1
|
Majumder S, Moriarty KL, Lee Y, Crombleholme TM. Placental Gene Therapy for Fetal Growth Restriction and Preeclampsia: Preclinical Studies and Prospects for Clinical Application. J Clin Med 2024; 13:5647. [PMID: 39337133 PMCID: PMC11432969 DOI: 10.3390/jcm13185647] [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: 07/30/2024] [Revised: 09/19/2024] [Accepted: 09/20/2024] [Indexed: 09/30/2024] Open
Abstract
In the last three decades, gene therapy has demonstrated significant progress. Over 700 active investigational new drug (IND) applications have been reported. Research on in utero gene therapy has advanced, but ethical and safety concerns persist. A novel approach under investigation is placental gene therapy, which holds promise for targeting diseases associated with placental dysfunction, such as fetal growth restriction (FGR) and preeclampsia. One of the underlying causes of placental insufficiency in these conditions is reduced placental growth factor-driven angiogenesis and endothelial cell dysfunction during fetal development. Studies have explored the overexpression of growth factor transgenes like IGF-1 to address FGR, yielding promising outcomes in animal models. Furthermore, intra-placental gene transfer, instead of systemic delivery of gene therapy vectors, has the potential to treat and cure these disorders. However, challenges and limitations akin to in utero gene therapy persist, including the risk of in utero infection, potential impairment of the mother's future fertility, the risk of germline integration, and possible off-target effects of gene transfer in the fetus or the mother. Consequently, additional research and deliberation within the scientific and medical communities are warranted to fully comprehend the potential benefits and risks of placental gene therapy.
Collapse
Affiliation(s)
- Sanjukta Majumder
- Molecular Fetal Therapy Laboratory, Fetal Care Center at Connecticut Children's Medical Center, Suite F254, 282 Washington Street, Hartford, CT 06106, USA
- Fetal Surgery Section, Division of Pediatric General and Thoracic Surgery, Department of Surgery, UConn Health, Farmington, CT 06030, USA
| | - Kristen Lee Moriarty
- Molecular Fetal Therapy Laboratory, Fetal Care Center at Connecticut Children's Medical Center, Suite F254, 282 Washington Street, Hartford, CT 06106, USA
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Farmington, CT 06030, USA
| | - Youngmok Lee
- Department of Pediatrics, School of Medicine, University of Connecticut, Farmington, CT 06030, USA
| | - Timothy M Crombleholme
- Molecular Fetal Therapy Laboratory, Fetal Care Center at Connecticut Children's Medical Center, Suite F254, 282 Washington Street, Hartford, CT 06106, USA
- Fetal Surgery Section, Division of Pediatric General and Thoracic Surgery, Department of Surgery, UConn Health, Farmington, CT 06030, USA
- Department of Pediatrics, School of Medicine, University of Connecticut, Farmington, CT 06030, USA
| |
Collapse
|
2
|
Larionov A, Hammer CM, Fiedler K, Filgueira L. Dynamics of Endothelial Cell Diversity and Plasticity in Health and Disease. Cells 2024; 13:1276. [PMID: 39120307 PMCID: PMC11312403 DOI: 10.3390/cells13151276] [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: 06/27/2024] [Revised: 07/19/2024] [Accepted: 07/19/2024] [Indexed: 08/10/2024] Open
Abstract
Endothelial cells (ECs) are vital structural units of the cardiovascular system possessing two principal distinctive properties: heterogeneity and plasticity. Endothelial heterogeneity is defined by differences in tissue-specific endothelial phenotypes and their high predisposition to modification along the length of the vascular bed. This aspect of heterogeneity is closely associated with plasticity, the ability of ECs to adapt to environmental cues through the mobilization of genetic, molecular, and structural alterations. The specific endothelial cytoarchitectonics facilitate a quick structural cell reorganization and, furthermore, easy adaptation to the extrinsic and intrinsic environmental stimuli, known as the epigenetic landscape. ECs, as universally distributed and ubiquitous cells of the human body, play a role that extends far beyond their structural function in the cardiovascular system. They play a crucial role in terms of barrier function, cell-to-cell communication, and a myriad of physiological and pathologic processes. These include development, ontogenesis, disease initiation, and progression, as well as growth, regeneration, and repair. Despite substantial progress in the understanding of endothelial cell biology, the role of ECs in healthy conditions and pathologies remains a fascinating area of exploration. This review aims to summarize knowledge and concepts in endothelial biology. It focuses on the development and functional characteristics of endothelial cells in health and pathological conditions, with a particular emphasis on endothelial phenotypic and functional heterogeneity.
Collapse
Affiliation(s)
- Alexey Larionov
- Faculty of Science and Medicine, Anatomy, University of Fribourg, Route Albert-Gockel 1, CH-1700 Fribourg, Switzerland; (C.M.H.); (L.F.)
| | - Christian Manfred Hammer
- Faculty of Science and Medicine, Anatomy, University of Fribourg, Route Albert-Gockel 1, CH-1700 Fribourg, Switzerland; (C.M.H.); (L.F.)
| | - Klaus Fiedler
- Independent Researcher, CH-1700 Fribourg, Switzerland;
| | - Luis Filgueira
- Faculty of Science and Medicine, Anatomy, University of Fribourg, Route Albert-Gockel 1, CH-1700 Fribourg, Switzerland; (C.M.H.); (L.F.)
| |
Collapse
|
3
|
Kwon M, Kim BS, Yoon S, Oh SO, Lee D. Hematopoietic Stem Cells and Their Niche in Bone Marrow. Int J Mol Sci 2024; 25:6837. [PMID: 38999948 PMCID: PMC11241602 DOI: 10.3390/ijms25136837] [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: 05/09/2024] [Revised: 06/18/2024] [Accepted: 06/19/2024] [Indexed: 07/14/2024] Open
Abstract
Extensive research has explored the functional correlation between stem cells and progenitor cells, particularly in blood. Hematopoietic stem cells (HSCs) can self-renew and regenerate tissues within the bone marrow, while stromal cells regulate tissue function. Recent studies have validated the role of mammalian stem cells within specific environments, providing initial empirical proof of this functional phenomenon. The interaction between bone and blood has always been vital to the function of the human body. It was initially proposed that during evolution, mammalian stem cells formed a complex relationship with the surrounding microenvironment, known as the niche. Researchers are currently debating the significance of molecular-level data to identify individual stromal cell types due to incomplete stromal cell mapping. Obtaining these data can help determine the specific activities of HSCs in bone marrow. This review summarizes key topics from previous studies on HSCs and their environment, discussing current and developing concepts related to HSCs and their niche in the bone marrow.
Collapse
Affiliation(s)
- Munju Kwon
- Department of Convergence Medicine, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea
| | - Byoung Soo Kim
- School of Biomedical Convergence Engineering, Pusan National University, Yangsan 50612, Republic of Korea
| | - Sik Yoon
- Department of Anatomy, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea
| | - Sae-Ock Oh
- Department of Anatomy, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea
| | - Dongjun Lee
- Department of Convergence Medicine, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea
- Transplantation Research Center, Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Republic of Korea
| |
Collapse
|
4
|
Liu J, Wang C, Qiu S, Sun W, Yang G, Yuan L. Toward Ultrasound Molecular Imaging of Endothelial Dysfunction in Diabetes: Targets, Strategies, and Challenges. ACS APPLIED BIO MATERIALS 2024; 7:1416-1428. [PMID: 38391247 DOI: 10.1021/acsabm.4c00053] [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] [Indexed: 02/24/2024]
Abstract
Diabetes vasculopathy is a significant complication of diabetes mellitus (DM), and early identification and timely intervention can effectively slow the progression. Accumulating studies have shown that diabetes causes vascular complications directly or indirectly through a variety of mechanisms. Direct imaging of the endothelial molecular changes not only identifies the early stage of diabetes vasculopathy but also sheds light on the precise treatment. Targeted ultrasound contrast agent (UCA)-based ultrasound molecular imaging (UMI) can noninvasively detect the expression status of molecular biomarkers overexpressed in the vasculature, thereby being a potential strategy for the diagnosis and treatment response evaluation of DM. Amounts of efforts have been focused on identification of the molecular targets expressed in the vasculature, manufacturing strategies of the targeted UCA, and the clinical translation for the diagnosis and evaluation of therapeutic efficacy in both micro- and macrovasculopathy in DM. This review summarizes the latest research progress on endothelium-targeted UCA and discusses their promising future and challenges in diabetes vasculopathy theranostics.
Collapse
Affiliation(s)
- Jiahan Liu
- Department of Ultrasound Medicine, Tangdu Hospital, Fourth Military Medical University, Shaanxi 710038, China
| | - Chen Wang
- Department of Ultrasound Medicine, Tangdu Hospital, Fourth Military Medical University, Shaanxi 710038, China
| | - Shuo Qiu
- Department of Ultrasound Medicine, Tangdu Hospital, Fourth Military Medical University, Shaanxi 710038, China
| | - Wenqi Sun
- Department of Ultrasound Medicine, Tangdu Hospital, Fourth Military Medical University, Shaanxi 710038, China
| | - Guodong Yang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Biochemistry and Molecular Biology, Fourth Military Medical University Xi'an, Shaanxi 710032, China
| | - Lijun Yuan
- Department of Ultrasound Medicine, Tangdu Hospital, Fourth Military Medical University, Shaanxi 710038, China
| |
Collapse
|
5
|
Tang SY, Lee YC, Tseng CW, Huang PH, Kuo KL, Tarng DC. Granulocyte Colony-Stimulating Factor Improves Endothelial Progenitor Cell-Mediated Neovascularization in Mice with Chronic Kidney Disease. Pharmaceutics 2023; 15:2380. [PMID: 37896140 PMCID: PMC10610103 DOI: 10.3390/pharmaceutics15102380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 09/13/2023] [Accepted: 09/20/2023] [Indexed: 10/29/2023] Open
Abstract
Patients with chronic kidney disease (CKD) have a higher prevalence of peripheral arterial disease (PAD), and endothelial progenitor cells (EPCs) play a pivotal role. We examined the impact of granulocyte colony-stimulating factor (G-CSF) on EPC function in response to tissue ischemia. Eight-week-old male C57BL/6J male mice were divided into sham operation and subtotal nephrectomy (SNx) groups, received hindlimb ischemic operation after seven weeks, then randomly received G-CSF or PBS intervention for four weeks with weekly follow-ups. SNx mice had significantly reduced limb reperfusion, decreased plasma EPC mobilization, and impaired angiogenesis in ischemic hindlimbs compared to the control group. However, G-CSF increased IL-10 and reversed these adverse changes. Additionally, ischemia-associated protein expressions, including IL-10, phospho-STAT3, VEGF, and phospho-eNOS, were significantly downregulated in the ischemic hindlimbs of SNx mice versus control, but these trends were reversed by G-CSF. Furthermore, in cultured EPCs, G-CSF significantly attenuated the decrease in EPC function initiated by indoxyl sulfate through IL-10. Overall, we discovered that G-CSF can improve EPC angiogenic function through a hypoxia/IL-10 signaling cascade and impede neovascular growth in response to ischemia of SNx mice. Our results highlight G-CSF's potential to restore angiogenesis in CKD patients with PAD via EPC-based methods.
Collapse
Affiliation(s)
- Shao-Yu Tang
- Department of Medical Education, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 23142, Taiwan;
| | - Yi-Chin Lee
- Department of Physiology, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan;
| | - Chien-Wei Tseng
- Department of Chinese Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 23142, Taiwan;
- School of Post-Baccalaureate Chinese Medicine, Tzu Chi University, Hualien 97004, Taiwan
| | - Po-Hsun Huang
- Institute of Clinical Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan;
- Cardiovascular Research Center, School of Medicine, National Yang Ming Chiao Tung, Taipei 11221, Taiwan
- Divisions of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei 11217, Taiwan
| | - Ko-Lin Kuo
- School of Post-Baccalaureate Chinese Medicine, Tzu Chi University, Hualien 97004, Taiwan
- Division of Nephrology, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 23142, Taiwan
- School of Medicine, Tzu Chi University, Hualien 97004, Taiwan
| | - Der-Cherng Tarng
- Institute of Clinical Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan;
- Division of Nephrology, Department of Medicine, Taipei Veterans General Hospital, Taipei 11217, Taiwan
| |
Collapse
|
6
|
Zhao ZA, Yan L, Wen J, Satyanarayanan SK, Yu F, Lu J, Liu YU, Su H. Cellular and molecular mechanisms in vascular repair after traumatic brain injury: a narrative review. BURNS & TRAUMA 2023; 11:tkad033. [PMID: 37675267 PMCID: PMC10478165 DOI: 10.1093/burnst/tkad033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 02/01/2023] [Accepted: 05/26/2023] [Indexed: 09/08/2023]
Abstract
Traumatic brain injury (TBI) disrupts normal brain function and is associated with high morbidity and fatality rates. TBI is characterized as mild, moderate or severe depending on its severity. The damage may be transient and limited to the dura matter, with only subtle changes in cerebral parenchyma, or life-threatening with obvious focal contusions, hematomas and edema. Blood vessels are often injured in TBI. Even in mild TBI, dysfunctional cerebral vascular repair may result in prolonged symptoms and poor outcomes. Various distinct types of cells participate in vascular repair after TBI. A better understanding of the cellular response and function in vascular repair can facilitate the development of new therapeutic strategies. In this review, we analyzed the mechanism of cerebrovascular impairment and the repercussions following various forms of TBI. We then discussed the role of distinct cell types in the repair of meningeal and parenchyma vasculature following TBI, including endothelial cells, endothelial progenitor cells, pericytes, glial cells (astrocytes and microglia), neurons, myeloid cells (macrophages and monocytes) and meningeal lymphatic endothelial cells. Finally, possible treatment techniques targeting these unique cell types for vascular repair after TBI are discussed.
Collapse
Affiliation(s)
- Zi-Ai Zhao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macau 999078, China
- Department of Neurology, General Hospital of Northern Theater Command, 83# Wen-Hua Road, Shenyang 110840, China
| | - Lingli Yan
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macau 999078, China
| | - Jing Wen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macau 999078, China
| | - Senthil Kumaran Satyanarayanan
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macau 999078, China
| | - Feng Yu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macau 999078, China
| | - Jiahong Lu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macau 999078, China
| | - Yong U Liu
- Laboratory of Neuroimmunology in Health and Disease Institute, Guangzhou First People’s Hospital School of Medicine, South China University of Technology, Guangzhou Higher Education Mega Center, Panyu District, Guangzhou 511400, China
| | - Huanxing Su
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macau 999078, China
| |
Collapse
|
7
|
Okita R, Kawamoto N, Okada M, Inokawa H, Yamamoto N, Murakami T, Ikeda E. Preoperative neutrophil-to-lymphocyte ratio correlates with PD-L1 expression in immune cells of patients with malignant pleural mesothelioma and predicts prognosis. Sci Rep 2023; 13:5263. [PMID: 37002247 PMCID: PMC10066199 DOI: 10.1038/s41598-023-31448-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 03/11/2023] [Indexed: 04/03/2023] Open
Abstract
We assessed the prognostic value of five complex inflammatory and nutritional parameters, namely neutrophil-to-lymphocyte ratio (NLR), prognostic nutritional index (PNI), C-reactive protein-to-NLR ratio (C/NLR), platelet-to-lymphocyte ratio (PLR), and lymphocyte-to-monocyte ratio (LMR) using data from patients with malignant pleural mesothelioma (MPM) undergoing extrapleural pneumonectomy (EPP). Moreover, the correlation between these five parameters and programmed cell death protein 1 ligand-1 (PD-L1) expression in the tumor microenvironment was evaluated. This study included consecutive MPM patients who underwent EPP. The histological subtype of the eligible patients (n = 61) correlated with all five parameters. Moreover, the PD-L1 expression scores for immune cells correlated with NLR and PLR, and the PD-L1 expression scores for both tumor cells and immune cells were inversely correlated with both PNI and LMR. Univariate analysis elucidated that NLR, PNI, and C/NLR were predictors of 5-year overall survival (OS), and multivariate analysis revealed that NLR was an independent predictor of 5-year OS, suggesting that NLR is a preoperative, prognostic factor for patients with MPM who are scheduled for EPP. To the best of our knowledge, this is the first study to evaluate the prognostic potentials of NLR, PNI, C/NLR, PLR, and LMR simultaneously in patients with MPM who underwent EPP.
