1
|
Kammala AK, Lintao RCV, Hoy R, Selim J, Luisi J, Yaklic JL, Ameredes BT, Menon R. Fetal microchimeric cells influence maternal lung health following term and preterm births. Sci Rep 2024; 14:28417. [PMID: 39557969 PMCID: PMC11574256 DOI: 10.1038/s41598-024-79795-0] [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: 08/06/2024] [Accepted: 11/11/2024] [Indexed: 11/20/2024] Open
Abstract
Fetal microchimerism, the presence of fetal cells in maternal tissues, has garnered interest for its potential role in maternal physiology. In this study, we aimed to explore the impact of fetal microchimeric cells on maternal lung health following term and preterm delivery, particularly in the context of infection-induced preterm birth and subsequent allergic challenges. We characterized the immune cells in maternal lungs using a transgenic mouse model (mT+ Ve, Td Tomato) and high dimensional mass cytometry (CyTOF) techniques. We evaluated their influence on lung function and inflammation. Our findings revealed distinct differences in the immune cell composition of maternal lungs between term and preterm deliveries. Mice delivered preterm significantly increased in fetal-specific cells, such as activated macrophages and Tbet + Ve memory B-cells, compared to term-delivered mice. Conversely, term deliveries showed elevated levels of CD4 cells. Furthermore, preterm-delivered dams demonstrated heightened airway hyperresponsiveness, pro-inflammatory cytokine expression, cellular infiltration, and lung mucous production compared to term-delivered dams. Co-culture experiments demonstrated that microchimeric cells from preterm births stimulated the production of inflammatory cytokines IL-6 and TNF-α in lung epithelial cells. These findings shed light on the complex immune dynamics postpartum and their role in lung complications after preterm birth. Understanding these mechanisms could provide insights for targeted interventions to improve maternal lung health in at-risk populations.
Collapse
Affiliation(s)
- Ananth Kumar Kammala
- Division of Basic Science and Translational Research, Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX, 77555-1062, USA.
| | - Ryan C V Lintao
- Division of Basic Science and Translational Research, Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX, 77555-1062, USA
- Department of Biochemistry and Molecular Biology, College of Medicine, University of the Philippines Manila, Manila, Philippines
| | - Richa Hoy
- John Sealy School of Medicine, The University of Texas Medical Branch at Galveston, Galveston, TX, USA
| | - Jessica Selim
- Division of Basic Science and Translational Research, Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX, 77555-1062, USA
- John Sealy School of Medicine, The University of Texas Medical Branch at Galveston, Galveston, TX, USA
| | - Jonathan Luisi
- Department of Internal Medicine, The University of Texas Medical Branch at Galveston, Galveston, TX, USA
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch at Galveston, Galveston, TX, USA
| | - Jerome L Yaklic
- Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX, USA
| | - Bill T Ameredes
- Department of Internal Medicine, The University of Texas Medical Branch at Galveston, Galveston, TX, USA
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch at Galveston, Galveston, TX, USA
| | - Ramkumar Menon
- Division of Basic Science and Translational Research, Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX, 77555-1062, USA.
| |
Collapse
|
2
|
Meneses-Preza YG, Martínez-Martínez R, Meixueiro-Calderón C, Hernández UM, Retana EA, Ponce-Regalado MD, Gamboa-Domínguez A, León-Contreras JC, Muñoz-Cruz S, Hernández-Pando R, Pérez-Tapia SM, Chávez-Blanco AD, Becerril-Villanueva E, Chacón-Salinas R. Mast Cell Carboxypeptidase A3 Is Associated with Pulmonary Fibrosis Secondary to COVID-19. Int J Mol Sci 2024; 25:12258. [PMID: 39596322 PMCID: PMC11594788 DOI: 10.3390/ijms252212258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 11/05/2024] [Accepted: 11/08/2024] [Indexed: 11/28/2024] Open
Abstract
COVID-19 is an infectious disease caused by SARS-CoV-2; over the course of the disease, a dysregulated immune response leads to excessive inflammation that damages lung parenchyma and compromises its function. One of the cell lineages classically associated with pathological inflammatory processes is mast cells (MCs). MCs and their mediators have been associated with COVID-19; we previously reported the role of carboxypeptidase A3 (CPA3) in severe COVID-19. However, sequelae of SARS-CoV-2 infection have been poorly studied. In patients who successfully resolve the infection, one of the reported sequelae is pulmonary fibrosis (PF). The etiology and exact mechanisms are unknown, and few studies exist. Therefore, the aim of this study was to evaluate whether MCs are associated with PF development after SARS-CoV-2 infection. Our findings demonstrate that during severe cases of SARS-CoV-2 infection, there is an increased amount of CPA3+ MCs in areas with pneumonia, around thrombotic blood vessels, and in fibrotic tissue. Moreover, higher numbers of CPA3-expressing MCs correlate with fibrotic tissue development (r = 0.8323; p = 0.001170). These results suggest that during COVID-19, exacerbated inflammation favors the recruitment or expansion of MCs and CPA3 expression in the lungs, which favors tissue damage and a failure of repair mechanisms, leading to fibrosis.
Collapse
Affiliation(s)
- Yatsiri G. Meneses-Preza
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, ENCB-IPN, Mexico City 11350, Mexico; (Y.G.M.-P.); (R.M.-M.); (S.M.P.-T.)
| | - Ricardo Martínez-Martínez
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, ENCB-IPN, Mexico City 11350, Mexico; (Y.G.M.-P.); (R.M.-M.); (S.M.P.-T.)
| | - Claudia Meixueiro-Calderón
- Departamento de Patología, Centro Médico Naval, Mexico City 04470, Mexico; (C.M.-C.); (U.M.H.); (E.A.R.)
| | - Ulises Manuel Hernández
- Departamento de Patología, Centro Médico Naval, Mexico City 04470, Mexico; (C.M.-C.); (U.M.H.); (E.A.R.)
| | - Elizabeth Angelica Retana
- Departamento de Patología, Centro Médico Naval, Mexico City 04470, Mexico; (C.M.-C.); (U.M.H.); (E.A.R.)
| | - María Dolores Ponce-Regalado
- Departamento de Ciencias de la Salud, Centro Universitario de los Altos, Universidad de Guadalajara, Tepatitlán de Morelos 47620, Mexico;
| | - Armando Gamboa-Domínguez
- Departamento de Patología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico; (A.G.-D.); (J.C.L.-C.)
| | - Juan Carlos León-Contreras
- Departamento de Patología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico; (A.G.-D.); (J.C.L.-C.)
| | - Samira Muñoz-Cruz
- Unidad de Investigación Médica en Inmunoquímica, UMAE Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico;
| | - Rogelio Hernández-Pando
- Sección de Patología Experimental, Departamento de Patología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico;
| | - Sonia M. Pérez-Tapia
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, ENCB-IPN, Mexico City 11350, Mexico; (Y.G.M.-P.); (R.M.-M.); (S.M.P.-T.)
