransferase and liver microscopic changes, suggesting that HA may be a sensitive diagnostic marker of the liver microvascular injury.Conclusions: These data support the conclusion that target-independent damage to liver SECs may be responsible for acute thrombocytopenia (through platelet sequestration in liver sinusoids) and development of SOS. Clin Cancer Res; 23(7); 1760-70. ©2016 AACR.
Collapse
Affiliation(s)
- Magali Guffroy
- Drug Safety Research and Development, Pfizer Inc., Pearl River, New York.
| | - Hadi Falahatpisheh
- Drug Safety Research and Development, Pfizer Inc., Pearl River, New York
| | - Kathleen Biddle
- Drug Safety Research and Development, Pfizer Inc., Groton, Connecticut
| | - John Kreeger
- Drug Safety Research and Development, Pfizer Inc., Groton, Connecticut
| | - Leslie Obert
- Drug Safety Research and Development, Pfizer Inc., Groton, Connecticut
| | - Karen Walters
- Drug Safety Research and Development, Pfizer Inc., Groton, Connecticut
| | - Richard Goldstein
- Drug Safety Research and Development, Pfizer Inc., Groton, Connecticut
| | - Germaine Boucher
- Drug Safety Research and Development, Pfizer Inc., Groton, Connecticut
| | - Timothy Coskran
- Drug Safety Research and Development, Pfizer Inc., Groton, Connecticut
| | - William Reagan
- Drug Safety Research and Development, Pfizer Inc., Groton, Connecticut
| | - Danielle Sullivan
- Drug Safety Research and Development, Pfizer Inc., Groton, Connecticut
| | - Chunli Huang
- Drug Safety Research and Development, Pfizer Inc., Groton, Connecticut
| | - Sharon Sokolowski
- Drug Safety Research and Development, Pfizer Inc., Groton, Connecticut
| | | | - Hans-Peter Gerber
- Oncology-Rinat Research & Development, Pfizer Inc., Pearl River, New York
| | - Martin Finkelstein
- Drug Safety Research and Development, Pfizer Inc., Pearl River, New York
| | - Nasir Khan
- Drug Safety Research and Development, Pfizer Inc., Groton, Connecticut
| |
Collapse
|
18
|
Qiao J, Yao H, Xia Y, Chu P, Li M, Wu Y, Li W, Ding L, Qi K, Li D, Xu K, Zeng L. Long non-coding RNAs expression profiles in hepatocytes of mice after hematopoietic stem cell transplantation. IUBMB Life 2016; 68:232-41. [PMID: 26805554 DOI: 10.1002/iub.1479] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Accepted: 01/06/2016] [Indexed: 11/12/2022]
Abstract
Hepatic veno-occlusive disease (HVOD), one serious complication following hematopoietic stem cell transplantation (HSCT), is mainly initiated by the damage to sinusoidal endothelial cells and hepatocytes. Long non-coding RNAs (lncRNAs) play an important role in the proliferation of hepatocytes and liver regeneration. lncRNAs profile in hepatocytes post-HSCT remains unclear. The aim of this study is to evaluate the profile of lncRNAs in hepatocytes of mice after HSCT. Mice HSCT model was established through infusion of 5 × 10(6) bone marrow mononuclear cells. On day 7, 14 and 33 after HSCT, mice were sacrificed for analysis of liver pathology, function and index. Total RNA was extracted from hepatocytes of mice on day 14 for microarray analysis of the expression profiles of lncRNAs by Arraystar Mouse lncRNA Microarray v2.0. Obvious edema and spotty necrosis of hepatocytes with inflammatory cells infiltration were observed post-HSCT. Meanwhile, increased levels of alkaline phosphatase, aspartate transaminase, and total bilirubin, as well as elevated liver index were also found. 2,918 up-regulated and 1,911 down-regulated lncRNAs in hepatocytes were identified. Some of differentially expressed mRNAs had adjacent lncRNAs that were also significantly dysregulated, with the same dysregulation direction. T-cell receptor (up-regulation) and VEGF signaling pathway (down-regulation) were identified as one of the most enriched pathways. Dysregulated lncRNAs might be involved in hepatocytes damage after HSCT, suggesting targeting them might be a novel approach in amelioration of hepatocytes damage.