Collapse
Affiliation(s)
- Riki Okita
- Department of Thoracic Surgery, National Hospital Organization Yamaguchi Ube Medical Center, 685 Higashikiwa, Ube Yamaguchi, 755-0241, Japan.
| | - Nobutaka Kawamoto
- Department of Thoracic Surgery, National Hospital Organization Yamaguchi Ube Medical Center, 685 Higashikiwa, Ube Yamaguchi, 755-0241, Japan
| | - Masanori Okada
- Department of Thoracic Surgery, National Hospital Organization Yamaguchi Ube Medical Center, 685 Higashikiwa, Ube Yamaguchi, 755-0241, Japan
| | - Hidetoshi Inokawa
- Department of Thoracic Surgery, National Hospital Organization Yamaguchi Ube Medical Center, 685 Higashikiwa, Ube Yamaguchi, 755-0241, Japan
| | - Naoki Yamamoto
- Health Administration Center, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Tomoyuki Murakami
- Department Clinical Research, National Hospital Organization Yamaguchi Ube Medical Center, 685 Higashikiwa, Ube Yamaguchi, 755-0241, Japan
- Department of Pathology, KYURIN/ KYURIN PACELL Corporation, 26-67 Morishita-Cho, Yahatanishi-Ku, Kitakyushu, Fukuoka, 806-0046, Japan
| | - Eiji Ikeda
- Department Clinical Research, National Hospital Organization Yamaguchi Ube Medical Center, 685 Higashikiwa, Ube Yamaguchi, 755-0241, Japan
- Department of Pathology, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| |
Collapse
|
8
|
Simulated microgravity affects stroma-dependent ex vivo myelopoiesis. Tissue Cell 2023; 80:101987. [PMID: 36481580 DOI: 10.1016/j.tice.2022.101987] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 11/15/2022] [Accepted: 11/23/2022] [Indexed: 11/25/2022]
Abstract
Microgravity is known negatively affect physiology of living beings, including hematopoiesis. Dysregulation of hematopoietic cells and supporting stroma relationships in bone marrow niche may be in charge. We compared the efficacy of ex vivo expansion of hematopoietic stem and progenitor cells (HSPCs) in presence of native or osteocommitted MSCs under simulated microgravity (Smg) using Random Positioning Machine (RPM). In comparison with 1 g, a decrease of MSC-associated HSPCs and an increase of floating HSPCs was observed after 7 days of Smg exposure. Among floating HSPCs, primitive progenitors were presented by late CD34+/133-. Total CFUs as well as erythroid (BFU-E) and granulocytic (CFU-G) numbers were lower. MSC-associated primitive HSPCs demonstrated increased proportion of late CD34+/133- in expense of early CD34-/133+. Osteo-MSCs preferentially supported late primitive CD34+ and more committed HSPCs as followed from increase of CFUs, and CD235a+ erythroid progenitors. Under Smg, an increased VEGF, eotaxin, and GRO-a levels, and a decrease in RANTES were found in the osteo-MSC-HSPC co-cultures. IL-6,-8, -13, G-CSF, GRO-a, MCP-3, MIP-1b, VEGF increased in co-culture with osteo-MSCs vs intact MSCs. Based on the findings, the misbalance between primitive/committed HSPCs and a decrease in hematopoiesis-supportive activity of osteocommitted cells are supposed to underline hematopoietic disorders during space flights.
Collapse
|
9
|
Li T, Ma J, Wang W, Lei B. Bioactive MXene Promoting Angiogenesis and Skeletal Muscle Regeneration through Regulating M2 Polarization and Oxidation Stress. Adv Healthc Mater 2023; 12:e2201862. [PMID: 36427290 DOI: 10.1002/adhm.202201862] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 11/04/2022] [Indexed: 11/26/2022]
Abstract
Complete repair of skeletal muscles caused by severe mechanical damage and muscle-related diseases remains a challenge. 2D Ti3 C2 Tx (MXene) possesses special photoelectromagnetic properties and has attracted considerable attention in materials science and engineering. However, the bioactive properties and potential mechanism of MXene in tissue engineering, especially in skeletal muscle regeneration, are unclear. Herein, the antioxidation and anti-inflammation activities of MXene and its effects on myogenic differentiation and regeneration of skeletal muscle in vivo are investigated. In vitro studies have shown that MXene has excellent antioxidation and anti-inflammatory properties, and promotes myogenic differentiation and angiogenesis. MXene can remove excess reactive oxygen species in macrophage cells to alleviate oxidative stress and induce the transformation of M1 macrophages into M2 macrophages to reduce excessive inflammation, which can significantly promote the proliferation and differentiation of myoblasts, as well as the proliferation, migration, and tube formation of endothelial cells. Animal experiments with rat tibial anterior muscle defects show that MXene can promote angiogenesis, muscle fiber formation, and skeletal muscle regeneration by regulating the cell microenvironment through anti-inflammatory and antioxidant pathways. The findings suggest that MXene can be used as a multifunctional bioactive material to enhance tissue regeneration through robust antioxidation, anti-inflammation, and angiogenesis activities.
Collapse
Affiliation(s)
- Ting Li
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, 710049, China.,Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, 710054, China
| | - Junping Ma
- Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, 710054, China
| | - Wensi Wang
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Bo Lei
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, 710049, China.,Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, 710054, China.,State Key Laboratory for Mechanical Behavior of Materials, Instrument Analysis Center, Xi'an Jiaotong University, Xi'an, 710054, China.,Instrumental Analysis Center, Xi'an Jiaotong University, Xi'an, 710049, China
| |
Collapse
|
10
|
The Role of Membrane-Type 1 Matrix Metalloproteinase-Substrate Interactions in Pathogenesis. Int J Mol Sci 2023; 24:ijms24032183. [PMID: 36768503 PMCID: PMC9917210 DOI: 10.3390/ijms24032183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/17/2023] [Accepted: 01/20/2023] [Indexed: 01/25/2023] Open
Abstract
A protease is an enzyme with a proteolytic activity that facilitates the digestion of its substrates. Membrane-type I matrix metalloproteinase (MT1-MMP), a member of the broader matrix metalloproteinases (MMP) family, is involved in the regulation of diverse cellular activities. MT1-MMP is a very well-known enzyme as an activator of pro-MMP-2 and two collagenases, MMP-8 and MMP-13, all of which are essential for cell migration. As an anchored membrane enzyme, MT1-MMP has the ability to interact with a diverse group of molecules, including proteins that are not part of the extracellular matrix (ECM). Therefore, MT1-MMP can regulate various cellular activities not only by changing the extra-cellular environment but also by regulating cell signaling. The presence of both intracellular and extra-cellular portions of MT1-MMP can allow it to interact with proteins on both sides of the cell membrane. Here, we reviewed the MT1-MMP substrates involved in disease pathogenesis.
Collapse
|
11
|
Wu C, Li X, Zhao H, Ling Y, Ying Y, He Y, Zhang S, Liang S, Wei J, Gan X. Resistance exercise promotes the resolution and recanalization of deep venous thrombosis in a mouse model via SIRT1 upregulation. BMC Cardiovasc Disord 2023; 23:18. [PMID: 36639616 PMCID: PMC9837998 DOI: 10.1186/s12872-022-02908-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 10/19/2022] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Early exercise for acute deep venous thrombosis (DVT) improves the patient's symptoms and does not increase the risk of pulmonary embolism. However, information about its effect on thrombus resolution is limited. The aim of this study was to investigate the role of resistance exercise (RE) in thrombus resolution and recanalization and determine its underlying mechanisms. METHODS: Ninety-six C57BL/6 J mice were randomly divided into four groups: Control group (C, n = 24); DVT group (D, n = 24); RE + DVT group (ED, n = 24); and inhibitor + RE + DVT group (IED, n = 24). A DVT model was induced by stenosis of the inferior vena cava (IVC). After undergoing IVC ultrasound within 24 h post-operation to confirm DVT formation, mice without thrombosis were excluded. Other mice were sacrificed and specimens were obtained 14 or 28 days after operation. Thrombus-containing IVC was weighed, and the thrombus area and recanalization rate were calculated using HE staining. Masson's trichrome staining was used to analyze the collagen content. RT-PCR and ELISA were performed to examine IL-6, TNF-α, IL-10, and VEGF expression levels. SIRT1 expression was assessed using immunohistochemistry staining and RT-PCR. VEGF-A protein expression and CD-31-positive microvascular density (MVD) in the thrombus were observed using immunohistochemistry. RESULTS: RE did not increase the incidence of pulmonary embolism. It reduced the weight and size of the thrombus and the collagen content. Conversely, it increased the recanalization rate. It also decreased the levels of the pro-inflammatory factors IL-6 and TNF-α and increased the expression levels of the anti-inflammatory factor IL-10. RE enhanced VEGF and SIRT1 expression levels and increased the MVD in the thrombosis area. After EX527 (SIRT1 inhibitor) was applied, the positive effects of exercise were suppressed. CONCLUSIONS RE can inhibit inflammatory responses, reduce collagen deposition, and increase angiogenesis in DVT mice, thereby promoting thrombus resolution and recanalization. Its underlying mechanism may be associated with the upregulation of SIRT1 expression.
Collapse
Affiliation(s)
- Caijiao Wu
- grid.412594.f0000 0004 1757 2961Department of Nursing, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021 Guangxi China
| | - Xiaorong Li
- grid.412594.f0000 0004 1757 2961Department of Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi China
| | - Huihan Zhao
- grid.412594.f0000 0004 1757 2961Department of Nursing, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021 Guangxi China
| | - Ying Ling
- grid.412594.f0000 0004 1757 2961Department of Nursing, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021 Guangxi China
| | - Yanping Ying
- grid.412594.f0000 0004 1757 2961Department of Nursing, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021 Guangxi China
| | - Yu He
- grid.412594.f0000 0004 1757 2961Medical Lab, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021 Guangxi China
| | - Shaohan Zhang
- grid.412594.f0000 0004 1757 2961Department of Nursing, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021 Guangxi China
| | - Shijing Liang
- grid.412594.f0000 0004 1757 2961Department of Nursing, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021 Guangxi China
| | - Jiani Wei
- grid.412594.f0000 0004 1757 2961Department of Nursing, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021 Guangxi China
| | - Xiao Gan
- grid.412594.f0000 0004 1757 2961Department of Nursing, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021 Guangxi China
| |
Collapse
|
12
|
Kugeratski FG, Santi A, Zanivan S. Extracellular vesicles as central regulators of blood vessel function in cancer. Sci Signal 2022; 15:eaaz4742. [PMID: 36166511 DOI: 10.1126/scisignal.aaz4742] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Blood vessels deliver oxygen and nutrients that sustain tumor growth and enable the dissemination of cancer cells to distant sites and the recruitment of intratumoral immune cells. In addition, the structural and functional abnormalities of the tumor vasculature foster the development of an aggressive tumor microenvironment and impair the efficacy of existing cancer therapies. Extracellular vesicles (EVs) have emerged as major players of tumor progression, and a growing body of evidence has demonstrated that EVs derived from cancer cells trigger multiple responses in endothelial cells that alter blood vessel function in tumors. EV-mediated signaling in endothelial cells can occur through the transfer of functional cargos such as miRNAs, lncRNAs, cirRNAs, and proteins. Moreover, membrane-bound proteins in EVs can elicit receptor-mediated signaling in endothelial cells. Together, these mechanisms reprogram endothelial cells and contribute to the sustained exacerbated angiogenic signaling typical of tumors, which, in turn, influences cancer progression. Targeting these angiogenesis-promoting EV-dependent mechanisms may offer additional strategies to normalize tumor vasculature. Here, we discuss the current knowledge pertaining to the contribution of cancer cell-derived EVs in mechanisms regulating blood vessel functions in tumors. Moreover, we discuss the translational opportunities in targeting the dysfunctional tumor vasculature using EVs and highlight the open questions in the field of EV biology that can be addressed using mass spectrometry-based proteomics analysis.
Collapse
Affiliation(s)
- Fernanda G Kugeratski
- Department of Immunology, University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Alice Santi
- Department of Experimental and Clinical Biomedical Sciences, Università degli Studi di Firenze, 50134 Firenze, Italy
| | - Sara Zanivan
- CRUK Beatson Institute, Switchback Road, Glasgow G61 1BD, UK
- School of Cancer Sciences, University of Glasgow, Switchback Road, Glasgow G61 1QH, UK
| |
Collapse
|
13
|
Custodia A, Ouro A, Sargento-Freitas J, Aramburu-Núñez M, Pías-Peleteiro JM, Hervella P, Rosell A, Ferreira L, Castillo J, Romaus-Sanjurjo D, Sobrino T. Unraveling the potential of endothelial progenitor cells as a treatment following ischemic stroke. Front Neurol 2022; 13:940682. [PMID: 36158970 PMCID: PMC9492921 DOI: 10.3389/fneur.2022.940682] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 08/15/2022] [Indexed: 11/13/2022] Open
Abstract
Ischemic stroke is becoming one of the most common causes of death and disability in developed countries. Since current therapeutic options are quite limited, focused on acute reperfusion therapies that are hampered by a very narrow therapeutic time window, it is essential to discover novel treatments that not only stop the progression of the ischemic cascade during the acute phase, but also improve the recovery of stroke patients during the sub-acute or chronic phase. In this regard, several studies have shown that endothelial progenitor cells (EPCs) can repair damaged vessels as well as generate new ones following cerebrovascular damage. EPCs are circulating cells with characteristics of both endothelial cells and adult stem cells presenting the ability to differentiate into mature endothelial cells and self-renew, respectively. Moreover, EPCs have the advantage of being already present in healthy conditions as circulating cells that participate in the maintenance of the endothelium in a direct and paracrine way. In this scenario, EPCs appear as a promising target to tackle stroke by self-promoting re-endothelization, angiogenesis and vasculogenesis. Based on clinical data showing a better neurological and functional outcome in ischemic stroke patients with higher levels of circulating EPCs, novel and promising therapeutic approaches would be pharmacological treatment promoting EPCs-generation as well as EPCs-based therapies. Here, we will review the latest advances in preclinical as well as clinical research on EPCs application following stroke, not only as a single treatment but also in combination with new therapeutic approaches.