- Unidad de Desarrollo e Investigación en Bioterapéuticos (UDIBI), Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, ENCB-IPN, Mexico City 11340, Mexico
| | - Alma D. Chávez-Blanco
- División de Ciencia Básica, Instituto Nacional de Cancerología (INCan, SS), Mexico City 14080, Mexico;
| | - Enrique Becerril-Villanueva
- Laboratorio de Psicoinmunología, Instituto Nacional de Psiquiatría Ramón de la Fuente, Mexico City 14370, Mexico
| | - Rommel Chacón-Salinas
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, ENCB-IPN, Mexico City 11350, Mexico; (Y.G.M.-P.); (R.M.-M.); (S.M.P.-T.)
| |
Collapse
|
3
|
Schwarzkopf L, Büttner P, Scholtyssek K, Schröter T, Hiller R, Hindricks G, Bollmann A, Laufs U, Ueberham L. C-kit pos cells in the human left atrial appendage. Heliyon 2023; 9:e21268. [PMID: 37954289 PMCID: PMC10637945 DOI: 10.1016/j.heliyon.2023.e21268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 10/09/2023] [Accepted: 10/18/2023] [Indexed: 11/14/2023] Open
Abstract
Background Subpopulations of myocardial c-kitpos cells have the ability to stimulate regeneration in ischemic heart disease by paracrine effects. The left atrial appendage (LAA), which is easy accessible during cardiac surgery, may represent a perfect source for c-kitpos cell extraction for autologous cell therapies in the living human. So far, frequency and distribution of c-kitpos cells in LAA are unknown. Methods LAAs of patients who underwent cardiac surgery due to coronary artery disease (coronary artery bypass graft, CABG), valvular heart disease or both and of two body donors were examined. Tissue was fixed in 4 % paraformaldehyde, embedded in paraffin, dissected in consecutive sections and stained for c-kitpos cells. In parallel, grade of fibrosis, amount of fat per section and cells positive for mast cell tryptase were examined. Results We collected 27 LAAs (37.0 % female, mean left ventricular ejection fraction 50.4 %, 63.0 % persistent atrial fibrillation (AF)). Most of the patients underwent combined CABG and valve surgery (51.9 %). C-kitpos cells were detected in 3 different regions: A) Attached to the epicardial fat layer, B) close to vascular structures and C) between cardiomyocytes. C-kitpos cells ranged from 0.05 c-kitpos cells per mm2 to 67.5 c-kitpos cells per mm2. We found no association between number of c-kitpos cells and type of AF, amount of fibrosis or amount of fat. Up to 72 % of c-kitpos cells also showed a positive staining for mast cell tryptase. Conclusion C-kitpos cells are frequent in LAAs of cardiovascular patients with a rather homogenous distribution throughout the LAA. The LAA can therefore be considered as a source for extraction of a reasonable quantity of autologous cardiac progenitor cells in the living human patient.
Collapse
Affiliation(s)
- Lea Schwarzkopf
- St. Elisabeth-Krankenhaus Leipzig, Department of Anaesthesiology, Leipzig, Germany
- German Heart Center Berlin, Department of Electrophysiology, Berlin, Germany
| | - Petra Büttner
- Heart Center Leipzig at University of Leipzig, Department of Cardiology, Leipzig, Germany
| | - Karl Scholtyssek
- German Heart Center Berlin, Department of Electrophysiology, Berlin, Germany
| | - Thomas Schröter
- Heart Center Leipzig at University of Leipzig, Department of Cardiac Surgery, Leipzig, Germany
| | - Ruth Hiller
- Insitut für Pathologie, University of Leipzig Medical Center, Leipzig, Germany
| | - Gerhard Hindricks
- German Heart Center Berlin, Department of Electrophysiology, Berlin, Germany
- Leipzig Heart Institute, Leipzig, Germany
| | - Andreas Bollmann
- German Heart Center Berlin, Department of Electrophysiology, Berlin, Germany
- Leipzig Heart Institute, Leipzig, Germany
| | - Ulrich Laufs
- Klinik und Poliklinik für Kardiologie, University of Leipzig Medical Center, Leipzig, Germany
| | - Laura Ueberham
- German Heart Center Berlin, Department of Electrophysiology, Berlin, Germany
- Leipzig Heart Institute, Leipzig, Germany
- Klinik und Poliklinik für Kardiologie, University of Leipzig Medical Center, Leipzig, Germany
| |
Collapse
|
4
|
Ibrahim NA, Buabeid MA, Shaimaa Arafa E, Elmorshedy KE. Zinc's protective role against hydroxychloroquine-induced cardiac effects in adult male albino rats. Saudi J Biol Sci 2023; 30:103733. [PMID: 37521750 PMCID: PMC10374629 DOI: 10.1016/j.sjbs.2023.103733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 06/22/2023] [Accepted: 06/30/2023] [Indexed: 08/01/2023] Open
Abstract
Background Long exposure to Hydroxychloroquine (HCQ) has been complicated by some dangerous though infrequent cardiotoxicity. Methods A total of 40 normal adult male albino rats dispersed into 4 groups were used. Group 1 (Control group), Group II (HCQ treated group), Group III (zinc [Zn]-treated group), and Group IV (HCQ and Zn treated group). Once the experimentation ended, rats were sacrificed and cardiac soft tissue sections were processed twenty-four hours at the end of the experiment for histological study. Results Cardiac-stained sections revealed that HCQ induced widespread necrosis, dilatation, and vacuolar degeneration. However, the combination of HCQ with Zn ameliorated these damaging effects. Cardiac enzyme parameters were also studied in the 4 groups and revealed CK-MB and troponin were considerably elevated in groups II associated to the control group. Conclusion It was concluded that Zn revealed a protective role against HCQ cardiomyopathy in adult male albino rats. This might signify an appreciated means for Zn-based treatment in the upcoming subsequent clinical records to adjust doses and guarantee patient safeguard.
Collapse
Affiliation(s)
- Nihal A. Ibrahim
- Department of Clinical Sciences, College of Pharmacy and Health Sciences, Ajman University, UAE
- Centre of Medical and Bio-allied Health Sciences Research (CMBAHSR), Ajman University, Ajman, UAE
| | | | - El Shaimaa Arafa
- Department of Clinical Sciences, College of Pharmacy and Health Sciences, Ajman University, UAE
- Centre of Medical and Bio-allied Health Sciences Research (CMBAHSR), Ajman University, Ajman, UAE
| | | |
Collapse
|
5
|
Pogontke C, Guadix JA, Sánchez-Tévar AM, Muñoz-Chápuli R, Ruiz-Villalba A, Pérez-Pomares JM. Dynamic Epicardial Contribution to Cardiac Interstitial c-Kit and Sca1 Cellular Fractions. Front Cell Dev Biol 2022; 10:864765. [PMID: 35706902 PMCID: PMC9189417 DOI: 10.3389/fcell.2022.864765] [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: 01/28/2022] [Accepted: 04/25/2022] [Indexed: 12/02/2022] Open
Abstract
Background: The cardiac interstitial cellular fraction is composed of multiple cell types. Some of these cells are known to express some well-known stem cell markers such as c-Kit and Sca1, but they are no longer accepted to be true cardiac stem cells. Although their existence in the cardiac interstitium has not been disputed, their dynamic throughout development, specific embryonic origin, and potential heterogeneity remain unknown. In this study, we hypothesized that both c-KitPOS and Sca1POS cardiac interstitial cell (CIC) subpopulations are related to the Wilms’ tumor 1 (Wt1) epicardial lineage. Methods: In this study, we have used genetic cell lineage tracing methods, immunohistochemistry, and FACS techniques to characterize cardiac c-KitPOS and Sca1POS cells. Results: Our data show that approximately 50% of cardiac c-KitPOS cells are derived from the Wt1-lineage at E15.5. This subpopulation decreased along with embryonic development, disappearing from P7 onwards. We found that a large proportion of cardiac c-KitPOS cells express specific markers strongly suggesting they are blood-borne cells. On the contrary, the percentage of Sca1POS cells within the Wt1-lineage increases postnatally. In accordance with these findings, 90% of adult epicardial-derived endothelial cells and 60% of mEFSK4POS cardiac fibroblasts expressed Sca1. Conclusion: Our study revealed a minor contribution of the Wt1-epicardial lineage to c-KitPOS CIC from embryonic stages to adulthood. Remarkably, a major part of the adult epicardial-derived cell fraction is enriched in Sca1, suggesting that this subpopulation of CICs is heterogeneous from their embryonic origin. The study of this heterogeneity can be instrumental to the development of diagnostic and prognostic tests for the evaluation of cardiac homeostasis and cardiac interstitium response to pathologic stimuli.