Collapse
Affiliation(s)
- Jianlin Qiao
- Blood Diseases Institute, Xuzhou Medical College, Xuzhou, China.,Key Laboratory of Bone Marrow Stem Cell, Xuzhou, Jiangsu Province, China.,Department of Hematology, The Affiliated Hospital of Xuzhou Medical College, Xuzhou, China
| | - Haina Yao
- Blood Diseases Institute, Xuzhou Medical College, Xuzhou, China.,Key Laboratory of Bone Marrow Stem Cell, Xuzhou, Jiangsu Province, China
| | - Yuan Xia
- Blood Diseases Institute, Xuzhou Medical College, Xuzhou, China.,Key Laboratory of Bone Marrow Stem Cell, Xuzhou, Jiangsu Province, China
| | - Peipei Chu
- Blood Diseases Institute, Xuzhou Medical College, Xuzhou, China.,Key Laboratory of Bone Marrow Stem Cell, Xuzhou, Jiangsu Province, China
| | - Mingfeng Li
- Blood Diseases Institute, Xuzhou Medical College, Xuzhou, China.,Key Laboratory of Bone Marrow Stem Cell, Xuzhou, Jiangsu Province, China
| | - Yulu Wu
- Blood Diseases Institute, Xuzhou Medical College, Xuzhou, China.,Key Laboratory of Bone Marrow Stem Cell, Xuzhou, Jiangsu Province, China
| | - Wen Li
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical College, Xuzhou, China
| | - Lan Ding
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical College, Xuzhou, China
| | - Kunming Qi
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical College, Xuzhou, China
| | - Depeng Li
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical College, Xuzhou, China
| | - Kailin Xu
- Blood Diseases Institute, Xuzhou Medical College, Xuzhou, China.,Key Laboratory of Bone Marrow Stem Cell, Xuzhou, Jiangsu Province, China.,Department of Hematology, The Affiliated Hospital of Xuzhou Medical College, Xuzhou, China
| | - Lingyu Zeng
- Blood Diseases Institute, Xuzhou Medical College, Xuzhou, China.,Key Laboratory of Bone Marrow Stem Cell, Xuzhou, Jiangsu Province, China.,Department of Hematology, The Affiliated Hospital of Xuzhou Medical College, Xuzhou, China
| |
Collapse
|
19
|
Qiao J, Huang Y, Xia Y, Chu P, Yao H, Xu L, Qi K, Liu Y, Xu K, Zeng L. Busulfan and cyclosphamide induce liver inflammation through NLRP3 activation in mice after hematopoietic stem cell transplantation. Sci Rep 2015; 5:17828. [PMID: 26635145 PMCID: PMC4669461 DOI: 10.1038/srep17828] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Accepted: 11/06/2015] [Indexed: 01/29/2023] Open
Abstract
The aim of this study was to evaluate the role of NLRP3 inflammasome on BU/CY-induced liver inflammation in mice after HSCT. HSCT mice model was established through infusion of 5 × 106 bone marrow mononuclear cells after conditioned with BU/CY. On day 7, 14, 21 and 28 after HSCT, mice were sacrificed for analysis of liver inflammation, cytokine secretion, NLRP3 expression and caspase-1 activation as well as release of ATP and high-mobility group protein B1 (HMGB1). Furthermore, NLRP3 selective inhibitor (BAY 11-7082) was administrated into mice after HSCT to evaluate its effects on liver inflammation. Severe liver inflammation and damage with elevated secretion of IL-1β and IL-18 were found in mice after HSCT. Meanwhile, elevated expressions of NLRP3 and caspase-1 activation in liver were found. In addition, increased release of ATP and HMGB1 were observed. Selective inhibition of NLRP3 decreased caspase-1 activation and secretion of IL-1β and IL-18. Furthermore, NLRP3 inhibition also reduced infiltration of macrophages and neutrophils and improved liver function. In conclusion, NLRP3 was involved in BU/CY-induced liver inflammation after HSCT and selectively inhibited it ameliorated liver inflammation and improved liver function, suggesting targeting NLRP3 might be a new approach in the prophylaxis of liver inflammation after HSCT.
Collapse
Affiliation(s)
- Jianlin Qiao
- Department of Hematology, the Affiliated Hospital of Xuzhou Medical College, Xuzhou 221002, China.,Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou 221002, China
| | - Yujin Huang
- Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou 221002, China
| | - Yuan Xia
- Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou 221002, China
| | - Peipei Chu
- Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou 221002, China
| | - Haina Yao
- Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou 221002, China
| | - Linyan Xu
- Department of Hematology, the Affiliated Hospital of Xuzhou Medical College, Xuzhou 221002, China.,Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou 221002, China
| | - Kunming Qi
- Department of Hematology, the Affiliated Hospital of Xuzhou Medical College, Xuzhou 221002, China
| | - Yun Liu
- Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou 221002, China
| | - Kailin Xu
- Department of Hematology, the Affiliated Hospital of Xuzhou Medical College, Xuzhou 221002, China.,Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou 221002, China
| | - Lingyu Zeng
- Department of Hematology, the Affiliated Hospital of Xuzhou Medical College, Xuzhou 221002, China.,Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou 221002, China
| |
Collapse
|