Collapse
Affiliation(s)
- Antía Custodia
- NeuroAging Laboratory (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Alberto Ouro
- NeuroAging Laboratory (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - João Sargento-Freitas
- Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
- Faculdade de Medicina da Universidade de Coimbra, Coimbra, Portugal
- Centro Neurociências e Biologia Celular, Coimbra, Portugal
| | - Marta Aramburu-Núñez
- NeuroAging Laboratory (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Juan Manuel Pías-Peleteiro
- NeuroAging Laboratory (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Pablo Hervella
- Neuroimaging and Biotechnology Laboratory (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Anna Rosell
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Lino Ferreira
- Faculdade de Medicina da Universidade de Coimbra, Coimbra, Portugal
- Centro Neurociências e Biologia Celular, Coimbra, Portugal
- CNC-Center for Neuroscience and Cell Biology, CIBB-Centre for Innovative Biomedicine and Biotechnology, UC, Biotech Parque Tecnológico de Cantanhede, University of Coimbra, Coimbra, Portugal
| | - José Castillo
- Neuroimaging and Biotechnology Laboratory (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Daniel Romaus-Sanjurjo
- NeuroAging Laboratory (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
- *Correspondence: Daniel Romaus-Sanjurjo
| | - Tomás Sobrino
- NeuroAging Laboratory (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
- Tomás Sobrino
| |
Collapse
|
14
|
Kale VP. A chimeric feeder comprising transforming growth factor beta 1- and basic fibroblast growth factor-primed bone marrow-derived mesenchymal stromal cells suppresses the expansion of hematopoietic stem and progenitor cells. Cell Biol Int 2022; 46:2132-2141. [PMID: 36073008 DOI: 10.1002/cbin.11904] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 08/20/2022] [Indexed: 12/19/2022]
Abstract
Bone marrow-derived mesenchymal stromal cells (BMSCs) physically associate with the hematopoietic stem cells (HSCs), forming a unique HSC niche. Owing to this proximity, the signaling mechanisms prevailing in the BMSCs affect the fate of the HSCs. In addition to cell-cell and cell-extracellular matrix interactions, various cytokines and growth factors present in the BM milieu evoke signaling mechanisms in the BMSCs. Previously, I have shown that priming of human BMSCs with transforming growth factor β1 (TGFβ1), a cytokine consistently found at active sites of hematopoiesis, boosts their hematopoiesis-supportive ability. Basic fibroblast growth factor (bFGF), another cytokine present in the marrow microenvironment, positively regulates hematopoiesis. Hence, I examined whether priming human BMSCs with bFGF improves their hematopoiesis-supportive ability. I found that bFGF-primed BMSCs stimulate hematopoiesis, as seen by a significant increase in colony formation from the bone marrow cells briefly interacted with them and the extensive proliferation of CD34+ HSCs cocultured with them. However, contrary to my expectation, I found that chimeric feeders comprising a mixture of TGF-primed and bFGF-primed BMSCs exerted a suppressive effect. These data demonstrate that though the TGF- and bFGF-primed BMSCs exert a salutary effect on hematopoiesis when used independently, they exert a suppressive effect when presented as a chimera. These findings suggest that the combinatorial effect of various priming agents and cytokines on the functionality of BMSCs toward the target tissues needs to be critically evaluated before they are clinically applied.
Collapse
Affiliation(s)
- Vaijayanti P Kale
- Symbiosis Centre for Stem Cell Research, Symbiosis School of Biological Sciences, Pune, Maharashtra, India.,National Centre for Cell Science, Ganeshkhind, Pune, India
| |
Collapse
|
15
|
Mallick R, Gurzeler E, Toivanen PI, Nieminen T, Ylä-Herttuala S. Novel Designed Proteolytically Resistant VEGF-B186R127S Promotes Angiogenesis in Mouse Heart by Recruiting Endothelial Progenitor Cells. Front Bioeng Biotechnol 2022; 10:907538. [PMID: 35992336 PMCID: PMC9385986 DOI: 10.3389/fbioe.2022.907538] [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: 03/29/2022] [Accepted: 06/21/2022] [Indexed: 11/23/2022] Open
Abstract
Background: Previous studies have indicated that vascular endothelial growth factor B186 (VEGF-B186) supports coronary vascular growth in normal and ischemic myocardium. However, previous studies also indicated that induction of ventricular arrhythmias is a severe side effect preventing the use of VEGF-B186 in cardiac gene therapy, possibly mediated by binding to neuropilin 1 (NRP1). We have designed a novel VEGF-B186 variant, VEGF-B186R127S, which is resistant to proteolytic processing and unable to bind to NRP1. Here, we studied its effects on mouse heart to explore the mechanism of VEGF-B186-induced vascular growth along with its effects on cardiac performance. Methods: Following the characterization of VEGF-B186R127S, we performed ultrasound-guided adenoviral VEGF-B186R127S gene transfers into the murine heart. Vascular growth and heart functions were analyzed using immunohistochemistry, RT-PCR, electrocardiogram and ultrasound examinations. Endothelial progenitor cells (EPCs) were isolated from the circulating blood and characterized. Also, in vitro experiments were carried out in cardiac endothelial cells with adenoviral vectors. Results: The proteolytically resistant VEGF-B186R127S significantly induced vascular growth in mouse heart. Interestingly, VEGF-B186R127S gene transfer increased the number of circulating EPCs that secreted VEGF-A. Other proangiogenic factors were also present in plasma and heart tissue after the VEGF-B186R127S gene transfer. Importantly, VEGF-B186R127S gene transfer did not cause any side effects, such as arrhythmias. Conclusion: VEGF-B186R127S induces vascular growth in mouse heart by recruiting EPCs. VEGF-B186R127S is a novel therapeutic agent for cardiac therapeutic angiogenesis to rescue myocardial tissue after an ischemic insult.
Collapse
Affiliation(s)
- Rahul Mallick
- A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Erika Gurzeler
- A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Pyry I. Toivanen
- A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Tiina Nieminen
- A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
- Kuopio Center for Gene and Cell Therapy, Kuopio, Finland
| | - Seppo Ylä-Herttuala
- A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
- Heart Center and Gene Therapy Unit, Kuopio University Hospital, Kuopio, Finland
- *Correspondence: Seppo Ylä-Herttuala,
| |
Collapse
|
16
|
Canjuga D, Steinle H, Mayer J, Uhde AK, Klein G, Wendel HP, Schlensak C, Avci-Adali M. Homing of mRNA-Modified Endothelial Progenitor Cells to Inflamed Endothelium. Pharmaceutics 2022; 14:pharmaceutics14061194. [PMID: 35745767 PMCID: PMC9229815 DOI: 10.3390/pharmaceutics14061194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 05/28/2022] [Accepted: 05/30/2022] [Indexed: 01/25/2023] Open
Abstract
Endothelial progenitor cells (EPCs) are one of the most important stem cells for the neovascularization of tissues damaged by ischemic diseases such as myocardial infarction, ischemic stroke, or critical limb ischemia. However, their low homing efficiency in the treatment of ischemic tissues limits their potential clinical applications. The use of synthetic messenger RNA (mRNA) for cell engineering represents a novel and promising technology for the modulation of cell behavior and tissue regeneration. To improve the therapeutic potential of EPCs, in this study, murine EPCs were engineered with synthetic mRNAs encoding C-X-C chemokine receptor 4 (CXCR4) and P-selectin glycoprotein ligand 1 (PSGL-1) to increase the homing and migration efficiency of EPCs to inflamed endothelium. Flow cytometric measurements revealed that the transfection of EPCs with CXCR4 and PSGL-1 mRNA resulted in increased expressions of CXCR4 and PSGL-1 on the cell surface compared with the unmodified EPCs. The transfection of EPCs with mRNAs did not affect cell viability. CXCR4-mRNA-modified EPCs showed significantly higher migration potential than unmodified cells in a chemotactic migration assay. The binding strength of the EPCs to inflamed endothelium was determined with single-cell atomic force microscopy (AFM). This showed that the mRNA-modified EPCs required a three-fold higher detachment force to be released from the TNF-α-activated endothelium than unmodified EPCs. Furthermore, in a dynamic flow model, significantly increased binding of the mRNA-modified EPCs to inflamed endothelium was detected. This study showed that the engineering of EPCs with homing factors encoding synthetic mRNAs increases the homing and migration potentials of these stem cells to inflamed endothelium. Thus, this strategy represents a promising strategy to increase the therapeutic potential of EPCs for the treatment of ischemic tissues.
Collapse
Affiliation(s)
- Denis Canjuga
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tuebingen, Calwerstraße 7/1, 72076 Tuebingen, Germany; (D.C.); (H.S.); (J.M.); (A.-K.U.); (H.P.W.); (C.S.)
| | - Heidrun Steinle
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tuebingen, Calwerstraße 7/1, 72076 Tuebingen, Germany; (D.C.); (H.S.); (J.M.); (A.-K.U.); (H.P.W.); (C.S.)
| | - Jana Mayer
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tuebingen, Calwerstraße 7/1, 72076 Tuebingen, Germany; (D.C.); (H.S.); (J.M.); (A.-K.U.); (H.P.W.); (C.S.)
| | - Ann-Kristin Uhde
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tuebingen, Calwerstraße 7/1, 72076 Tuebingen, Germany; (D.C.); (H.S.); (J.M.); (A.-K.U.); (H.P.W.); (C.S.)
| | - Gerd Klein
- Center for Medical Research, Department of Medicine II, University of Tuebingen, Waldhörnlestraße 22, 72072 Tuebingen, Germany;
| | - Hans Peter Wendel
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tuebingen, Calwerstraße 7/1, 72076 Tuebingen, Germany; (D.C.); (H.S.); (J.M.); (A.-K.U.); (H.P.W.); (C.S.)
| | - Christian Schlensak
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tuebingen, Calwerstraße 7/1, 72076 Tuebingen, Germany; (D.C.); (H.S.); (J.M.); (A.-K.U.); (H.P.W.); (C.S.)
| | - Meltem Avci-Adali
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tuebingen, Calwerstraße 7/1, 72076 Tuebingen, Germany; (D.C.); (H.S.); (J.M.); (A.-K.U.); (H.P.W.); (C.S.)
- Correspondence: ; Tel.: +49-7071-29-86605; Fax: +49-7071-29-3617
| |
Collapse
|
17
|
Heinisch PP, Bello C, Emmert MY, Carrel T, Dreßen M, Hörer J, Winkler B, Luedi MM. Endothelial Progenitor Cells as Biomarkers of Cardiovascular Pathologies: A Narrative Review. Cells 2022; 11:cells11101678. [PMID: 35626716 PMCID: PMC9139418 DOI: 10.3390/cells11101678] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 05/11/2022] [Accepted: 05/13/2022] [Indexed: 01/25/2023] Open
Abstract
Endothelial progenitor cells (EPC) may influence the integrity and stability of the vascular endothelium. The association of an altered total EPC number and function with cardiovascular diseases (CVD) and risk factors (CVF) was discussed; however, their role and applicability as biomarkers for clinical purposes have not yet been defined. Endothelial dysfunction is one of the key mechanisms in CVD. The assessment of endothelial dysfunction in vivo remains a major challenge, especially for a clinical evaluation of the need for therapeutic interventions or for primary prevention of CVD. One of the main challenges is the heterogeneity of this particular cell population. Endothelial cells (EC) can become senescent, and the majority of circulating endothelial cells (CEC) show evidence of apoptosis or necrosis. There are a few viable CECs that have properties similar to those of an endothelial progenitor cell. To use EPC levels as a biomarker for vascular function and cumulative cardiovascular risk, a correct definition of their phenotype, as well as an update on the clinical application and practicability of current isolation methods, are an urgent priority.
Collapse
Affiliation(s)
- Paul Philipp Heinisch
- Department of Congenital and Pediatric Heart Surgery, German Heart Center Munich, School of Medicine, Technical University of Munich, 80636 Munich, Germany;
- Division of Congenital and Pediatric Heart Surgery, University Hospital of Munich, Ludwig-Maximilians-Universität, 80636 Munich, Germany
- Department of Anaesthesiology and Pain Medicine, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland; (C.B.); (M.M.L.)
- Correspondence:
| | - Corina Bello
- Department of Anaesthesiology and Pain Medicine, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland; (C.B.); (M.M.L.)
| | - Maximilian Y. Emmert
- Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, 13353 Berlin, Germany;
- Institute of Regenerative Medicine (IREM), University of Zurich, 8952 Schlieren, Switzerland
- Department of Cardiovascular Surgery, Charité Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Thierry Carrel
- Department of Cardiac Surgery, University Hospital Zurich, 8091 Zurich, Switzerland;
| | - Martina Dreßen
- Department of Cardiovascular Surgery, Institute Insure, German Heart Center Munich, School of Medicine & Health, Technical University of Munich, Lazarettstrasse 36, 80636 Munich, Germany;
| | - Jürgen Hörer
- Department of Congenital and Pediatric Heart Surgery, German Heart Center Munich, School of Medicine, Technical University of Munich, 80636 Munich, Germany;
- Division of Congenital and Pediatric Heart Surgery, University Hospital of Munich, Ludwig-Maximilians-Universität, 80636 Munich, Germany
| | - Bernhard Winkler
- Department of Cardiovascular Surgery, Hospital Hietzing, 1130 Vienna, Austria;
| | - Markus M. Luedi
- Department of Anaesthesiology and Pain Medicine, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland; (C.B.); (M.M.L.)
| |
Collapse
|
18
|
Xu Z, Wang Y, Liu G, Chen J, Wang W, Cheng Y, Ren Q, Cui Y, Yang W, Liu Z, Chen X, Xue J, Chang T, Qu X, Yu S, Zhou Y, Xu K, Su Z, Deng Q, Zhao Y, Yang H. A randomized, open-label, single-dose, two-cycle crossover study to evaluate the bioequivalence and safety of lenvatinib and Lenvima® in Chinese healthy subjects. Expert Opin Investig Drugs 2022; 31:737-746. [PMID: 35427205 DOI: 10.1080/13543784.2022.2067528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Affiliation(s)
- Zhongnan Xu
- Department of clinical research center, Chia Tai Tianqing Pharmaceutical Group Co.Ltd, Jiangsu, China
| | - Yanli Wang
- Phase I Clinical Trial Laboratory, Affiliated Hospital to Changchun University of Chinese Medicine, Jilin, China
| | - Guangwen Liu
- Phase I Clinical Trial Laboratory, Affiliated Hospital to Changchun University of Chinese Medicine, Jilin, China
| | - Jiahui Chen
- Phase I Clinical Trial Laboratory, Affiliated Hospital to Changchun University of Chinese Medicine, Jilin, China
| | - Wanhua Wang
- Phase I Clinical Trial Laboratory, Affiliated Hospital to Changchun University of Chinese Medicine, Jilin, China
| | - Yang Cheng
- Phase I Clinical Trial Laboratory, Affiliated Hospital to Changchun University of Chinese Medicine, Jilin, China
| | - Qing Ren
- Phase I Clinical Trial Laboratory, Affiliated Hospital to Changchun University of Chinese Medicine, Jilin, China
| | - Yingzi Cui
- Phase I Clinical Trial Laboratory, Affiliated Hospital to Changchun University of Chinese Medicine, Jilin, China
| | - Wei Yang
- Phase I Clinical Trial Laboratory, Affiliated Hospital to Changchun University of Chinese Medicine, Jilin, China
| | - Zhengzhi Liu
- Phase I Clinical Trial Laboratory, Affiliated Hospital to Changchun University of Chinese Medicine, Jilin, China
| | - Xuesong Chen
- Ansiterui Medical Technology Consulting Co.,Ltd., Jilin, China
| | - Jinling Xue
- Department of clinical research center, Chia Tai Tianqing Pharmaceutical Group Co.Ltd, Jiangsu, China
| | - Tianying Chang
- Phase I Clinical Trial Laboratory, Affiliated Hospital to Changchun University of Chinese Medicine, Jilin, China
| | - Xinyao Qu
- Phase I Clinical Trial Laboratory, Affiliated Hospital to Changchun University of Chinese Medicine, Jilin, China
| | - Shuang Yu
- Phase I Clinical Trial Laboratory, Affiliated Hospital to Changchun University of Chinese Medicine, Jilin, China
| | - Yannan Zhou
- Phase I Clinical Trial Laboratory, Affiliated Hospital to Changchun University of Chinese Medicine, Jilin, China
| | - Kaibo Xu
- Phase I Clinical Trial Laboratory, Affiliated Hospital to Changchun University of Chinese Medicine, Jilin, China
| | - Zhengjie Su
- Phase I Clinical Trial Laboratory, Affiliated Hospital to Changchun University of Chinese Medicine, Jilin, China
| | - Qiaohuan Deng
- Phase I Clinical Trial Laboratory, Affiliated Hospital to Changchun University of Chinese Medicine, Jilin, China
| | - Yicheng Zhao
- Clinical Medical College, Changchun University of Chinese Medicine, Jilin, China
| | - Haimiao Yang
- Phase I Clinical Trial Laboratory, Affiliated Hospital to Changchun University of Chinese Medicine, Jilin, China
| |
Collapse
|
19
|
Lee SM, Heo T, Kim G, Kim H. Current status and development direction of hyperbaric medicine in Korea. JOURNAL OF THE KOREAN MEDICAL ASSOCIATION 2022. [DOI: 10.5124/jkma.2022.65.4.232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Background: The indications of hyperbaric oxygen therapy (HBOT) covered by the health insurance in Korea increased to 16 in 2019, which includes acute central retinal artery obstruction within 24 hours of vision loss; anemia due to excessive bleeding, since blood transfusion is impractical; chronic refractory osteomyelitis (Wagner grade ≥3); and intracranial abscess.Current Concepts: HBOT affects the treatment in the primary and secondary mechanisms. According to the Boyle’s law, the primary mechanism is important in treating decompression sickness and intravascular air embolism by decreasing the volume of air bubbles when pressure increases, whereas the secondary mechanism involves hyperoxygenation of the primary mechanism and various effects, such as vasoconstriction, angiogenesis, immune function enhancement, reperfusion injury prevention, antimicrobial action, and gas washout effect, occur. In the past 5 years, domestic HBOT has made significant progress. However, there are many non-therapeutic lowpressure HBOT facilities that are limited by insurance coverage issues, quality equipment management, and medical personnel in HBOT facilities.Discussion and Conclusion: To solve the problem, the introduction of the definition of therapeutic hyperbaric pressure and certification system of HBOT facilities must be initiated. Moreover, the system should be improved so that insurance can be applied for a more indication of HBOT.