Collapse
Affiliation(s)
- C. Pogontke
- Department of Animal Biology, Faculty of Sciences, University of Málaga, Málaga, Spain
- Instituto Malagueño de Biomedicina (IBIMA)-Plataforma BIONAND, Universidad de Málaga, Málaga, Spain
| | - J. A. Guadix
- Department of Animal Biology, Faculty of Sciences, University of Málaga, Málaga, Spain
- Instituto Malagueño de Biomedicina (IBIMA)-Plataforma BIONAND, Universidad de Málaga, Málaga, Spain
| | - A. M. Sánchez-Tévar
- Department of Animal Biology, Faculty of Sciences, University of Málaga, Málaga, Spain
- Instituto Malagueño de Biomedicina (IBIMA)-Plataforma BIONAND, Universidad de Málaga, Málaga, Spain
| | - R. Muñoz-Chápuli
- Department of Animal Biology, Faculty of Sciences, University of Málaga, Málaga, Spain
| | - A. Ruiz-Villalba
- Department of Animal Biology, Faculty of Sciences, University of Málaga, Málaga, Spain
- Instituto Malagueño de Biomedicina (IBIMA)-Plataforma BIONAND, Universidad de Málaga, Málaga, Spain
- *Correspondence: A. Ruiz-Villalba, ; J. M. Pérez-Pomares,
| | - J. M. Pérez-Pomares
- Department of Animal Biology, Faculty of Sciences, University of Málaga, Málaga, Spain
- Instituto Malagueño de Biomedicina (IBIMA)-Plataforma BIONAND, Universidad de Málaga, Málaga, Spain
- *Correspondence: A. Ruiz-Villalba, ; J. M. Pérez-Pomares,
| |
Collapse
|
6
|
Odintsova IA, Slutskaya DR, Berezovskaya TI. Telocytes: localization, structure, functions and significance in pathology. GENES & CELLS 2022; 17:6-12. [DOI: 10.23868/202205001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
The review article presents the material analyzing and summarizing the information about the localization, ultramicroscopic structure, functions and significance in pathological processes of newly discovered cells - telocytes. The information about molecular markers of telocytes are considered. The questions about the tissue belonging of these cells are discussed, the relevance of studying their role in the development of pathological processes in different organs is emphasized. The analysis of literature data was carried out taking into account the fundamental provisions of the doctrine of the cell-differon organization of tissues.
Collapse
|
7
|
Ramirez-GarciaLuna JL, Rangel-Berridi K, Olasubulumi OO, Rosenzweig DH, Henderson JE, Gawri R, Martineau PA. Enhanced Bone Remodeling After Fracture Priming. Calcif Tissue Int 2022; 110:349-366. [PMID: 34668029 DOI: 10.1007/s00223-021-00921-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 10/01/2021] [Indexed: 11/30/2022]
Abstract
The immune system is an active component of bone repair. Mast cells influence the recruitment of macrophages, osteoclasts and blood vessels into the repair tissue. We hypothesized that if mast cells and other immune cells are sensitized to recognize broken bone, they will mount an increased response to subsequent fractures that may be translated into enhanced healing. To test this, we created a bone defect on the left leg of anesthetized mice and 2 weeks later, a second one on the right leg. Bone repair in the right legs was then compared to control mice that underwent the creation of bilateral window bone defects at the same time. Mice were euthanized at 14 and 56 days. Mineralized tissue quantity and morphometric parameters were assessed using micro-CT and histology. The activity of osteoblasts, osteoclasts, vascular endothelial cells, mast cells, and macrophages was evaluated using histochemistry. Our main findings were (1) no significant differences in the amount of bone produced at 14- or 56 days post-operative between groups; (2) mice exposed to subsequent fractures showed significantly better bone morphometric parameters after 56 days post-operative; and (3) significant increases in the content of blood vessels, osteoclasts, and the number of macrophages in the subsequent fracture group. Our results provide strong evidence that a transient increase in the inflammatory state of a healing injury promotes faster bone remodelling and increased neo-angiogenesis. This phenomenon is also characterized by changes in mast cell and macrophage content that translate into more active recruitment of mesenchymal stromal cells.
Collapse
Affiliation(s)
- Jose L Ramirez-GarciaLuna
- Bone Engineering Labs, Injury, Repair & Recovery Program, Research Institute, McGill University Health Centre, 1650 Cedar Ave., Montreal, QC, H3G 1A4, Canada
- Experimental Surgery, Faculty of Medicine, McGill University, 3605 Rue de la Montagne, Montreal, QC, H3G 2M1, Canada
| | - Karla Rangel-Berridi
- Bone Engineering Labs, Injury, Repair & Recovery Program, Research Institute, McGill University Health Centre, 1650 Cedar Ave., Montreal, QC, H3G 1A4, Canada
- Biofabrication and Bioengineering Labs, Injury, Repair & Recovery Program, Research Institute, McGill University Health Centre, 1650 Cedar Ave., Montreal, QC, H3G 1A4, Canada
- Experimental Surgery, Faculty of Medicine, McGill University, 3605 Rue de la Montagne, Montreal, QC, H3G 2M1, Canada
| | - Ore-Oluwa Olasubulumi
- Bone Engineering Labs, Injury, Repair & Recovery Program, Research Institute, McGill University Health Centre, 1650 Cedar Ave., Montreal, QC, H3G 1A4, Canada
| | - Derek H Rosenzweig
- Biofabrication and Bioengineering Labs, Injury, Repair & Recovery Program, Research Institute, McGill University Health Centre, 1650 Cedar Ave., Montreal, QC, H3G 1A4, Canada
- Experimental Surgery, Faculty of Medicine, McGill University, 3605 Rue de la Montagne, Montreal, QC, H3G 2M1, Canada
| | - Janet E Henderson
- Bone Engineering Labs, Injury, Repair & Recovery Program, Research Institute, McGill University Health Centre, 1650 Cedar Ave., Montreal, QC, H3G 1A4, Canada
- Experimental Surgery, Faculty of Medicine, McGill University, 3605 Rue de la Montagne, Montreal, QC, H3G 2M1, Canada
- Experimental Medicine, Faculty of Medicine, McGill University, 3605 Rue de la Montagne, Montreal, QC, H3G 2M1, Canada
| | - Rahul Gawri
- Regenerative Orthopaedics and Innovation Laboratory, Injury, Repair & Recovery Program, Research Institute-McGill University Health Centre, 1650 Cedar Ave., Montreal, QC, H3G 1A4, Canada.