Collapse
|
20
|
Sen A, Singh A, Roy A, Mohanty S, Naik N, Kalaivani M, Ramakrishnan L. Role of endothelial colony forming cells (ECFCs) Tetrahydrobiopterin (BH4) in determining ECFCs functionality in coronary artery disease (CAD) patients. Sci Rep 2022; 12:3076. [PMID: 35197509 PMCID: PMC8866483 DOI: 10.1038/s41598-022-06758-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Accepted: 01/31/2022] [Indexed: 01/05/2023] Open
Abstract
Nitric oxide (NO.) is critical for functionality of endothelial colony forming cells (ECFCs). Dimerization of endothelial nitric oxide synthase (eNOS) is must to produce NO. and tetrahydrobiopterin (BH4) plays a crucial role in stabilizing this state. We investigated BH4 level in ECFCs and its effect on ECFCs functionality in CAD patients. Intracellular biopterin levels and ECFCs functionality in terms of cell viability, adhesion, proliferation, in vitro wound healing and angiogenesis were assessed. Guanosine Triphosphate Cyclohydrolase-1 (GTPCH-1) expression was studied in ECFCs. Serum total reactive oxygen/nitrogen species was measured and effect of nitrosative stress on ECFC's biopterins level and functionality were evaluated by treating with 3-morpholino sydnonimine (SIN-1). BH4 level was significantly lower in ECFCs from CAD patients. Cell proliferation, wound closure reflecting cellular migration as well as in vitro angiogenesis were impaired in ECFCs from CAD patients. Wound healing capacity and angiogenesis were positively correlated with ECFC's BH4. A negative effect of nitrosative stress on biopterins level and cell functionality was observed in SIN-1 treated ECFCs. ECFCs from CAD exhibited impaired functionality and lower BH4 level. Association of BH4 with wound healing capacity and angiogenesis suggest its role in maintaining ECFC's functionality. Oxidative stress may be a determinant of intracellular biopterin levels.
Collapse
Affiliation(s)
- Atanu Sen
- Department of Cardiac Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Archna Singh
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Ambuj Roy
- Department of Cardiology, All India Institute of Medical Sciences, New Delhi, India
| | - Sujata Mohanty
- Centre of Excellence for Stem Cell Research, All India Institute of Medical Sciences, New Delhi, India
| | - Nitish Naik
- Department of Cardiology, All India Institute of Medical Sciences, New Delhi, India
| | - Mani Kalaivani
- Department of Biostatistics, All India Institute of Medical Sciences, New Delhi, India
| | - Lakshmy Ramakrishnan
- Department of Cardiac Biochemistry, All India Institute of Medical Sciences, New Delhi, India.
| |
Collapse
|
21
|
Perdomo S, Brugnini A, Trias N, Menyou A, Silveira G, Ranero S, Lens D, Díaz L, Grille S. Mobilized and apheresis-collected endothelial progenitor cells with plerixafor. J Clin Apher 2022; 37:245-252. [PMID: 35114004 DOI: 10.1002/jca.21967] [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: 10/21/2021] [Revised: 01/11/2022] [Accepted: 01/13/2022] [Indexed: 11/10/2022]
Abstract
BACKGROUND Endothelial progenitor cells (EPCs) are immature cells able to proliferate and contribute to endothelial repair, vascular homeostasis, neovascularization, and angiogenesis. It therefore seems likely that circulating EPCs have therapeutic potential in ischemic and vascular diseases. In this study we evaluated the efficiency of EPC mobilization and collection by large volume leukapheresis in subjects with hematological diseases, treated with plerixafor in association with G-CSF. METHODS Twenty-two patients with lymphoid malignancies underwent rHuG-CSF and plerixafor treatment followed by leukapheresis. Blood samples before and after treatment and apheresis liquid sample were taken and analyzed by flow cytometry in order to quantified EPC. RESULTS The percentage of CD34+ cells and EPCs among circulating total nuclear cells (TNCs) increased significantly by approximately 2-fold and 3-fold, respectively, after plerixafor treatment. Consequently, the absolute number of CD34+ cells and EPCs were increased 4-fold after plerixafor treatment. The median PB concentration of EPCs before and after treatment were 0.77/μL (0.31-2.15) and 3.41/μL (1.78-4.54), respectively, P < .0001. The total EPCs collected per patient were 3.3×107 (0.8×107 -6.8×107 ). CONCLUSION We have shown that plerixafor in combination with G-CSF allows the mobilization and collection of large amounts of EPCs along with CD34+ cells in lymphoid neoplasm patients. The possibility to collect and to store these cells could represent a promising therapeutic tool for the treatment of ischemic complications without the need of in vitro expansion.
Collapse
Affiliation(s)
- Susana Perdomo
- Servicio Médico Integral, Centro de Trasplante de Médula Ósea, Montevideo, Uruguay
| | - Andreina Brugnini
- Laboratorio de Citometría y Biología Molecular, Departamento Básico de Medicina, Hospital de Clínicas, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Natalia Trias
- Laboratorio de Citometría y Biología Molecular, Departamento Básico de Medicina, Hospital de Clínicas, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Alba Menyou
- Servicio Médico Integral, Centro de Trasplante de Médula Ósea, Montevideo, Uruguay
| | - Gonzalo Silveira
- Laboratorio de Citometría y Biología Molecular, Departamento Básico de Medicina, Hospital de Clínicas, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Sabrina Ranero
- Laboratorio de Citometría y Biología Molecular, Departamento Básico de Medicina, Hospital de Clínicas, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay.,Facultad de Medicina, Hospital de Clínicas, Universidad de la República, Montevideo, Uruguay
| | - Daniela Lens
- Laboratorio de Citometría y Biología Molecular, Departamento Básico de Medicina, Hospital de Clínicas, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Lilián Díaz
- Servicio Médico Integral, Centro de Trasplante de Médula Ósea, Montevideo, Uruguay
| | - Sofía Grille
- Servicio Médico Integral, Centro de Trasplante de Médula Ósea, Montevideo, Uruguay.,Laboratorio de Citometría y Biología Molecular, Departamento Básico de Medicina, Hospital de Clínicas, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay.,Facultad de Medicina, Hospital de Clínicas, Universidad de la República, Montevideo, Uruguay
| |
Collapse
|
22
|
Oo MW, Kawai H, Takabatake K, Shan Q, Eain HS, Sukegawa S, Nakano K, Nagatsuka H. Cancer-Associated Stromal Cells Promote the Contribution of MMP2-Positive Bone Marrow-Derived Cells to Oral Squamous Cell Carcinoma Invasion. Cancers (Basel) 2021; 14:cancers14010137. [PMID: 35008304 PMCID: PMC8750016 DOI: 10.3390/cancers14010137] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/22/2021] [Accepted: 12/25/2021] [Indexed: 12/14/2022] Open
Abstract
Simple Summary Based on its invasiveness, oral squamous cell carcinoma (OSCC) shows two different subtypes: less-invasive verrucous squamous carcinoma (VSCC) or highly invasive squamous cell carcinoma (SCC). The stromal component influences OSCC progression and invasion. On the other hand, bone marrow-derived cells (BMDCs) are recruited into tumors and involved in tumor development. We hypothesized that stromal factors might also affect the relation of BMDCs and tumor invasion. We established the OSCC models transplanted with stromal cells from VSCC and SCC, and we compared the potential stromal factors of VSCC and SCC for the involvement of BMDCs in tumor invasion. Our study showed that stromal factors IL6 and IL1B might promote the contribution of MMP-2 positive BMDCs to OSCC invasion. Abstract Tumor stromal components contribute to tumor development and invasion. However, the role of stromal cells in the contribution of bone marrow-derived cells (BMDCs) in oral squamous cell carcinoma (OSCC) invasion is unclear. In the present study, we created two different invasive OSCC patient-derived stroma xenografts (PDSXs) and analyzed and compared the effects of stromal cells on the relation of BMDCs and tumor invasion. We isolated stromal cells from two OSCC patients: less invasive verrucous OSCC (VSCC) and highly invasive conventional OSCC (SCC) and co-xenografted with the OSCC cell line (HSC-2) on green fluorescent protein (GFP)-positive bone marrow (BM) cells transplanted mice. We traced the GFP-positive BM cells by immunohistochemistry (IHC) and detected matrix metalloproteinase 2 (MMP2) expression on BM cells by double fluorescent IHC. The results indicated that the SCC-PDSX promotes MMP2-positive BMDCs recruitment to the invasive front line of the tumor. Furthermore, microarray analysis revealed that the expressions of interleukin 6; IL-6 mRNA and interleukin 1 beta; IL1B mRNA were higher in SCC stromal cells than in VSCC stromal cells. Thus, our study first reports that IL-6 and IL1B might be the potential stromal factors promoting the contribution of MMP2-positive BMDCs to OSCC invasion.
Collapse
Affiliation(s)
- May Wathone Oo
- Department of Oral Pathology and Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8525, Japan; (M.W.O.); (K.T.); (Q.S.); (H.S.E.); (S.S.); (K.N.); (H.N.)
| | - Hotaka Kawai
- Department of Oral Pathology and Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8525, Japan; (M.W.O.); (K.T.); (Q.S.); (H.S.E.); (S.S.); (K.N.); (H.N.)
- Correspondence: ; Tel.: +81-86-235-6651
| | - Kiyofumi Takabatake
- Department of Oral Pathology and Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8525, Japan; (M.W.O.); (K.T.); (Q.S.); (H.S.E.); (S.S.); (K.N.); (H.N.)
| | - Qiusheng Shan
- Department of Oral Pathology and Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8525, Japan; (M.W.O.); (K.T.); (Q.S.); (H.S.E.); (S.S.); (K.N.); (H.N.)
- Department of Oral and Maxillofacial Surgery, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8525, Japan
| | - Htoo Shwe Eain
- Department of Oral Pathology and Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8525, Japan; (M.W.O.); (K.T.); (Q.S.); (H.S.E.); (S.S.); (K.N.); (H.N.)
- Department of Oral and Maxillofacial Reconstructive Surgery, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8525, Japan
| | - Shintaro Sukegawa
- Department of Oral Pathology and Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8525, Japan; (M.W.O.); (K.T.); (Q.S.); (H.S.E.); (S.S.); (K.N.); (H.N.)
- Department of Oral and Maxillofacial Surgery, Kagawa Prefectural Central Hospital, Takamatsu 760-0065, Japan
| | - Keisuke Nakano
- Department of Oral Pathology and Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8525, Japan; (M.W.O.); (K.T.); (Q.S.); (H.S.E.); (S.S.); (K.N.); (H.N.)
| | - Hitoshi Nagatsuka
- Department of Oral Pathology and Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8525, Japan; (M.W.O.); (K.T.); (Q.S.); (H.S.E.); (S.S.); (K.N.); (H.N.)
| |
Collapse
|
23
|
Eltanahy AM, Koluib YA, Gonzales A. Pericytes: Intrinsic Transportation Engineers of the CNS Microcirculation. Front Physiol 2021; 12:719701. [PMID: 34497540 PMCID: PMC8421025 DOI: 10.3389/fphys.2021.719701] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 07/29/2021] [Indexed: 12/15/2022] Open
Abstract
Pericytes in the brain are candidate regulators of microcirculatory blood flow because they are strategically positioned along the microvasculature, contain contractile proteins, respond rapidly to neuronal activation, and synchronize microvascular dynamics and neurovascular coupling within the capillary network. Analyses of mice with defects in pericyte generation demonstrate that pericytes are necessary for the formation of the blood-brain barrier, development of the glymphatic system, immune homeostasis, and white matter function. The development, identity, specialization, and progeny of different subtypes of pericytes, however, remain unclear. Pericytes perform brain-wide 'transportation engineering' functions in the capillary network, instructing, integrating, and coordinating signals within the cellular communicome in the neurovascular unit to efficiently distribute oxygen and nutrients ('goods and services') throughout the microvasculature ('transportation grid'). In this review, we identify emerging challenges in pericyte biology and shed light on potential pericyte-targeted therapeutic strategies.
Collapse
Affiliation(s)
- Ahmed M. Eltanahy
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, NV, United States
| | - Yara A. Koluib
- Tanta University Hospitals, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Albert Gonzales
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, NV, United States
| |
Collapse
|
24
|
Smith-Berdan S, Bercasio A, Kramer L, Petkus B, Hinck L, Forsberg EC. Acute and endothelial-specific Robo4 deletion affect hematopoietic stem cell trafficking independent of VCAM1. PLoS One 2021; 16:e0255606. [PMID: 34388149 PMCID: PMC8362960 DOI: 10.1371/journal.pone.0255606] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 07/20/2021] [Indexed: 11/18/2022] Open
Abstract
Hematopoietic stem cell (HSC) trafficking is regulated by a number of complex mechanisms. Among them are the transmembrane protein Robo4 and the vascular cell adhesion molecule, VCAM1. Endothelial VCAM1 is a well-known regulator of hematopoietic cell trafficking, and our previous studies revealed that germline deletion of Robo4 led to impaired HSC trafficking, with an increase in vascular endothelial cell (VEC) numbers and downregulation of VCAM1 protein on sinusoidal VECs. Here, we utilized two Robo4 conditional deletion models in parallel with Robo4 germline knockout mice (R4KO) to evaluate the effects of acute and endothelial cell-specific Robo4 deletion on HSC trafficking. Strikingly similar to the R4KO, the acute deletion of Robo4 resulted in altered HSC distribution between the bone marrow and blood compartments, despite normal numbers of VECs and wild-type levels of VCAM1 cell surface protein on sinusoidal VECs. Additionally, consistent with the R4KO mice, acute loss of Robo4 in the host perturbed long-term engraftment of donor wild-type HSCs and improved HSC mobilization to the peripheral blood. These data demonstrate the significant role that endothelial Robo4 plays in directional HSC trafficking, independent of alterations in VEC numbers and VCAM1 expression.