- Experimental Surgery, Faculty of Medicine, McGill University, 3605 Rue de la Montagne, Montreal, QC, H3G 2M1, Canada.
| | - Paul A Martineau
- Bone Engineering Labs, Injury, Repair & Recovery Program, Research Institute, McGill University Health Centre, 1650 Cedar Ave., Montreal, QC, H3G 1A4, Canada
- Regenerative Orthopaedics and Innovation Laboratory, Injury, Repair & Recovery Program, Research Institute-McGill University Health Centre, 1650 Cedar Ave., Montreal, QC, H3G 1A4, Canada
- Experimental Surgery, Faculty of Medicine, McGill University, 3605 Rue de la Montagne, Montreal, QC, H3G 2M1, Canada
| |
Collapse
|
8
|
Grigorev I, Korzhevskii D. Modern Imaging Technologies of Mast Cells for Biology and Medicine (Review). Sovrem Tekhnologii Med 2021; 13:93-107. [PMID: 34603768 PMCID: PMC8482833 DOI: 10.17691/stm2021.13.4.10] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Indexed: 01/03/2023] Open
Abstract
Mast cells play an important role in the body defense against allergens, pathogens, and parasites by participating in inflammation development. However, there is evidence for their contributing to the pathogenesis of a number of atopic, autoimmune, as well as cardiovascular, oncologic, neurologic, and other diseases (allergy, asthma, eczema, rhinitis, anaphylaxis, mastocytosis, multiple sclerosis, rheumatoid arthritis, inflammatory gastrointestinal and pulmonary diseases, migraine, etc.). The diagnosis of many diseases and the study of mast cell functions in health and disease require their identification; so, the knowledge on adequate imaging techniques for mast cells in humans and different species of animals is of particular importance. The present review summarizes the data on major methods of mast cell imaging: enzyme histochemistry, immunohistochemistry, as well as histochemistry using histological stains. The main histological stains bind to heparin and other acidic mucopolysaccharides contained in mast cells and stain them metachromatically. Among these are toluidine blue, methylene blue (including that contained in May-Grünwald-Giemsa stain), thionin, pinacyanol, and others. Safranin and fluorescent dyes: berberine and avidin - also bind to heparin. Longer staining with histological dyes or alcian blue staining is needed to label mucosal and immature mast cells. Advanced techniques - enzyme histochemistry and especially immunohistochemistry - enable to detect mast cells high-selectively using a reaction to tryptases and chymases (specific proteases of these cells). In the immunohistochemical study of tryptases and chymases, species-specific differences in the distribution of the proteases in mast cells of humans and animals should be taken into account for their adequate detection. The immunohistochemical reaction to immunoglobulin E receptor (FcεRI) and c-kit receptor is not specific to mast cells, although the latter is important to demonstrate their proliferation in normal and malignant growth. Correct fixation of biological material is also discussed in the review as it is of great significance for histochemical and immunohistochemical mast cell detection. Fluorescent methods of immunohistochemistry and a multimarker analysis in combination with confocal microscopy are reported to be new technological approaches currently used to study various mast cell populations.
Collapse
Affiliation(s)
- I.P. Grigorev
- Senior Researcher, Laboratory of Functional Morphology of the Central and Peripheral Nervous System, Department of General and Specific Morphology; Institute of Experimental Medicine, 12 Akademika Pavlova St., Saint Petersburg, 197376, Russia
| | - D.E. Korzhevskii
- Professor of the Russian Academy of Sciences, Head of the Laboratory of Functional Morphology of the Central and Peripheral Nervous System, Department of General and Specific Morphology; Institute of Experimental Medicine, 12 Akademika Pavlova St., Saint Petersburg, 197376, Russia
| |
Collapse
|
9
|
Characterization of encapsulated porcine cardiosphere-derived cells embedded in 3D alginate matrices. Int J Pharm 2021; 599:120454. [PMID: 33676988 DOI: 10.1016/j.ijpharm.2021.120454] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 02/23/2021] [Accepted: 03/01/2021] [Indexed: 12/22/2022]
Abstract
Myocardial infarction is caused by an interruption of coronary blood flow, leading to one of the main death causes worldwide. Current therapeutic approaches are palliative and not able to solve the loss of cardiac tissue. Cardiosphere derived cells (CDCs) reduce scarring, and increase viable myocardium, with safety and adequate biodistribution, but show a low rate engraftment and survival after implantation. In order to solve the low retention, we propose the encapsulation of CDCs within three-dimensional alginate-poly-L-lysine-alginate matrix as therapy for cardiac regeneration. In this work, we demonstrate the encapsulation of CDCs in alginate matrix, with no decrease in viability over a month, and showing the preservation of CDCs phenotype, differentiation potential, gene expression profile and growth factor release after encapsulation, moving a step forward to clinical translation of CDCs therapy in regeneration in heart failure.
Collapse
|
10
|
Selviler-Sizer S, Kabak YB, Kabak M. Telocytes in the hearts of Saanen goats. Microsc Res Tech 2020; 84:548-554. [PMID: 33017500 DOI: 10.1002/jemt.23612] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 09/10/2020] [Accepted: 09/19/2020] [Indexed: 12/21/2022]
Abstract
Telocytes, new interstitial cells that have received significant attention in recent years, have been detected in many organs, including the heart. The distinction between telocytes and other interstitial cells can only be made based on their ultrastructural characterization and immunophenotypic features. In this study, we examined the interstitial cells in the healthy heart tissues of Saanen goats to determine whether they are telocytes or not, by using a scanning electron microscope (SEM) and immunohistochemical and immunofluorescence staining methods. The SEM revealed oval and round telocytes with two to four telopodes. Some telopodes also had podoms. The staining for immunohistochemical and immunofluorescence methods used for CD34, c-kit (CD117), and vimentin antibodies. Positive cells were detected in the heart muscle and heart valves by immunohistochemical staining. As these antigens can also be expressed by other non-telocyte cells, we used double immunofluorescence staining with CD34/c-kit and CD34/vimentin antibodies to identify true telocytes. Telocytes were determined in the right atrium and aortic valve. While telocytes were CD34+/c-kit+ and CD34+/vimentin+, fibroblasts were CD34-/vimentin+. These results confirm the presence of telocytes in the hearts of Saanen goats.
Collapse
Affiliation(s)
- Sedef Selviler-Sizer
- Department of Anatomy, Faculty of Veterinary Medicine, Ondokuz Mayis University, Samsun, Turkey
| | - Yonca Betil Kabak
- Department of Pathology, Faculty of Veterinary Medicine, Ondokuz Mayis University, Samsun, Turkey
| | - Murat Kabak
- Department of Anatomy, Faculty of Veterinary Medicine, Ondokuz Mayis University, Samsun, Turkey
| |
Collapse
|
11
|
Motta Junior JDS, Miggiolaro AFRDS, Nagashima S, de Paula CBV, Baena CP, Scharfstein J, de Noronha L. Mast Cells in Alveolar Septa of COVID-19 Patients: A Pathogenic Pathway That May Link Interstitial Edema to Immunothrombosis. Front Immunol 2020; 11:574862. [PMID: 33042157 PMCID: PMC7530169 DOI: 10.3389/fimmu.2020.574862] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 08/27/2020] [Indexed: 12/22/2022] Open
Abstract
It is currently believed that innate immunity is unable to prevent the spread of SARS-CoV-2 from the upper airways to the alveoli of high-risk groups of patients. SARS-CoV-2 replication in ACE-2-expressing pneumocytes can drive the diffuse alveolar injury through the cytokine storm and immunothrombosis by upregulating the transcription of chemokine/cytokines, unlike several other respiratory viruses. Here we report histopathology data obtained in post-mortem lung biopsies of COVID-19, showing the increased density of perivascular and septal mast cells (MCs) and IL-4-expressing cells (n = 6), in contrast to the numbers found in pandemic H1N1-induced pneumonia (n = 10) or Control specimens (n = 10). Noteworthy, COVID-19 lung biopsies showed a higher density of CD117+ cells, suggesting that c-kit positive MCs progenitors were recruited earlier to the alveolar septa. These findings suggest that MC proliferation/differentiation in the alveolar septa might be harnessed by the shift toward IL-4 expression in the inflamed alveolar septa. Future studies may clarify whether the fibrin-dependent generation of the hyaline membrane, processes that require the diffusion of procoagulative plasma factors into the alveolar lumen and the endothelial dysfunction, are preceded by MC-driven formation of interstitial edema in the alveolar septa.