Collapse
Affiliation(s)
- Stephanie Smith-Berdan
- Institute for the Biology of Stem Cells, University of California-Santa Cruz, Santa Cruz, CA, United States of America
- Department of Biomolecular Engineering, University of California-Santa Cruz, Santa Cruz, CA, United States of America
| | - Alyssa Bercasio
- Institute for the Biology of Stem Cells, University of California-Santa Cruz, Santa Cruz, CA, United States of America
| | - Leah Kramer
- Institute for the Biology of Stem Cells, University of California-Santa Cruz, Santa Cruz, CA, United States of America
| | - Bryan Petkus
- Institute for the Biology of Stem Cells, University of California-Santa Cruz, Santa Cruz, CA, United States of America
| | - Lindsay Hinck
- Institute for the Biology of Stem Cells, University of California-Santa Cruz, Santa Cruz, CA, United States of America
- Department of Molecular, Cell and Developmental Biology, University of California-Santa Cruz, Santa Cruz, CA, United States of America
| | - E. Camilla Forsberg
- Institute for the Biology of Stem Cells, University of California-Santa Cruz, Santa Cruz, CA, United States of America
- Department of Biomolecular Engineering, University of California-Santa Cruz, Santa Cruz, CA, United States of America
- * E-mail:
| |
Collapse
|
25
|
Alterations in microRNA Expression during Hematopoietic Stem Cell Mobilization. BIOLOGY 2021; 10:biology10070668. [PMID: 34356523 PMCID: PMC8301406 DOI: 10.3390/biology10070668] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 07/09/2021] [Accepted: 07/12/2021] [Indexed: 01/01/2023]
Abstract
Simple Summary Lymphoproliferative disorders comprise a heterogeneous group of hematological malignancies characterized by abnormal lymphocyte proliferation. Autologous hematopoietic stem cell transplantation plays a very important role in the treatment of lymphoproliferative diseases. The key element in this process is the effective mobilization of hematopoietic cells from the marrow niche to the peripheral blood. Mobilization of HSC is regulated by many factors, out of which miRNAs present in the hematopoietic niche via targeting cytokines, and signaling pathways may play an important regulatory role. This study investigated the expression of selected miRNAs in patients with multiple myeloma, Hodgkin’s lymphomas, and non-Hodgkin’s lymphomas undergoing mobilization procedures. The aim of the study was to evaluate the expression of hsa-miR-15a-5p, hsa-miR-16-5p, hsa-miR-34a-5p, hsa-miR-126-3p, hsa-miR-146a-5p, hsa-miR-155-5p, and hsa-miR-223-3p during the mobilization procedure, and to assess their role in mobilization efficacy. The level of miRNAs was tested at two time points before the initiation of mobilization and on the day of the first apheresis. Our results suggest that the investigated miRNAs, especially hsa-miR-146a-5p, may influence the efficacy of HSC mobilization. Abstract microRNAs play an important role in the regulation of gene expression, cell fate, hematopoiesis, and may influence the efficacy of CD34+ cell mobilization. The present study examines the role of hsa-miR-15a-5p, hsa-miR-16-5p, hsa-miR-34a-5p, hsa-miR-126-3p, hsa-miR-146a-5p, hsa-miR-155-5p, and hsa-miR-223-3p in the course of hematopoietic stem cell mobilization. The numbers of CD34+ cells collected in patients with hematological malignancies (39 multiple myelomas, 11 lymphomas) were determined during mobilization for an autologous hematopoietic stem cell transplantation. The miRNA level was evaluated by RT-PCR. Compared to baseline, a significant decline in hsa-miR-15a-5p, hsa-miR-16-5p, hsa-miR-126-3p, hsa-miR-146a-5p, and hsa-miR-155-5p was observed on the day of the first apheresis (day A). An increase was observed only in the expression of hsa-miR-34a-5p. On day A, a negative correlation was found between hsa-miR-15a-5p and hsa-miR-146a-5p levels and the number of CD34+ cells in peripheral blood. A negative correlation was observed between hsa-miR-146a-5p and the number of collected CD34+ cells after the first apheresis. Good mobilizers, defined according to GITMO criteria, demonstrated a lower hsa-miR-146a-5p level on day A than poor mobilizers. Patients from the hsa-miR-146a-5p “low expressors” collected more CD34+ cells than “high expressors”. Our results suggest that the investigated miRNAs, especially hsa-miR-146a-5p, may influence the efficacy of HSC mobilization.
Collapse
|
26
|
Snake venom vascular endothelial growth factors (svVEGFs): Unravelling their molecular structure, functions, and research potential. Cytokine Growth Factor Rev 2021; 60:133-143. [PMID: 34090786 DOI: 10.1016/j.cytogfr.2021.05.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/18/2021] [Accepted: 05/24/2021] [Indexed: 02/07/2023]
Abstract
Vascular endothelial growth factor (VEGF) is a key regulator of angiogenesis, a physiological process characterized by the formation of new vessels from a preexisting endothelium. VEGF has also been implicated in pathologic states, such as neoplasias, intraocular neovascular disorders, among other conditions. VEGFs are distributed in seven different families: VEGF-A, B, C, D, and PIGF (placental growth factor), which are identified in mammals; VEGF-E, which are encountered in viruses; and VEGF-F or svVEGF (snake venom VEGF) described in snake venoms. This is the pioneer review of svVEGF family, exploring its distribution among the snake venoms, molecular structure, main functions, and potential applications.
Collapse
|
27
|
Simioni C, Conti I, Varano G, Brenna C, Costanzi E, Neri LM. The Complexity of the Tumor Microenvironment and Its Role in Acute Lymphoblastic Leukemia: Implications for Therapies. Front Oncol 2021; 11:673506. [PMID: 34026651 PMCID: PMC8131840 DOI: 10.3389/fonc.2021.673506] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 03/30/2021] [Indexed: 12/15/2022] Open
Abstract
The microenvironment that surrounds a tumor, in addition to the tumor itself, plays an important role in the onset of resistance to molecularly targeted therapies. Cancer cells and their microenvironment interact closely between them by means of a molecular communication that mutually influences their biological characteristics and behavior. Leukemia cells regulate the recruitment, activation and program of the cells of the surrounding microenvironment, including those of the immune system. Studies on the interactions between the bone marrow (BM) microenvironment and Acute Lymphoblastic Leukemia (ALL) cells have opened a scenario of potential therapeutic targets which include cytokines and their receptors, signal transduction networks, and hypoxia-related proteins. Hypoxia also enhances the formation of new blood vessels, and several studies show how angiogenesis could have a key role in the pathogenesis of ALL. Knowledge of the molecular mechanisms underlying tumor-microenvironment communication and angiogenesis could contribute to the early diagnosis of leukemia and to personalized molecular therapies. This article is part of a Special Issue entitled: Innovative Multi-Disciplinary Approaches for Precision Studies in Leukemia edited by Sandra Marmiroli (University of Modena and Reggio Emilia, Modena, Italy) and Xu Huang (University of Glasgow, Glasgow, United Kingdom).
Collapse
Affiliation(s)
- Carolina Simioni
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy.,Laboratory for Technologies of Advanced Therapies (LTTA) - Electron Microscopy Center, University of Ferrara, Ferrara, Italy
| | - Ilaria Conti
- Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| | - Gabriele Varano
- Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| | - Cinzia Brenna
- Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| | - Eva Costanzi
- Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| | - Luca M Neri
- Laboratory for Technologies of Advanced Therapies (LTTA) - Electron Microscopy Center, University of Ferrara, Ferrara, Italy.,Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| |
Collapse
|
28
|
Crippa S, Santi L, Berti M, De Ponti G, Bernardo ME. Role of ex vivo Expanded Mesenchymal Stromal Cells in Determining Hematopoietic Stem Cell Transplantation Outcome. Front Cell Dev Biol 2021; 9:663316. [PMID: 34017834 PMCID: PMC8129582 DOI: 10.3389/fcell.2021.663316] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 03/17/2021] [Indexed: 02/06/2023] Open
Abstract
Overall, the human organism requires the production of ∼1 trillion new blood cells per day. Such goal is achieved via hematopoiesis occurring within the bone marrow (BM) under the tight regulation of hematopoietic stem and progenitor cell (HSPC) homeostasis made by the BM microenvironment. The BM niche is defined by the close interactions of HSPCs and non-hematopoietic cells of different origin, which control the maintenance of HSPCs and orchestrate hematopoiesis in response to the body’s requirements. The activity of the BM niche is regulated by specific signaling pathways in physiological conditions and in case of stress, including the one induced by the HSPC transplantation (HSCT) procedures. HSCT is the curative option for several hematological and non-hematological diseases, despite being associated with early and late complications, mainly due to a low level of HSPC engraftment, impaired hematopoietic recovery, immune-mediated graft rejection, and graft-versus-host disease (GvHD) in case of allogenic transplant. Mesenchymal stromal cells (MSCs) are key elements of the BM niche, regulating HSPC homeostasis by direct contact and secreting several paracrine factors. In this review, we will explore the several mechanisms through which MSCs impact on the supportive activity of the BM niche and regulate HSPC homeostasis. We will further discuss how the growing understanding of such mechanisms have impacted, under a clinical point of view, on the transplantation field. In more recent years, these results have instructed the design of clinical trials to ameliorate the outcome of HSCT, especially in the allogenic setting, and when low doses of HSPCs were available for transplantation.
Collapse
Affiliation(s)
- Stefania Crippa
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Ludovica Santi
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Margherita Berti
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Giada De Ponti
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Centro Ricerca M. Tettamanti, Department of Pediatrics, University of Milano-Bicocca, Monza, Italy
| | - Maria Ester Bernardo
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Pediatric Immunohematology and Bone Marrow Transplantation Unit, San Raffaele Scientific Institute, Milan, Italy.,University Vita-Salute San Raffaele, Faculty of Medicine, Milan, Italy
| |
Collapse
|
29
|
Abstract
PURPOSE OF REVIEW Liver transplantation is the gold standard for the treatment of end-stage liver disease. However, a shortage of donor organs, high cost, and surgical complications limit the use of this treatment. Cellular therapies using hepatocytes, hematopoietic stem cells, bone marrow mononuclear cells, and mesenchymal stem cells (MSCs) are being investigated as alternative treatments to liver transplantation. The purpose of this review is to describe studies using MSC transplantation for liver diseases based on the reported literature and to discuss prospective research designed to improve the efficacy of MSC therapy. RECENT FINDINGS MSCs have several properties that show potential to regenerate injured tissues or organs, such as homing, transdifferentiation, immunosuppression, and cellular protective capacity. Additionally, MSCs can be noninvasively isolated from various tissues and expanded ex vivo in sufficient numbers for clinical evaluation. SUMMARY Currently, there is no approved MSC therapy for the treatment of liver disease. However, MSC therapy is considered a promising alternative treatment for end-stage liver diseases and is reported to improve liver function safely with no side effects. Further robust preclinical and clinical studies will be needed to improve the therapeutic efficacy of MSC transplantation.
Collapse
|
30
|
The Role of VEGF Receptors as Molecular Target in Nuclear Medicine for Cancer Diagnosis and Combination Therapy. Cancers (Basel) 2021; 13:cancers13051072. [PMID: 33802353 PMCID: PMC7959315 DOI: 10.3390/cancers13051072] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 02/13/2021] [Accepted: 02/24/2021] [Indexed: 12/17/2022] Open
Abstract
Simple Summary The rapid development of diagnostic and therapeutic methods of the cancer treatment causes that these diseases are becoming better known and the fight against them is more and more effective. Substantial contribution in this development has nuclear medicine that enables very early cancer diagnosis and early start of the so-called targeted therapy. This therapeutic concept compared to the currently used chemotherapy, causes much fewer undesirable side effects, due to targeting a specific lesion in the body. This review article discusses the possible applications of radionuclide-labelled tracers (peptides, antibodies or synthetic organic molecules) that can visualise cancer cells through pathological blood vessel system in close tumour microenvironment. Hence, at a very early step of oncological disease, targeted therapy can involve in tumour formation and growth. Abstract One approach to anticancer treatment is targeted anti-angiogenic therapy (AAT) based on prevention of blood vessel formation around the developing cancer cells. It is known that vascular endothelial growth factor (VEGF) and vascular endothelial growth factor receptors (VEGFRs) play a pivotal role in angiogenesis process; hence, application of angiogenesis inhibitors can be an effective approach in anticancer combination therapeutic strategies. Currently, several types of molecules have been utilised in targeted VEGF/VEGFR anticancer therapy, including human VEGF ligands themselves and their derivatives, anti-VEGF or anti-VEGFR monoclonal antibodies, VEGF binding peptides and small molecular inhibitors of VEGFR tyrosine kinases. These molecules labelled with diagnostic or therapeutic radionuclides can become, respectively, diagnostic or therapeutic receptor radiopharmaceuticals. In targeted anti-angiogenic therapy, diagnostic radioagents play a unique role, allowing the determination of the emerging tumour, to monitor the course of treatment, to predict the treatment outcomes and, first of all, to refer patients for AAT. This review provides an overview of design, synthesis and study of radiolabelled VEGF/VEGFR targeting and imaging agents to date. Additionally, we will briefly discuss their physicochemical properties and possible application in combination targeted radionuclide tumour therapy.
Collapse
|
31
|
Jarajapu YPR. Targeting Angiotensin-Converting Enzyme-2/Angiotensin-(1-7)/Mas Receptor Axis in the Vascular Progenitor Cells for Cardiovascular Diseases. Mol Pharmacol 2021; 99:29-38. [PMID: 32321734 PMCID: PMC7725063 DOI: 10.1124/mol.119.117580] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 03/31/2020] [Indexed: 12/20/2022] Open
Abstract
Bone marrow-derived hematopoietic stem/progenitor cells are vasculogenic and play an important role in endothelial health and vascular homeostasis by participating in postnatal vasculogenesis. Progenitor cells are mobilized from bone marrow niches in response to remote ischemic injury and migrate to the areas of damage and stimulate revascularization largely by paracrine activation of angiogenic functions in the peri-ischemic vasculature. This innate vasoprotective mechanism is impaired in certain chronic clinical conditions, which leads to the development of cardiovascular complications. Members of the renin-angiotensin system-angiotensin-converting enzymes (ACEs) ACE and ACE2, angiotensin II (Ang II), Ang-(1-7), and receptors AT1 and Mas-are expressed in vasculogenic progenitor cells derived from humans and rodents. Ang-(1-7), generated by ACE2, is known to produce cardiovascular protective effects by acting on Mas receptor and is considered as a counter-regulatory mechanism to the detrimental effects of Ang II. Evidence has now been accumulating in support of the activation of the ACE2/Ang-(1-7)/Mas receptor pathway by pharmacologic or molecular maneuvers, which stimulates mobilization of progenitor cells from bone marrow, migration to areas of vascular damage, and revascularization of ischemic areas in pathologic conditions. This minireview summarizes recent studies that have enhanced our understanding of the physiology and pharmacology of vasoprotective axis in bone marrow-derived progenitor cells in health and disease. SIGNIFICANCE STATEMENT: Hematopoietic stem progenitor cells (HSPCs) stimulate revascularization of ischemic areas. However, the reparative potential is diminished in certain chronic clinical conditions, leading to the development of cardiovascular diseases. ACE2 and Mas receptor are key members of the alternative axis of the renin-angiotensin system and are expressed in HSPCs. Accumulating evidence points to activation of ACE2 or Mas receptor as a promising approach for restoring the reparative potential, thereby preventing the development of ischemic vascular diseases.
Collapse
Affiliation(s)
- Yagna P R Jarajapu
- Department of Pharmaceutical Sciences, College of Health Professions, North Dakota State University, Fargo, North Dakota
| |
Collapse
|
32
|
Lu X, Jiang M, Tian J, Liu W, Wu F, Yu L, Feng G, Zhong S, Xiang Y, Wen H. Growth Arrest-Specific Transcript 5 (GAS5) Exerts Important Roles on the Treatment of BM45 Cells of Liver Cirrhosis. MOLECULAR THERAPY. NUCLEIC ACIDS 2020; 22:1154-1163. [PMID: 33312752 PMCID: PMC7701024 DOI: 10.1016/j.omtn.2020.10.024] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 10/13/2020] [Indexed: 12/13/2022]
Abstract
Bone marrow (BM)-derived CD45 (BM45) cells were demonstrated to exhibit an improved antifibrotic effect on the treatment of CCL4-induced liver fibrosis by significantly increasing the level of matrix metalloproteinase 9 (MMP-9). In this study, we aimed to validate the therapeutic effect of BM45 on the treatment of liver cirrhosis and to further investigate the molecular mechanism underlying the effect of growth arrest-specific transcript 5 (GAS5) on BM45. Accordingly, GAS5 significantly suppressed miR-222 and miR-21 expression but enhanced p27 and MMP-9 expression in HepG2 and LX2 cells. Additionally, GAS5 obstructed transforming growth factor (TGF)-β-induced dysregulation of miR-222, p27, and α-smooth muscle actin (α-SMA) in mice. GAS5 showed a considerable potential to enhance the capability of BM45 in restoring the normal expression of CCL4, miR-222, miR-21, MMP-9, p27, and α-SMA that was dysregulated by alanine aminotransferase (ALT), albumin, and fibrosis. In summary, our study validated the regulatory relationship between miR-21 and MMP-9, as well as between miR-222 and p27. The overexpression of GAS5 upregulated the expression of MMP-9 and p27 via respectively reducing the miR-222 and miR-21 expression, resulting in higher BM45-induced activation of hepatic stellate cells (HSCs). Accordingly, same results were obtained in an animal model, indicating that GAS5 may exert a positive effect on the treatment of BM45 of liver cirrhosis.
Collapse
Affiliation(s)
- Xing Lu
- Key Laboratory of Freshwater Biodiversity Conservation, Ministry of Agriculture and Rural Affairs, Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan 430223, Hubei, China
| | - Ming Jiang
- Key Laboratory of Freshwater Biodiversity Conservation, Ministry of Agriculture and Rural Affairs, Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan 430223, Hubei, China
| | - Juan Tian
- Key Laboratory of Freshwater Biodiversity Conservation, Ministry of Agriculture and Rural Affairs, Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan 430223, Hubei, China
| | - Wei Liu
- Key Laboratory of Freshwater Biodiversity Conservation, Ministry of Agriculture and Rural Affairs, Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan 430223, Hubei, China
| | - Fan Wu
- Key Laboratory of Freshwater Biodiversity Conservation, Ministry of Agriculture and Rural Affairs, Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan 430223, Hubei, China
| | - Lijuan Yu
- Key Laboratory of Freshwater Biodiversity Conservation, Ministry of Agriculture and Rural Affairs, Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan 430223, Hubei, China
| | - Guohui Feng
- School of Basic Medical Sciences, Wuhan University, Wuhan 430071, Hubei, China
| | - Shan Zhong
- School of Basic Medical Sciences, Wuhan University, Wuhan 430071, Hubei, China
- Hubei Province Key Laboratory of Allergy and Immunology, Wuhan 430071, Hubei, China
| | - Ying Xiang
- School of Basic Medical Sciences, Wuhan University, Wuhan 430071, Hubei, China
| | - Hua Wen
- Key Laboratory of Freshwater Biodiversity Conservation, Ministry of Agriculture and Rural Affairs, Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan 430223, Hubei, China
| |
Collapse
|
33
|
The role of vascular niche and endothelial cells in organogenesis and regeneration. Exp Cell Res 2020; 398:112398. [PMID: 33271129 DOI: 10.1016/j.yexcr.2020.112398] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 11/21/2020] [Accepted: 11/22/2020] [Indexed: 02/08/2023]
Abstract
The term vascular niche indicate the physical and biochemical microenvironment around blood vessel where endothelial cells, pericytes, and smooth muscle cells organize themselves to form blood vessels and release molecules involved in the recruitment of hematopoietic stem cells, endothelial progenitor cells and mesenchymal stem cells. The vascular niche creates a permissive environment that enables different cell types to realize their developmental or regenerative programs. In this context, the proximity between the endothelium and the new-forming cellular components of organs suggests an essential role of endothelial cells in the organs maturation. Dynamic interactions between specific organ endothelial cells and different cellular conponents are crucial for different organ morphogenesis and function. Conversely, organs provide cues shaping vascular network structure.
Collapse
|
34
|
Wen C, Zhang J, Li Y, Zheng W, Liu M, Zhu Y, Sui X, Zhang X, Han Q, Lin Y, Yang J, Zhang L. A zwitterionic hydrogel coated titanium surface with high-efficiency endothelial cell selectivity for rapid re-endothelialization. Biomater Sci 2020; 8:5441-5451. [PMID: 32996913 DOI: 10.1039/d0bm00671h] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Coronary stent implantation is an effective procedure for percutaneous coronary intervention treatment. However, its long-term safety and efficacy are still hindered by the in-stent restenosis and late thrombus formation. Herein, an anti-biofouling and endothelial cell selective zwitterionic hydrogel coating was developed to simultaneously enhance the nonspecific resistance and rapid re-endothelialization of the titanium surface. An endothelial cell selective peptide, REDV, could be simply conjugated on the zwitterionic carboxybetaine (CB) hydrogel to prepare the REDV/CB coating. It was found that the REDV/CB hydrogel layer maintained antifouling properties, which could inhibit the protein adsorption, bacterial adhesion, platelet activation and aggregation, and smooth muscle cell proliferation. More importantly, the co-culture study confirmed that the conjugated REVD peptide could specifically capture endothelial cells and promote their migration and proliferation, and simultaneously decrease the adhesion and proliferation of smooth muscle cells. Therefore, the antifouling and endothelial cell selective coating proposed in this work provides a promising strategy to develop an intravascular stent for promoted re-endothelialization and inhibited neointimal hyperplasia in clinical applications.
Collapse
Affiliation(s)
- Chiyu Wen
- Department of Biochemical Engineering, Frontier Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (MOE), School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300350, China.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Improved in vitro angiogenic behavior of human umbilical vein endothelial cells with oxidized polydopamine coating. Colloids Surf B Biointerfaces 2020; 194:111176. [DOI: 10.1016/j.colsurfb.2020.111176] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 05/18/2020] [Accepted: 06/02/2020] [Indexed: 02/06/2023]
|
36
|
Harding JS, Herbath M, Chen Y, Rayasam A, Ritter A, Csoka B, Hasko G, Michael IP, Fabry Z, Nagy A, Sandor M. VEGF-A from Granuloma Macrophages Regulates Granulomatous Inflammation by a Non-angiogenic Pathway during Mycobacterial Infection. Cell Rep 2020; 27:2119-2131.e6. [PMID: 31091450 DOI: 10.1016/j.celrep.2019.04.072] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 02/11/2019] [Accepted: 04/16/2019] [Indexed: 12/22/2022] Open
Abstract
Many autoimmune and infectious diseases are characterized by the formation of granulomas which are inflammatory lesions that consist of spatially organized immune cells. These sites protect the host and control pathogens like Mycobacterium tuberculosis (Mtb), but are highly inflammatory and cause pathology. Using bacille Calmette-Guerin (BCG) and Mtb infection in mice that induce sarcoid or caseating granulomas, we show that a subpopulation of granuloma macrophages produces vascular endothelial growth factor (VEGF-A), which recruits immune cells to the granuloma by a non-angiogenic pathway. Selective blockade of VEGF-A in myeloid cells, combined with granuloma transplantation, shows that granuloma VEGF-A regulates granulomatous inflammation. The severity of granuloma-related inflammation can be ameliorated by pharmaceutical or genetic inhibition of VEGF-A, which improves survival of mice infected with virulent Mtb without altering host protection. These data show that VEGF-A inhibitors could be used as a host-directed therapy against granulomatous diseases like tuberculosis and sarcoidosis, thereby expanding the value of already existing and approved anti-VEGF-A drugs.
Collapse
Affiliation(s)
- Jeffrey S Harding
- Department of Pathology and Laboratory Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53706, USA; Cellular and Molecular Pathology Training Program, University of Wisconsin-Madison, Madison, WI 53706, USA; Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5T 3H7, Canada
| | - Melinda Herbath
- Department of Pathology and Laboratory Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Yuli Chen
- Department of Pathology and Laboratory Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Aditya Rayasam
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Anna Ritter
- Department of Pathology and Laboratory Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Balazs Csoka
- Department of Anesthesiology, Irving Medical Center, Columbia University, New York, NY 10032, USA
| | - George Hasko
- Department of Anesthesiology, Irving Medical Center, Columbia University, New York, NY 10032, USA
| | - Iacovos P Michael
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5T 3H7, Canada
| | - Zsuzsanna Fabry
- Department of Pathology and Laboratory Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53706, USA; Cellular and Molecular Pathology Training Program, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Andras Nagy
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5T 3H7, Canada; Department of Obstetrics and Gynecology, and Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Matyas Sandor
- Department of Pathology and Laboratory Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53706, USA; Cellular and Molecular Pathology Training Program, University of Wisconsin-Madison, Madison, WI 53706, USA.
| |
Collapse
|
37
|
Liu H, Tang F, Su J, Ma J, Qin Y, Ji L, Geng H, Wang S, Zhang P, Liu J, Cui S, Ge RL, Li Z. EPAS1 regulates proliferation of erythroblasts in chronic mountain sickness. Blood Cells Mol Dis 2020; 84:102446. [PMID: 32470757 DOI: 10.1016/j.bcmd.2020.102446] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 05/14/2020] [Accepted: 05/15/2020] [Indexed: 01/13/2023]
Abstract
Excessive erythrocytosis (EE) is a characteristic of chronic mountain sickness (CMS). Currently, the pathogenesis of CMS remains unclear. This study was intended to investigate the role of EPAS1 in the proliferation of erythroblasts in CMS. Changes of HIF-1α and EPAS1/HIF-2α in the bone marrow erythroblasts of 21 patients with CMS and 14 control subjects residing at the same altitudes were determined by RT-qPCR and western blotting. We also developed a lentiviral vector, Lv-EPAS1/sh-EPAS1, to over-express/silence EPAS1 in K562 cells. Cells cycle and proliferation were detected by flow cytometry. Transcriptome analyses were carried out on Illumina. CMS patients showed a higher expression of EPAS1/HIF-2α in the bone marrow erythroblasts than those of controls. Variations in EPAS1 expression in CMS patients were positively correlated with RBC levels, and negatively correlated with SaO2. Over-expressing of EPAS1 in K562 cells accelerated the erythroid cells cycle progression and promoted the erythroid cells proliferation-and vice versa. Transcriptome data indicated that proliferation-related DEGs were significantly enriched in EPAS1 overexpression/silencing K562 cells. Our results suggest that EPAS1 might participate in the pathogenesis of EE by regulating the proliferation of erythroblasts.
Collapse
Affiliation(s)
- Huihui Liu
- Research Center for High Altitude Medicine, Qinghai University, Xining, China; Qinghai Key Laboratory of Science and Technology for High Altitude Medicine, Xining, China; Qinghai-Utah Joint Research Key Lab for High Altitude Medicine, Xining, China; Department of Rheumatology, Affiliated Hospital of Qinghai University, Xining, China
| | - Feng Tang
- Research Center for High Altitude Medicine, Qinghai University, Xining, China; Qinghai Key Laboratory of Science and Technology for High Altitude Medicine, Xining, China; Qinghai-Utah Joint Research Key Lab for High Altitude Medicine, Xining, China
| | - Juan Su
- Department of Rheumatology, Affiliated Hospital of Qinghai University, Xining, China
| | - Jie Ma
- Department of Hematology, Affiliated Hospital of Qinghai University, Xining, China
| | - Yajing Qin
- Department of Rheumatology, Affiliated Hospital of Qinghai University, Xining, China
| | - Linhua Ji
- Department of Hematology, Affiliated Hospital of Qinghai University, Xining, China
| | - Hui Geng
- Department of Rheumatology, Affiliated Hospital of Qinghai University, Xining, China
| | - Shengyan Wang
- Research Center for High Altitude Medicine, Qinghai University, Xining, China; Qinghai Key Laboratory of Science and Technology for High Altitude Medicine, Xining, China; Qinghai-Utah Joint Research Key Lab for High Altitude Medicine, Xining, China
| | - Peili Zhang
- Research Center for High Altitude Medicine, Qinghai University, Xining, China; Qinghai Key Laboratory of Science and Technology for High Altitude Medicine, Xining, China; Qinghai-Utah Joint Research Key Lab for High Altitude Medicine, Xining, China
| | - Junli Liu
- Research Center for High Altitude Medicine, Qinghai University, Xining, China; Qinghai Key Laboratory of Science and Technology for High Altitude Medicine, Xining, China; Qinghai-Utah Joint Research Key Lab for High Altitude Medicine, Xining, China
| | - Sen Cui
- Department of Hematology, Affiliated Hospital of Qinghai University, Xining, China
| | - Ri-Li Ge
- Research Center for High Altitude Medicine, Qinghai University, Xining, China; Qinghai Key Laboratory of Science and Technology for High Altitude Medicine, Xining, China; Qinghai-Utah Joint Research Key Lab for High Altitude Medicine, Xining, China
| | - Zhanquan Li
- Department of Rheumatology, Affiliated Hospital of Qinghai University, Xining, China.
| |
Collapse
|
38
|
Yanishi K, Shoji K, Fujioka A, Hori Y, Yukawa A, Matoba S. Impact of Therapeutic Angiogenesis Using Autologous Bone Marrow-derived Mononuclear Cell Implantation in Patients with No-option Critical Limb Ischemia. Ann Vasc Dis 2020; 13:13-22. [PMID: 32273917 PMCID: PMC7140169 DOI: 10.3400/avd.ra.20-00002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Recently, the limb salvage rate of patients with critical limb ischemia (CLI) has been improved due to the development of revascularization and wound care treatment. However, many patients with CLI are refractory to standard treatments, including revascularization such as endovascular treatment or surgical bypass. Establishment of a new cell therapy is required to improve the limb salvage rate and prognosis in patients with CLI. In 1997, endothelial progenitor cells were found to be derived from the bone marrow to circulate as CD34 surface antigen positive cells in peripheral blood and to affect therapeutic angiogenesis in ischemic tissues. Later, therapeutic angiogenesis using autologous bone marrow-derived mononuclear cell (BM-MNC) implantation was performed for patients with no-option CLI in clinical practice. Several reports showed the safety and efficacy of the BM-MNC implantation in patients with CLI caused by arteriosclerosis obliterans, thromboangiitis obliterans (TAO), and collagen diseases. In particular, in patients with CLI caused by TAO, limb salvage rate was significantly improved compared with standard treatments. The BM-MNC implantation may be feasible and safe in patients with no-option CLI. Here, we review the efficacy of BM-MNC implantation in no-option CLI, with a focus on therapeutic angiogenesis.