Collapse
Affiliation(s)
- Jarbas da Silva Motta Junior
- School of Medicine, Pontifícia Universidade Católica do Paraná PUCPR, Curitiba, Brazil
- Hospital Marcelino Champagnat, Curitiba, Brazil
| | | | - Seigo Nagashima
- School of Medicine, Pontifícia Universidade Católica do Paraná PUCPR, Curitiba, Brazil
| | | | - Cristina Pellegrino Baena
- School of Medicine, Pontifícia Universidade Católica do Paraná PUCPR, Curitiba, Brazil
- Hospital Marcelino Champagnat, Curitiba, Brazil
| | - Julio Scharfstein
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Lucia de Noronha
- School of Medicine, Pontifícia Universidade Católica do Paraná PUCPR, Curitiba, Brazil
| |
Collapse
|
12
|
Kotov G, Landzhov B, Stamenov N, Stanchev S, Iliev A. Changes in the number of mast cells, expression of fibroblast growth factor-2 and extent of interstitial fibrosis in established and advanced hypertensive heart disease. Ann Anat 2020; 232:151564. [PMID: 32603827 DOI: 10.1016/j.aanat.2020.151564] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 06/12/2020] [Accepted: 06/17/2020] [Indexed: 12/16/2022]
Abstract
INTRODUCTION An increasing number of studies have shed light on the role of cardiac mast cells in the pathogenesis of hypertension-induced myocardial remodeling. Mast cells promote fibroblast activation, myofibroblast differentiation and subsequent collagen accumulation through the action of tryptase, chymase, histamine and fibroblast growth factor-2. The aim of the present study was to report on the changes in the number of mast cells as evaluated through toluidine blue, tryptase and c-kit staining, to assess the extent of interstitial fibrosis and correlate it with the changes in the number of mast cells and to analyze the immunohistochemical expression of fibroblast growth factor-2 in two groups of spontaneously hypertensive rats indicative of established and advanced hypertensive heart disease. A novel aspect of our work was the analysis of all parameters in the right ventricle. MATERIAL AND METHODS For the present study, we used 6- and 12-month-old spontaneously hypertensive rats. A light microscopic study was conducted on sections stained with hematoxylin and eosin and toluidine blue. For the immunohistochemical study we used monoclonal antibodies against mast cell tryptase and fibroblast growth factor-2 and a polyclonal antibody against c-kit. The expression of fibroblast growth factor-2 was assessed semi-quantitatively through ImageJ. The number of mast cells was evaluated on toluidine blue-, tryptase- and c-kit-stained sections and a comparative statistical analysis with the Mann-Whitney test was conducted between the two age groups. A separate statistical analysis between results obtained through immunostaining for tryptase and for c-kit was conducted in each age group with the Wilcoxon signed-rank test. The extent of fibrosis was assessed quantitatively on slides stained with Mallory's trichrome stain as a percentage of the whole tissue and compared between the two age groups. Spearman's correlation was used to test whether a correlation exists between the number of mast cells and the percentage of interstitial fibrosis. RESULTS Mast cells with typical cytoplasmic granules were visualized in the interstitial tissue and in the perivascular zone in both age groups. In both ventricles, their number increased significantly in 12-month-old animals as evaluated through all three staining methods. Moreover, immunostaining for tryptase and for c-kit yielded comparable results. The immunoreactivity of fibroblast growth factor-2 increased in both ventricles in older animals. Expression of this protein was particularly intensive in the cytoplasm of connective tissue cells with the characteristic features of mast cells mainly found in the areas of fibrotic alterations in 12-month-old spontaneously hypertensive rats. In both ventricles, interstitial fibrosis was more extensive throughout the myocardium of older animals and was positively correlated with the changes in the number of mast cells in both age groups. CONCLUSION The present study reported for the first time that the increase in the number of mast cells, observed as hypertension-induced myocardial changes progress, is statistically significant and confirmed that this process takes place in both ventricles. This increase is accompanied by a higher expression of fibroblast growth factor-2 and is more strongly correlated with the more pronounced interstitial fibrosis in older animals, further supporting the role of mast cells in the structural changes taking place in the myocardium in response to systemic hypertension.
Collapse
Affiliation(s)
- Georgi Kotov
- Department of Anatomy, Histology and Embryology, Medical University of Sofia, Bulgaria.
| | - Boycho Landzhov
- Department of Anatomy, Histology and Embryology, Medical University of Sofia, Bulgaria
| | - Nikola Stamenov
- Department of Anatomy, Histology and Embryology, Medical University of Sofia, Bulgaria
| | - Stancho Stanchev
- Department of Anatomy, Histology and Embryology, Medical University of Sofia, Bulgaria
| | - Alexandar Iliev
- Department of Anatomy, Histology and Embryology, Medical University of Sofia, Bulgaria
| |
Collapse
|
13
|
Rozsívalová K, Pierzynová A, Kratochvílová H, Lindner J, Lipš M, Kotulák T, Ivák P, Netuka I, Haluzík M, Kučera T. Increased Number of Mast Cells in Epicardial Adipose Tissue of Cardiac Surgery Patients With Coronary Artery Disease. Physiol Res 2020; 69:621-631. [PMID: 32584133 DOI: 10.33549/physiolres.934344] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Chronic inflammation of adipose tissue is associated with the pathogenesis of cardiovascular diseases. Mast cells represent an important component of the innate defense system of the organism. In our work, we quantified mast cell number in epicardial adipose tissue (EAT), subcutaneous adipose tissue (SAT), and right atrial myocardium (RA) in patients undergoing open heart surgery (n=57). Bioptic samples of EAT (n=44), SAT (n=42) and RA (n=17) were fixed by 4 % paraformaldehyde and embedded into paraffin. An anti-mast cell tryptase antibody was used for immunohistochemical detection and quantification of mast cells. We also demonstrated immunohistochemically the expression of CD117 and chymase markers. In EAT of patients with coronary artery disease (CAD), higher incidence of mast cells has been found compared to patients without CAD (3.7±2.6 vs. 2.1±1.2 cells/mm(2)). In SAT and RA, there was no difference in the number of mast cells in CAD and non-CAD patients. Mast cells in SAT, EAT and RA expressed CD117 and chymase. An increased incidence of mast cells in EAT of CAD patients may indicate the specific role of these inflammatory cells in relation to EAT and coronary arteries affected by atherosclerosis.