Collapse
Affiliation(s)
- Kenji Yanishi
- Department of Cardiovascular Medicine, Kyoto Prefectural University of Medicine, Kyoto, Kyoto, Japan
| | - Keisuke Shoji
- Department of Cardiovascular Medicine, Kyoto Prefectural University of Medicine, Kyoto, Kyoto, Japan
| | - Ayumu Fujioka
- Department of Cardiovascular Medicine, Kyoto Prefectural University of Medicine, Kyoto, Kyoto, Japan
| | - Yusuke Hori
- Department of Cardiovascular Medicine, Kyoto Prefectural University of Medicine, Kyoto, Kyoto, Japan
| | - Arito Yukawa
- Department of Cardiovascular Medicine, Kyoto Prefectural University of Medicine, Kyoto, Kyoto, Japan
| | - Satoaki Matoba
- Department of Cardiovascular Medicine, Kyoto Prefectural University of Medicine, Kyoto, Kyoto, Japan
| |
Collapse
|
39
|
Lu R, Wang Q, Li J, Miao D. P27 deletion enhances hematopoiesis by paracrine action of IL22 secreted from bone marrow mesenchymal stem cells. Am J Transl Res 2020; 12:787-799. [PMID: 32269712 PMCID: PMC7137050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Accepted: 10/12/2019] [Indexed: 06/11/2023]
Abstract
Previous studies have reported that p27 deletion stimulates the proliferation of bone marrow mesenchymal stem cells (BM-MSCs) and their differentiation into osteoblasts, it also increases bone marrow hematopoietic progenitor cells (HPCs). However, it is unknown whether the enhanced hematopoiesis induced by p27 deficiency was associated with releasing hematopoietic stem cell (HSC) and HPC supporting factors by BM-MSCs. To answer this question, we cultured the BM-MSCs from wild-type (WT) or p27 knockout (KO) mice, analyzed their proliferation, apoptosis and osteogenesis and harvested their conditioned medium (CM); We also cultured the bone marrow cells (BMCs) with normal medium or CM from WT or KO BM-MSCs and analyzed changes of HSCs and HPCs and colony forming cells (CFCs). Our results showed that the proliferation and osteogenic differentiation of BM-MSCs were increased significantly and their apoptosis was reduced significantly in p27 deficient mice. Simultaneously, we demonstrated that the CM from p27 deficient BM-MSCs stimulated the expansion of HSCs/HPCs more dramatically than that from WT BM-MSCs. Five 2-fold up-regulated proteins were identified in the CM from p27 deficient BM-MSCs by protein chip assays, including interleukin-22 (IL-22), transforming growth factor-β type I receptor, tumor necrosis factor-related Apoptosis-inducing ligands, VE-cadherin and vascular endothelial growth factor B. We confirmed that expression of IL22 at both mRNA and protein levels were up-regulated significantly in p27 deficient BM-MSCs. The treatment of IL22 increased the percentages of HSCs and HPCs in BMC cultures and the number of CFCs in the colony formation assay, whereas the increased HSC/HPC expansion induced by the CM derived from p27 deficient BM-MSCs was blocked by the addition of anti-IL22 antibody in a dose dependent manner. We also found that the percentages of IL22R1, Stat3 and p-Stat3-S727 positive HSCs and HPCs were increased significantly in p27 deficient BMCs. Our findings in this study indicate that p27 deficiency stimulates HSC/HPC expansion by increasing secretion of IL22 by BM-MSCs and activating IL22-Stat3 signaling in HSCs and HPCs.
Collapse
Affiliation(s)
- Ruinan Lu
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical UniversityNanjing, China
- Research Center for Bone and Stem Cells, Key Laboratory for Aging & Disease, Nanjing Medical UniversityNanjing 211166, China
| | - Qian Wang
- Department of Human Anatomy, School of Medicine, Jiangsu UniversityZhenjiang 212013, China
| | - Jianyong Li
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical UniversityNanjing, China
| | - Dengshun Miao
- The Research Center for Aging, Friendship Affiliated Plastic Surgery Hospital of Nanjing Medical UniversityNanjing, China
- Research Center for Bone and Stem Cells, Key Laboratory for Aging & Disease, Nanjing Medical UniversityNanjing 211166, China
| |
Collapse
|
40
|
Zhang J, Cui Y, Li X, Xiao Y, Liu L, Jia F, He J, Xie X, Parthasarathy S, Hao H, Fang N. 5F peptide promotes endothelial differentiation of bone marrow stem cells through activation of ERK1/2 signaling. Eur J Pharmacol 2020; 876:173051. [PMID: 32145325 DOI: 10.1016/j.ejphar.2020.173051] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Revised: 02/25/2020] [Accepted: 02/28/2020] [Indexed: 11/29/2022]
Abstract
Synthetic apolipoprotein A-I (apoA-I) mimetic peptide 5F exhibits anti-atherosclerotic ability with largely unknown mechanism(s). Bone marrow (BM)-derived endothelial progenitor cells (EPCs) play a critical role in vascular integrity and function. The objective of the present study was to evaluate the effect of 5F on endothelial differentiation of BM stem cells and related mechanisms. Murine BM multipotent adult progenitor cells (MAPCs) were induced to differentiate into endothelial cells in vitro with or without 5F. The expression of endothelial markers vWF, Flk-1 and CD31 was significantly increased in the cells treated with 5F with enhanced in vitro vascular tube formation and LDL uptake without significant changes on proliferation and stem cell maker Oct-4 expression. Phosphorylated ERK1/2, not Akt, was significantly increased in 5F-treated cells. Treatment of MAPCs with PD98059 or small interfering RNA against ERK2 substantially attenuated ERK1/2 phosphorylation, and effectively prevented 5F-induced enhancement of endothelial differentiation of MAPCs. In vivo studies revealed that 5F increased EPCs number in the BM in mice after acute hindlimb ischemia that was effectively prevented with PD98059 treatment. These data supported the conclusion that 5F promoted endothelial differentiation of MAPCs through activation of ERK1/2 signaling.
Collapse
Affiliation(s)
- Jia Zhang
- Department of Geriatrics, Renji Hospital, School of Medicine, Shanghai Jiao Tong University , Shanghai, 200127, China; Davis Heart & Lung Research Institute and Division of Cardiovascular Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Yuqi Cui
- Davis Heart & Lung Research Institute and Division of Cardiovascular Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Xin Li
- Davis Heart & Lung Research Institute and Division of Cardiovascular Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Yuan Xiao
- Davis Heart & Lung Research Institute and Division of Cardiovascular Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Lingjuan Liu
- Davis Heart & Lung Research Institute and Division of Cardiovascular Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Fengpeng Jia
- Davis Heart & Lung Research Institute and Division of Cardiovascular Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Jianfeng He
- Davis Heart & Lung Research Institute and Division of Cardiovascular Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Xiaoyun Xie
- Davis Heart & Lung Research Institute and Division of Cardiovascular Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Sampath Parthasarathy
- Burnett School of Biomedical Sciences, University of Central Florida College of Medicine, USA
| | - Hong Hao
- Davis Heart & Lung Research Institute and Division of Cardiovascular Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Ningyuan Fang
- Department of Geriatrics, Renji Hospital, School of Medicine, Shanghai Jiao Tong University , Shanghai, 200127, China.
| |
Collapse
|
41
|
Patry C, Doniga T, Lenz F, Viergutz T, Weiss C, Tönshoff B, Kalenka A, Yard B, Krebs J, Schaible T, Beck G, Rafat N. Increased mobilization of mesenchymal stem cells in patients with acute respiratory distress syndrome undergoing extracorporeal membrane oxygenation. PLoS One 2020; 15:e0227460. [PMID: 31986159 PMCID: PMC6984734 DOI: 10.1371/journal.pone.0227460] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Accepted: 12/18/2019] [Indexed: 01/31/2023] Open
Abstract
Background The acute respiratory distress syndrome (ARDS) is characterized by pulmonary epithelial and endothelial barrier dysfunction and injury. In severe forms of ARDS, extracorporeal membrane oxygenation (ECMO) is often the last option for life support. Endothelial progenitor (EPC) and mesenchymal stem cells (MSC) can regenerate damaged endothelium and thereby improve pulmonary endothelial dysfunction. However, we still lack sufficient knowledge about how ECMO might affect EPC- and MSC-mediated regenerative pathways in ARDS. Therefore, we investigated if ECMO impacts EPC and MSC numbers in ARDS patients. Methods Peripheral blood mononuclear cells from ARDS patients undergoing ECMO (n = 16) and without ECMO support (n = 12) and from healthy volunteers (n = 16) were isolated. The number and presence of circulating EPC and MSC was detected by flow cytometry. Serum concentrations of vascular endothelial growth factor (VEGF) and angiopoietin 2 (Ang2) were determined. Results In the ECMO group, MSC subpopulations were higher by 71% compared to the non-ECMO group. Numbers of circulating EPC were not significantly altered. During ECMO, VEGF and Ang2 serum levels remained unchanged compared to the non-ECMO group (p = 0.16), but Ang2 serum levels in non-survivors of ARDS were significantly increased by 100% (p = 0.02) compared to survivors. Conclusions ECMO support in ARDS is specifically associated with an increased number of circulating MSC, most likely due to enhanced mobilization, but not with a higher numbers of EPC or serum concentrations of VEGF and Ang2.
Collapse
Affiliation(s)
- Christian Patry
- Department of Pediatrics I, University Children’s Hospital Heidelberg, University of Heidelberg, Heidelberg, Germany
| | - Thalia Doniga
- Department of Neonatology, University Children’s Hospital Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Franziska Lenz
- Department of Anaesthesiology and Intensive Care Medicine, University Medical Center Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Tim Viergutz
- Department of Anaesthesiology and Intensive Care Medicine, University Medical Center Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Christel Weiss
- Department of Medical Statistics and Biomathematics, Medical Faculty Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Burkhard Tönshoff
- Department of Pediatrics I, University Children’s Hospital Heidelberg, University of Heidelberg, Heidelberg, Germany
| | - Armin Kalenka
- Department of Anaesthesiology and Intensive Care Medicine, University Medical Center Mannheim, University of Heidelberg, Heidelberg, Germany
- Department of Anaesthesiology and Intensive Care Medicine, Hospital Bergstraße, Heppenheim, Germany
| | - Benito Yard
- Department of Medicine V, University Medical Center Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Jörg Krebs
- Department of Anaesthesiology and Intensive Care Medicine, University Medical Center Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Thomas Schaible
- Department of Neonatology, University Children’s Hospital Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Grietje Beck
- Department of Anaesthesiology and Intensive Care Medicine, Dr. Horst-Schmidt Clinic, Wiesbaden, Germany
| | - Neysan Rafat
- Department of Pediatrics I, University Children’s Hospital Heidelberg, University of Heidelberg, Heidelberg, Germany
- Department of Neonatology, University Children’s Hospital Mannheim, University of Heidelberg, Heidelberg, Germany
- Department of Pharmaceutical Sciences, Bahá'í Institute of Higher Education (BIHE), Teheran, Iran
- * E-mail:
| |
Collapse
|
42
|
Rakkar K, Othman O, Sprigg N, Bath P, Bayraktutan U. Endothelial progenitor cells, potential biomarkers for diagnosis and prognosis of ischemic stroke: protocol for an observational case-control study. Neural Regen Res 2020; 15:1300-1307. [PMID: 31960816 PMCID: PMC7047808 DOI: 10.4103/1673-5374.269028] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Ischemic stroke is a devastating, life altering event which can severely reduce patient quality of life. Despite years of research there have been minimal therapeutic advances. Endothelial progenitor cells (EPCs), stem cells involved in both vasculogenesis and angiogenesis, may be a potential therapeutic target. After a stroke, EPCs migrate to the site of ischemic injury to repair cerebrovascular damage, and their numbers and functional capacity may determine patients’ outcome. This study aims to determine whether the number of circulating EPCs and their functional aspects may be used as biomarkers to identify the type (cortical or lacunar) and/or severity of ischemic stroke. The study will also investigate if there are any differences in these characteristics between healthy volunteers over and under 65 years of age. 100 stroke patients (50 lacunar and 50 cortical strokes) will be recruited in this prospective, observational case-controlled study. Blood samples will be taken from stroke patients at baseline (within 48 hours of stroke) and days 7, 30 and 90. EPCs will be counted with flow cytometry. The plasma levels of pro- and anti-angiogenic factors and inflammatory cytokines will also be determined. Outgrowth endothelial cells will be cultured to be used in tube formation, migration and proliferation functional assays. Primary outcome is disability or dependence on day 90 after stroke, assessed by the modified Rankin Scale. Secondary outcomes are changes in circulating EPC numbers and/or functional capacity between patient and healthy volunteers, between patient subgroups and between elderly and young healthy volunteers. Recruitment started in February 2017, 167 participants have been recruited. Recruitment will end in November 2019. West Midlands - Coventry & Warwickshire Research Ethics Committee approved this study (REC number: 16/WM/0304) on September 8, 2016. Protocol version: 2.0. The Bayraktutan Dunhill Medical Trust EPC Study was registered in ClinicalTrials.gov (NCT02980354) on November 15, 2016. This study will determine whether the number of EPCs can be used as a prognostic or diagnostic marker for ischemic strokes and is a step towards discovering if transplantation of EPCs may aid patient recovery.
Collapse
Affiliation(s)
- Kamini Rakkar
- Stroke, Division of Clinical Neuroscience, University of Nottingham, Clinical Sciences Building, City Hospital, Hucknall Road, NG5 1PB, UK
| | - Othman Othman
- Stroke, Division of Clinical Neuroscience, University of Nottingham, Clinical Sciences Building, City Hospital, Hucknall Road, NG5 1PB, UK
| | - Nikola Sprigg
- Stroke, Division of Clinical Neuroscience, University of Nottingham, Clinical Sciences Building, City Hospital, Hucknall Road, NG5 1PB, UK
| | - Philip Bath
- Stroke, Division of Clinical Neuroscience, University of Nottingham, Clinical Sciences Building, City Hospital, Hucknall Road, NG5 1PB, UK
| | - Ulvi Bayraktutan
- Stroke, Division of Clinical Neuroscience, University of Nottingham, Clinical Sciences Building, City Hospital, Hucknall Road, NG5 1PB, UK
| |
Collapse
|
43
|
Albakri M, Tashkandi H, Zhou L. A Review of Advances in Hematopoietic Stem Cell Mobilization and the Potential Role of Notch2 Blockade. Cell Transplant 2020; 29:963689720947146. [PMID: 32749152 PMCID: PMC7563033 DOI: 10.1177/0963689720947146] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 07/09/2020] [Accepted: 07/14/2020] [Indexed: 12/15/2022] Open
Abstract
Hematopoietic stem cell (HSC) transplantation can be a potential cure for hematological malignancies and some nonhematologic diseases. Hematopoietic stem and progenitor cells (HSPCs) collected from peripheral blood after mobilization are the primary source to provide HSC transplantation. In most of the cases, mobilization by the cytokine granulocyte colony-stimulating factor with chemotherapy, and in some settings, with the CXC chemokine receptor type 4 antagonist plerixafor, can achieve high yield of hematopoietic progenitor cells (HPCs). However, adequate mobilization is not always successful in a significant portion of donors. Research is going on to find new agents or strategies to increase HSC mobilization. Here, we briefly review the history of HSC transplantation, current mobilization regimens, some of the novel agents that are under investigation for clinical practice, and our recent findings from animal studies regarding Notch and ligand interaction as potential targets for HSPC mobilization.
Collapse
Affiliation(s)
- Marwah Albakri
- Department of Pathology, Case Western Reserve University, Cleveland,
OH, USA
| | - Hammad Tashkandi
- Department of Pathology, University of Pittsburgh Medical Center,
PA, USA
| | - Lan Zhou
- Department of Pathology, Case Western Reserve University, Cleveland,
OH, USA
| |
Collapse
|
44
|
Abstract
Enforced egress of hematopoietic stem cells (HSCs) out of the bone marrow (BM) into the peripheral circulation, termed mobilization, has come a long way since its discovery over four decades ago. Mobilization research continues to be driven by the need to optimize the regimen currently available in the clinic with regard to pharmacokinetic and pharmacodynamic profile, costs, and donor convenience. In this review, we describe the most recent findings in the field and how we anticipate them to affect the development of mobilization strategies in the future. Furthermore, the significance of mobilization beyond HSC collection, i.e. for chemosensitization, conditioning, and gene therapy as well as a means to study the interactions between HSCs and their BM microenvironment, is reviewed. Open questions, controversies, and the potential impact of recent technical progress on mobilization research are also highlighted.
Collapse
Affiliation(s)
- Darja Karpova
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, 69120, Germany
| | - Michael P Rettig
- Division of Oncology, Department of Medicine, Washington University School of Medicine,, St. Louis, Missouri, 63110, USA
| | - John F DiPersio
- Division of Oncology, Department of Medicine, Washington University School of Medicine,, St. Louis, Missouri, 63110, USA
| |
Collapse
|
45
|
Lugano R, Ramachandran M, Dimberg A. Tumor angiogenesis: causes, consequences, challenges and opportunities. Cell Mol Life Sci 2019; 77:1745-1770. [PMID: 31690961 PMCID: PMC7190605 DOI: 10.1007/s00018-019-03351-7] [Citation(s) in RCA: 1079] [Impact Index Per Article: 179.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 10/10/2019] [Accepted: 10/21/2019] [Indexed: 02/07/2023]
Abstract
Tumor vascularization occurs through several distinct biological processes, which not only vary between tumor type and anatomic location, but also occur simultaneously within the same cancer tissue. These processes are orchestrated by a range of secreted factors and signaling pathways and can involve participation of non-endothelial cells, such as progenitors or cancer stem cells. Anti-angiogenic therapies using either antibodies or tyrosine kinase inhibitors have been approved to treat several types of cancer. However, the benefit of treatment has so far been modest, some patients not responding at all and others acquiring resistance. It is becoming increasingly clear that blocking tumors from accessing the circulation is not an easy task to accomplish. Tumor vessel functionality and gene expression often differ vastly when comparing different cancer subtypes, and vessel phenotype can be markedly heterogeneous within a single tumor. Here, we summarize the current understanding of cellular and molecular mechanisms involved in tumor angiogenesis and discuss challenges and opportunities associated with vascular targeting.
Collapse
Affiliation(s)
- Roberta Lugano
- The Rudbeck Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, 75185, Uppsala, Sweden
| | - Mohanraj Ramachandran
- The Rudbeck Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, 75185, Uppsala, Sweden
| | - Anna Dimberg
- The Rudbeck Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, 75185, Uppsala, Sweden.
| |
Collapse
|
46
|
Metalloproteases: On the Watch in the Hematopoietic Niche. Trends Immunol 2019; 40:1053-1070. [DOI: 10.1016/j.it.2019.09.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 08/15/2019] [Accepted: 09/20/2019] [Indexed: 12/19/2022]
|
47
|
El-Aswad BEDW, Sonbol AA, Moharm IM, El-Refai SA, Seleem HEDM, Soliman SS. Circulating endothelial cells in severe Plasmodium falciparum infection. Parasitol Int 2019; 72:101926. [PMID: 31100355 DOI: 10.1016/j.parint.2019.101926] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2019] [Revised: 05/11/2019] [Accepted: 05/13/2019] [Indexed: 12/28/2022]
Abstract
Plasmodium falciparum infection is associated with diffuse vascular dys-regulation. Levels of blood circulating endothelial cells (CECs; CD146+CD45-) are a marker of vascular injury. This study aimed to measure blood CECs by flow cytometery in patients with acute malaria infection before and after treatment and to evaluate the prognostic value of that measurement for that disease. The subjects were allocated into: Group I: uncomplicated malaria (UM, n = 32), Group II: severe malaria (SM, n = 12), Group III: the treated UM (TUM, n = 32), Group IV: the treated SM (TSM, n = 12) and Group V: healthy controls (HC, n = 25). Before treatment, SM patients showed the highest mean number of CECs (30,658.3 ± 2658.2/5 × 106 peripheral blood mononuclear cells (PBMCs)), followed by UM patients (19,481.56 ± 866.83/5x106PBMCs) and both groups were significantly higher than HC (2034 ± 300.59/5x106PBMCs, P < .001). The level of CECs decreased significantly in both infected groups after treatment; in TUM it became 5602.18 ± 509.72/5 × 106PBMCs and in TSM it reached 8457.5 ± 452.4/5 × 106 PBMCs (both values P < .001 in comparison with SM). By receiver operating characteristic curve analysis, the best cut-off count for CECs which enables prediction of the occurrence of severe malaria infection was 27,150/5 × 106 PBMCs or more, with 100% sensitivity, 100% specificity, and 100% accuracy. CECs had a significant positive correlation with parasitemia index and serum creatinine and a significant negative correlation with hemoglobin concentration in patients with acute malaria. In conclusion, the level of CECs could be used as a biomarker denoting endothelium damage during acute P. falciparum infection, and it correlated with infection severity and predicted its prognosis.
Collapse
Affiliation(s)
- Bahaa El-Deen W El-Aswad
- Department of Medical Parasitology, Faculty of Medicine, Menoufia University, Shebin al-Kom, Egypt.
| | - Ahmed A Sonbol
- Department of Clinical Pathology, Faculty of Medicine, Menoufia University, Shebin al-Kom, Egypt
| | - Ismail M Moharm
- Department of Medical Parasitology, Faculty of Medicine, Menoufia University, Shebin al-Kom, Egypt
| | - Samar A El-Refai
- Department of Medical Parasitology, Faculty of Medicine, Menoufia University, Shebin al-Kom, Egypt
| | - Hosam El-Din M Seleem
- Department of Tropical medicine, Faculty of Medicine, Menoufia University, Shebin al-Kom, Egypt
| | - Shiamaa S Soliman
- Department of Public Health and Community Medicine, Faculty of Medicine, Menoufia University, Shebin al-Kom, Egypt
| |
Collapse
|
48
|
Bisht K, Brunck ME, Matsumoto T, McGirr C, Nowlan B, Fleming W, Keech T, Magor G, Perkins AC, Davies J, Walkinshaw G, Flippin L, Winkler IG, Levesque JP. HIF prolyl hydroxylase inhibitor FG-4497 enhances mouse hematopoietic stem cell mobilization via VEGFR2/KDR. Blood Adv 2019; 3:406-418. [PMID: 30733301 PMCID: PMC6373754 DOI: 10.1182/bloodadvances.2018017566] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2018] [Accepted: 01/06/2019] [Indexed: 02/06/2023] Open
Abstract
In normoxia, hypoxia-inducible transcription factors (HIFs) are rapidly degraded within the cytoplasm as a consequence of their prolyl hydroxylation by oxygen-dependent prolyl hydroxylase domain (PHD) enzymes. We have previously shown that hematopoietic stem and progenitor cells (HSPCs) require HIF-1 for effective mobilization in response to granulocyte colony-stimulating factor (G-CSF) and CXCR4 antagonist AMD3100/plerixafor. Conversely, HIF PHD inhibitors that stabilize HIF-1 protein in vivo enhance HSPC mobilization in response to G-CSF or AMD3100 in a cell-intrinsic manner. We now show that extrinsic mechanisms involving vascular endothelial growth factor receptor-2 (VEGFR2), via bone marrow (BM) endothelial cells, are also at play. PTK787/vatalanib, a tyrosine kinase inhibitor selective for VEGFR1 and VEGFR2, and neutralizing anti-VEGFR2 monoclonal antibody DC101 blocked enhancement of HSPC mobilization by FG-4497. VEGFR2 was absent on mesenchymal and hematopoietic cells and was detected only in Sca1+ endothelial cells in the BM. We propose that HIF PHD inhibitor FG-4497 enhances HSPC mobilization by stabilizing HIF-1α in HSPCs as previously demonstrated, as well as by activating VEGFR2 signaling in BM endothelial cells, which facilitates HSPC egress from the BM into the circulation.
Collapse
Affiliation(s)
- Kavita Bisht
- Cancer Care and Biology Program, Mater Research Institute-The University of Queensland, Translational Research Institute, Woolloongabba, QLD, Australia
| | - Marion E Brunck
- Cancer Care and Biology Program, Mater Research Institute-The University of Queensland, Translational Research Institute, Woolloongabba, QLD, Australia
| | - Taichi Matsumoto
- Cancer Care and Biology Program, Mater Research Institute-The University of Queensland, Translational Research Institute, Woolloongabba, QLD, Australia
- Faculty of Pharmacological Sciences, Fukuoka University, Fukuoka, Japan
| | - Crystal McGirr
- Cancer Care and Biology Program, Mater Research Institute-The University of Queensland, Translational Research Institute, Woolloongabba, QLD, Australia
| | - Bianca Nowlan
- Cancer Care and Biology Program, Mater Research Institute-The University of Queensland, Translational Research Institute, Woolloongabba, QLD, Australia
| | - Whitney Fleming
- Cancer Care and Biology Program, Mater Research Institute-The University of Queensland, Translational Research Institute, Woolloongabba, QLD, Australia
| | - Thomas Keech
- Cancer Care and Biology Program, Mater Research Institute-The University of Queensland, Translational Research Institute, Woolloongabba, QLD, Australia
| | - Graham Magor
- Cancer Care and Biology Program, Mater Research Institute-The University of Queensland, Translational Research Institute, Woolloongabba, QLD, Australia
| | - Andrew C Perkins
- Cancer Care and Biology Program, Mater Research Institute-The University of Queensland, Translational Research Institute, Woolloongabba, QLD, Australia
| | - Julie Davies
- Cancer Care and Biology Program, Mater Research Institute-The University of Queensland, Translational Research Institute, Woolloongabba, QLD, Australia
| | | | | | - Ingrid G Winkler
- Cancer Care and Biology Program, Mater Research Institute-The University of Queensland, Translational Research Institute, Woolloongabba, QLD, Australia
- Faculty of Medicine, The University of Queensland, Herston, QLD, Australia
| | - Jean-Pierre Levesque
- Cancer Care and Biology Program, Mater Research Institute-The University of Queensland, Translational Research Institute, Woolloongabba, QLD, Australia
- Faculty of Medicine, The University of Queensland, Herston, QLD, Australia
| |
Collapse
|
49
|
Ravindran K, Salem MM, Alturki AY, Thomas AJ, Ogilvy CS, Moore JM. Endothelialization following Flow Diversion for Intracranial Aneurysms: A Systematic Review. AJNR Am J Neuroradiol 2019; 40:295-301. [PMID: 30679207 DOI: 10.3174/ajnr.a5955] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 12/08/2018] [Indexed: 01/07/2023]
Abstract
BACKGROUND The underlying mechanism of action of flow diverters is believed to be the induction of aneurysm thrombosis and simultaneous endothelial cell growth along the device struts, thereby facilitating aneurysm exclusion from the circulation. Although extensive attention has been paid to the role of altered cerebrovascular hemodynamics using computational fluid dynamics analyses, relatively less emphasis has been placed on the role of the vascular endothelium in promoting aneurysm healing. PURPOSE Our aim was to systematically review all available literature investigating the mechanism of action of flow diverters in both human patients and preclinical models. DATA SOURCES A systematic search of PubMed, Cochrane Central Register of Controlled Trials MEDLINE, EMBASE, and the Web of Science electronic data bases was conducted in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines. STUDY SELECTION We selected articles assessing the role of endothelialization in flow-diverter treatment of cerebral aneurysms, including both preclinical and clinical studies. DATA ANALYSIS Ten articles were eligible for inclusion in this review. Two assessed endothelialization in human patients, while the other 8 used preclinical models (either rabbits or pigs). DATA SYNTHESIS Methods used to assess endothelialization included optical coherence tomography and scanning electron microscopy. LIMITATIONS A limitation was the heterogeneity of studies. CONCLUSIONS Current data regarding the temporal relationship to flow-diverter placement has largely been derived from work in preclinical animal models. Whether these cells along the device struts originate from adjacent endothelial cells or are the result of homing of circulating endothelial progenitor cells is equivocal.
Collapse
Affiliation(s)
- K Ravindran
- From the Neurosurgical Service, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - M M Salem
- From the Neurosurgical Service, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - A Y Alturki
- From the Neurosurgical Service, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - A J Thomas
- From the Neurosurgical Service, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - C S Ogilvy
- From the Neurosurgical Service, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - J M Moore
- From the Neurosurgical Service, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
50
|
Rafat N, Patry C, Sabet U, Viergutz T, Weiss C, Tönshoff B, Beck G, Schaible T. Endothelial Progenitor and Mesenchymal Stromal Cells in Newborns With Congenital Diaphragmatic Hernia Undergoing Extracorporeal Membrane Oxygenation. Front Pediatr 2019; 7:490. [PMID: 31824902 PMCID: PMC6882772 DOI: 10.3389/fped.2019.00490] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Accepted: 11/07/2019] [Indexed: 02/03/2023] Open
Abstract
Background: Endothelial progenitor (EPC) and mesenchymal stromal cells (MSC) can regenerate damaged endothelium and thereby improve pulmonary endothelial dysfunction. We do not know, how extracorporeal membrane oxygenation (ECMO) might affect EPC- and MSC-mediated regenerative pathways in patients with congenital diaphragmatic hernia (CDH). Therefore, we investigated, if ECMO support impacts EPC and MSC numbers in CDH patients. Methods: Peripheral blood mononuclear cells from newborns with ECMO-dependent (n = 18) and ECMO-independent CDH (n = 12) and from healthy controls (n = 12) were isolated. The numbers of EPC and MSC were identified by flowcytometry. Serum levels of vascular endothelial growth factor (VEGF) and angiopoietin (Ang)-2 were determined. Results: EPC and MSC were elevated in newborns with CDH. ECMO-dependent infants had higher EPC subpopulation counts (2,1-7,6-fold) before treatment compared to ECMO-independent infants. In the disease course, EPC and MSC subpopulation counts in ECMO-dependent infants were lower than before ECMO initiation. During ECMO, VEGF serum levels were significantly reduced (by 90.5%) and Ang2 levels significantly increased (by 74.8%). Conclusions: Our data suggest that ECMO might be associated with a rather impaired mobilization of EPC and MSC and with a depression of VEGF serum levels in newborns with CDH.
Collapse
Affiliation(s)
- Neysan Rafat
- Department of Neonatology, University Children's Hospital Mannheim, University of Heidelberg, Mannheim, Germany.,Department of Pediatrics I, University Children's Hospital Heidelberg, Heidelberg, Germany.,Department of Pharmaceutical Sciences, Bahá'í Institute of Higher Education (BIHE), Teheran, Iran
| | - Christian Patry
- Department of Pediatrics I, University Children's Hospital Heidelberg, Heidelberg, Germany
| | - Ursula Sabet
- Department of Pediatrics I, University Children's Hospital Heidelberg, Heidelberg, Germany
| | - Tim Viergutz
- Department of Anesthesiology and Critical Care Medicine, University Medical Center Mannheim, University of Heidelberg, Mannheim, Germany
| | - Christel Weiss
- Department for Medical Statistics and Biomathematics, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Burkhard Tönshoff
- Department of Pediatrics I, University Children's Hospital Heidelberg, Heidelberg, Germany
| | - Grietje Beck
- Department of Anesthesiology, Helios Dr. Horst-Schmidt Clinic, Wiesbaden, Germany
| | - Thomas Schaible
- Department of Neonatology, University Children's Hospital Mannheim, University of Heidelberg, Mannheim, Germany
| |
Collapse
|