Collapse
Affiliation(s)
- K Rozsívalová
- Institute of Histology and Embryology, First Faculty of Medicine, Charles University, Prague, Czech Republic.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
DeSimone CV, McLeod CJ, Gomez Pinilla PJ, Beyder A, Farrugia G, Asirvatham SJ, Kapa S. Telocytes express ANO-1-encoded chloride channels in canine ventricular myocardium. J Arrhythm 2019; 35:515-521. [PMID: 31293701 PMCID: PMC6595329 DOI: 10.1002/joa3.12176] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Revised: 01/30/2019] [Accepted: 02/22/2019] [Indexed: 12/17/2022] Open
Abstract
INTRODUCTION It is unknown if ANO-1 is expressed in the heart, though the presence of a calcium-activated chloride current has been proposed to mediate some cardiac dysrhythmias. Furthermore, a specific cell type termed telocytes, morphologically mimicking Cajal cells which use ANO-1 to modulate their pacemaker activity in the gut, have been described in the heart. We therefore sought to determine whether this channel is expressed in the canine heart. METHODS Myocardium was sampled from the ventricles of five canines. Sections were labeled with anti-Kit and anti-ANO-1 antibodies. Slides were reviewed by four investigators looking at cell morphology, distribution, and co-localization. Identification of telocytes was based on criteria including morphology, Kit positivity (+), and ANO-1 positivity (+). RESULTS Clusters of cells meeting criteria for telocytes were seen in the epicardium, sub-epicardium, and mid-myocardium. A small subset of cells that were morphologically similar to myocytes was ANO-1 (+) but Kit (-). In total, three different cell classes were found: (i) Kit (+), ANO-1 (+) cells with the appearance of telocytes; (ii) Kit (+), ANO-1 (-) cells; and (iii) Kit (-), ANO-1 (+) cells with the morphologic appearance of cardiac myocytes. CONCLUSIONS Telocytes are present in the canine ventricle and express ANO-1. These data merit further study to elucidate the functional expression of these channels in the heart and whether they may be targets for cardiac arrhythmias.
Collapse
Affiliation(s)
| | | | | | - Arthur Beyder
- Division of GastroenterologyDepartment of Internal MedicineMayo ClinicRochesterMNUSA
| | - Gianrico Farrugia
- Division of GastroenterologyDepartment of Internal MedicineMayo ClinicRochesterMNUSA
| | - Samuel J. Asirvatham
- Division of CardiologyDepartment of Internal MedicineMayo ClinicRochesterMNUSA
- Division of Pediatric CardiologyDepartment of Pediatrics and Adolescent MedicineMayo ClinicRochesterMNUSA
| | - Suraj Kapa
- Division of CardiologyDepartment of Internal MedicineMayo ClinicRochesterMNUSA
| |
Collapse
|
15
|
Abstract
A great interest has developed over the last several years in research on interstitial Cajal-like cells (ICLCs), later renamed to telocytes (TCs). Such studies are restricted by diverse limitations. We aimed to critically review (sub)epicardial ICLCs/TCs and to bring forward supplemental immunohistochemical evidence on (sub)epicardial stromal niche inhabitants. We tested the epicardial expressions of CD117/c-kit, CD34, Cytokeratin 7 (CK7), Ki67, Platelet-Derived Growth Factor Receptor (PDGFR)-α and D2-40 in adult human cardiac samples. The mesothelial epicardial cells expressed D2-40, CK7, CD117/c-kit and PDGFR-α. Subepicardial D2-40-positive lymphatic vessels and isolated D2-40-positive and CK7-positive subepicardial cells were also found. Immediate submesothelial spindle-shaped cells expressed Ki-67. Submesothelial stromal cells and endothelial tubes were PDGFR-α-positive and CD34-positive. The expression of CD34 was pan-stromal, so a particular stromal cell type could not be distinguished. The stromal expression of CD117/c-kit was also noted. It seems that epicardial TCs could not be regarded as belonging to a unique cell type until (pre)lymphatic endothelial cells are inadequately excluded. Markers such as CD117/c-kit or CD34 seem to be improper for identifying TCs as a distinctive cell type. Care should be taken when using the immunohistochemical method and histological interpretations, as they may not produce accurate results.
Collapse
|
16
|
Legere SA, Haidl ID, Légaré JF, Marshall JS. Mast Cells in Cardiac Fibrosis: New Insights Suggest Opportunities for Intervention. Front Immunol 2019; 10:580. [PMID: 31001246 PMCID: PMC6455071 DOI: 10.3389/fimmu.2019.00580] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 03/04/2019] [Indexed: 12/19/2022] Open
Abstract
Mast cells (MC) are innate immune cells present in virtually all body tissues with key roles in allergic disease and host defense. MCs recognize damage-associated molecular patterns (DAMPs) through expression of multiple receptors including Toll-like receptors and the IL-33 receptor ST2. MCs can be activated to degranulate and release pre-formed mediators, to synthesize and secrete cytokines and chemokines without degranulation, and/or to produce lipid mediators. MC numbers are generally increased at sites of fibrosis. They are potent, resident, effector cells producing mediators that regulate the fibrotic process. The nature of the secretory products produced by MCs depend on micro-environmental signals and can be both pro- and anti-fibrotic. MCs have been repeatedly implicated in the pathogenesis of cardiac fibrosis and in angiogenic responses in hypoxic tissues, but these findings are controversial. Several rodent studies have indicated a protective role for MCs. MC-deficient mice have been reported to have poorer outcomes after coronary artery ligation and increased cardiac function upon MC reconstitution. In contrast, MCs have also been implicated as key drivers of fibrosis. MC stabilization during a hypertensive rat model and an atrial fibrillation mouse model rescued associated fibrosis. Discrepancies in the literature could be related to problems with mouse models of MC deficiency. To further complicate the issue, mice generally have a much lower density of MCs in their cardiac tissue than humans, and as such comparing MC deficient and MC containing mouse models is not necessarily reflective of the role of MCs in human disease. In this review, we will evaluate the literature regarding the role of MCs in cardiac fibrosis with an emphasis on what is known about MC biology, in this context. MCs have been well-studied in allergic disease and multiple pharmacological tools are available to regulate their function. We will identify potential opportunities to manipulate human MC function and the impact of their mediators with a view to preventing or reducing harmful fibrosis. Important therapeutic opportunities could arise from increased understanding of the impact of such potent, resident immune cells, with the ability to profoundly alter long term fibrotic processes.
Collapse
Affiliation(s)
- Stephanie A. Legere
- Departments of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
| | - Ian D. Haidl
- Departments of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
| | - Jean-François Légaré
- Department of Pathology, Dalhousie University, Halifax, NS, Canada
- Department of Surgery, Dalhousie Medicine New Brunswick, Saint John, NB, Canada
| | - Jean S. Marshall
- Departments of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
- Department of Pathology, Dalhousie University, Halifax, NS, Canada
| |
Collapse
|
17
|
Iancu CB, Rusu MC, Mogoantă L, Hostiuc S, Grigoriu M. Myocardial Telocyte-Like Cells: A Review Including New Evidence. Cells Tissues Organs 2019; 206:16-25. [PMID: 30879002 DOI: 10.1159/000497194] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Accepted: 01/17/2019] [Indexed: 11/19/2022] Open
Abstract
Telocytes (TCs) are a controversial cell type characterized by the presence of a particular kind of prolongations, known as telopodes, which are long, thin, and moniliform. A number of attempts has been made to establish the molecular phenotype of cardiac TCs (i.e., expression of c-kit, CD34, vimentin, PDGRFα, PDGRFβ, etc.). We designed an immunohistochemical study involving cardiac tissue samples obtained from 10 cadavers with the aim of determining whether there are TC-like interstitial cells that populate the interstitial space other than the mural microvascular cells. We applied the markers for CD31, CD34, PDGRFα, CD117/c-kit, and α-smooth muscle actin (α-SMA). We found that, in relation to two-dimensional cuts, the endothelial tubes could be misidentified as TC-like cells, the difference being the positive identification of endothelial lumina. Moreover, we found that cardiac pericytes express PDGRFα, CD117/c-kit, and α-SMA, and that they could also be misidentified as TCs when using light microscopy. We reviewed the respective values of the previously identified markers for achieving a clear-cut identification of cardiac TCs, highlighting the critical lack of specificity.
Collapse
Affiliation(s)
- Cristian B Iancu
- Division of Anatomy, Faculty of Dental Medicine, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Mugurel C Rusu
- Division of Anatomy, Faculty of Dental Medicine, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania,
| | - Laurenţiu Mogoantă
- Department of Histology, University of Medicine and Pharmacy Craiova, Craiova, Romania
| | - Sorin Hostiuc
- Department of Legal Medicine and Bioethics, Faculty of Dental Medicine, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Mihai Grigoriu
- Division of Surgery, University Emergency Hospital Bucharest, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| |
Collapse
|
18
|
López-Giraldo A, Cruz T, Molins L, Guirao Á, Saco A, Cuerpo S, Ramirez J, Agustí Á, Faner R. Characterization, localization and comparison of c-Kit+ lung cells in never smokers and smokers with and without COPD. BMC Pulm Med 2018; 18:123. [PMID: 30064386 PMCID: PMC6066937 DOI: 10.1186/s12890-018-0688-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 07/10/2018] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND c-Kit + lung stem cells have been described in the human healthy lung. Their potential relation with smoking and/or chronic obstructive pulmonary disease (COPD) is unknown. METHODS We characterized and compared c-Kit+ cells in lung tissue of 12 never smokers (NS), 15 smokers with normal spirometry (S) and 44 COPD patients who required lung resectional surgery. Flow cytometry (FACS) was used to characterize c-Kit+ cells in fresh lung tissue disaggregates, and immunofluorescence (IF) for further characterization and to determine their location in OCT- embedded lung tissue. RESULTS We identified 4 c-Kit+ cell populations, with similar proportions in NS, S and COPD: (1) By FACS, c-Kithigh/CD45+ cells (4.03 ± 2.97% (NS), 3.96 ± 5.30% (S), and 5.20 ± 3.44% (COPD)). By IF, these cells were tryptase+ (hence, mast cells) and located around the airways; (2) By IF, c-Kitlow/CD45+/triptase- (0.07 ± 0.06 (NS), 0.03 ± 0.02 (S), and 0.06 ± 0.07 (COPD) cells/field), which likely correspond to innate lymphoid cells; (3) By FACS, c-Kitlow/CD45-/CD34+ (0.95 ± 0.84% (NS), 1.14 ± 0.94% (S) and 0.95 ± 1.38% (COPD)). By IF these cells were c-Kitlow/CD45-/CD31+, suggesting an endothelial lineage, and were predominantly located in the alveolar wall; and, (4) by FACS, an infrequent c-Kitlow/CD45-/CD34- population (0.09 ± 0.14% (NS), 0.08 ± 0.09% (S) and 0.08 ± 0.11% (COPD)) compatible with a putative lung stem cell population. Yet, IF failed to detect them and we could not isolate or grow them, thus questioning the existence of c-Kit+ lung stem-cells. CONCLUSIONS The adult human lung contains a mixture of c-Kit+ cells, unlikely to be lung stem cells, which are independent of smoking status and/or presence of COPD.
Collapse
Affiliation(s)
- Alejandra López-Giraldo
- Respiratory Institute, Hospital Clinic, University of Barcelona, Barcelona, Spain.,Institut d'investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,CIBER Enfermedades Respiratorias(CIBERES), Instituto de Salud Carlos III, Madrid, Spain
| | - Tamara Cruz
- Institut d'investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,CIBER Enfermedades Respiratorias(CIBERES), Instituto de Salud Carlos III, Madrid, Spain
| | - Laureano Molins
- Respiratory Institute, Hospital Clinic, University of Barcelona, Barcelona, Spain.,Institut d'investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Ángela Guirao
- Respiratory Institute, Hospital Clinic, University of Barcelona, Barcelona, Spain.,Institut d'investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Adela Saco
- Institut d'investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Department of Pathology, Hospital Clinic, Barcelona, Spain
| | - Sandra Cuerpo
- Respiratory Institute, Hospital Clinic, University of Barcelona, Barcelona, Spain.,Institut d'investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,CIBER Enfermedades Respiratorias(CIBERES), Instituto de Salud Carlos III, Madrid, Spain
| | - Josep Ramirez
- Institut d'investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Department of Pathology, Hospital Clinic, Barcelona, Spain
| | - Álvar Agustí
- Respiratory Institute, Hospital Clinic, University of Barcelona, Barcelona, Spain.,Institut d'investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,CIBER Enfermedades Respiratorias(CIBERES), Instituto de Salud Carlos III, Madrid, Spain
| | - Rosa Faner
- Institut d'investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain. .,CIBER Enfermedades Respiratorias(CIBERES), Instituto de Salud Carlos III, Madrid, Spain. .,, Barcelona, Spain.
| |
Collapse
|
19
|
Varga I, Kyselovič J, Galfiova P, Danisovic L. The Non-cardiomyocyte Cells of the Heart. Their Possible Roles in Exercise-Induced Cardiac Regeneration and Remodeling. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 999:117-136. [PMID: 29022261 DOI: 10.1007/978-981-10-4307-9_8] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The non-cardiomyocyte cellular microenvironment of the heart includes diverse types of cells of mesenchymal origin. During development, the majority of these cells derive from the epicardium, while a subset derives from the endothelium/endocardium and neural crest derived mesenchyme. This subset includes cardiac fibroblasts and telocytes, the latter of which are a controversial type of "connecting cell" that support resident cardiac progenitors in the postnatal heart. Smooth muscle cells, pericytes, and endothelial cells are also present, in addition to adipocytes, which accumulate as epicardial adipose connective tissue. Furthermore, the heart harbors many cells of hematopoietic origin, such as mast cells, macrophages, and other immune cell populations. Most of these control immune reactions and inflammation. All of the above-mentioned non-cardiomyocyte cells of the heart contribute to this organ's well-orchestrated physiology. These cells also contribute to regeneration as a result of injury or age, in addition to tissue remodeling triggered by chronic disease or increased physical activity (exercise-induced cardiac growth). These processes in the heart, the most important vital organ in the human body, are not only fascinating from a scientific standpoint, but they are also clinically important. It is well-known that regular exercise can help prevent many cardiovascular diseases. However, the precise mechanisms underpinning myocardial remodeling triggered by physical activity are still unknown. Surprisingly, exercise-induced adaptation mechanisms are often identical or very similar to tissue remodeling caused by pathological conditions, such as hypertension, cardiac hypertrophy, and cardiac fibrosis. This review provides a summary of our current knowledge regarding the cardiac cellular microenvironment, focusing on the clinical applications this information to the study of heart remodeling during regular physical exercise.
Collapse
Affiliation(s)
- Ivan Varga
- Institute of Histology and Embryology, Faculty of Medicine, Comenius University, Bratislava, Slovak Republic.
| | - Jan Kyselovič
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University Bratislava, Bratislava, Slovak Republic
| | - Paulina Galfiova
- Institute of Histology and Embryology, Faculty of Medicine, Comenius University, Bratislava, Slovak Republic
| | - Lubos Danisovic
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, Bratislava, Slovak Republic
| |
Collapse
|
20
|
Gerisch M, Smettan J, Ebert S, Athelogou M, Brand-Saberi B, Spindler N, Mueller WC, Giri S, Bader A. Qualitative and Quantitative Analysis of Cardiac Progenitor Cells in Cases of Myocarditis and Cardiomyopathy. Front Genet 2018; 9:72. [PMID: 29559994 PMCID: PMC5845648 DOI: 10.3389/fgene.2018.00072] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 02/16/2018] [Indexed: 11/24/2022] Open
Abstract
We aimed to identify and quantify CD117+ and CD90+ endogenous cardiac progenitor cells (CPC) in human healthy and diseased hearts. We hypothesize that these cells perform a locally acting, contributing function in overcoming medical conditions of the heart by endogenous means. Human myocardium biopsies were obtained from 23 patients with the following diagnoses: Dilatative cardiomyopathy (DCM), ischemic cardiomyopathy (ICM), myocarditis, and controls from healthy cardiac patients. High-resolution scanning microscopy of the whole slide enabled a computer-based immunohistochemical quantification of CD117 and CD90. Those signals were evaluated by Definiens Tissue Phenomics® Technology. Co-localization of CD117 and CD90 was determined by analyzing comparable serial sections. CD117+/CD90+ cardiac cells were detected in all biopsies. The highest expression of CD90 was revealed in the myocarditis group. CD117 was significantly higher in all patient groups, compared to healthy specimens (*p < 0.05). The highest co-expression was found in the myocarditis group (6.75 ± 3.25 CD90+CD117+ cells/mm2) followed by ICM (4 ± 1.89 cells/mm2), DCM (1.67 ± 0.58 cells/mm2), and healthy specimens (1 ± 0.43 cells/mm2). We conclude that the human heart comprises a fraction of local CD117+ and CD90+ cells. We hypothesize that these cells are part of local endogenous progenitor cells due to the co-expression of CD90 and CD117. With novel digital image analysis technologies, a quantification of the CD117 and CD90 signals is available. Our experiments reveal an increase of CD117 and CD90 in patients with myocarditis.
Collapse
Affiliation(s)
- Marie Gerisch
- Applied Stem Cell Biology and Cell Technology, Biomedical and Biotechnological Center, University of Leipzig, Leipzig, Germany
| | - Jan Smettan
- Division of Cardiology and Angiology, Department of Internal Medicine, Neurology and Dermatology, University Hospital Leipzig, Leipzig, Germany
| | - Sabine Ebert
- Applied Stem Cell Biology and Cell Technology, Biomedical and Biotechnological Center, University of Leipzig, Leipzig, Germany
| | | | - Beate Brand-Saberi
- Department of Anatomy and Molecular Embryology, Institute of Anatomy, Faculty of Medicine, Ruhr-University Bochum, Bochum, Germany
| | - Nick Spindler
- Department of Orthopedics, Trauma and Plastic Surgery, University Hospital Leipzig, Leipzig, Germany
| | - Wolf C Mueller
- Department of Neuropathology, University Hospital Leipzig, Leipzig, Germany
| | - Shibashish Giri
- Applied Stem Cell Biology and Cell Technology, Biomedical and Biotechnological Center, University of Leipzig, Leipzig, Germany.,Department of Plastic and Hand Surgery, University Hospital Rechts der Isar, Munich Technical University, Munich, Germany
| | - Augustinus Bader
- Applied Stem Cell Biology and Cell Technology, Biomedical and Biotechnological Center, University of Leipzig, Leipzig, Germany
| |
Collapse
|
21
|
Cardiac Progenitor Cells and the Interplay with Their Microenvironment. Stem Cells Int 2017; 2017:7471582. [PMID: 29075298 PMCID: PMC5623801 DOI: 10.1155/2017/7471582] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 07/26/2017] [Indexed: 02/06/2023] Open
Abstract
The microenvironment plays a crucial role in the behavior of stem and progenitor cells. In the heart, cardiac progenitor cells (CPCs) reside in specific niches, characterized by key components that are altered in response to a myocardial infarction. To date, there is a lack of knowledge on these niches and on the CPC interplay with the niche components. Insight into these complex interactions and into the influence of microenvironmental factors on CPCs can be used to promote the regenerative potential of these cells. In this review, we discuss cardiac resident progenitor cells and their regenerative potential and provide an overview of the interactions of CPCs with the key elements of their niche. We focus on the interaction between CPCs and supporting cells, extracellular matrix, mechanical stimuli, and soluble factors. Finally, we describe novel approaches to modulate the CPC niche that can represent the next step in recreating an optimal CPC microenvironment and thereby improve their regeneration capacity.
Collapse
|
22
|
Hashimoto R, Joshi SR, Jiang H, Capdevila JH, McMurtry IF, Laniado Schwartzman M, Gupte SA. Cyp2c44 gene disruption is associated with increased hematopoietic stem cells: implication in chronic hypoxia-induced pulmonary hypertension. Am J Physiol Heart Circ Physiol 2017; 313:H293-H303. [PMID: 28550179 DOI: 10.1152/ajpheart.00785.2016] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 05/05/2017] [Accepted: 05/20/2017] [Indexed: 01/02/2023]
Abstract
We have recently demonstrated that disruption of the murine cytochrome P-450 2c44 gene (Cyp2c44) exacerbates chronic hypoxia-induced pulmonary artery remodeling and hypertension in mice. Subsequently, we serendipitously found that Cyp2c44 gene disruption also increases hematopoietic stem cell (HSC) numbers in bone marrow and blood. Therefore, the objective of the present study was to investigate whether CYP2C44-derived eicosanoids regulate HSC proliferation/cell growth and whether increased HSCs contribute to chronic hypoxia-induced remodeling of pulmonary arteries in Cyp2c44 knockout mice. Our findings demonstrated that lack of CYP2C44 epoxygenase, which catalyzed the oxidation of arachidonic acid to epoxyeicosatrienoic (EETs) and hydroxyeicosatetraenoic (HETE) acids, increases the numbers of 1) HSCs (CD34+, CD117+, and CD133+), 2) proangiogenic (CD34+CD133+ and CD34+CD117+CD133+) cells, and 3) immunogenic/inflammatory (CD34+CD11b+, CD133+CD11b+, F4/80+, CD11b+, and F4/80+CD11b+) macrophages in bone marrow and blood compared with wild-type mice. Among the various CYP2C44-derived arachidonic acids, only 15-HETE decreased CD117+ cell numbers when applied to bone marrow cell cultures. Interestingly, CD133+ and von Willebrand factor-positive cells, which are derived from proangiogenic stem cells, are increased in the bone marrow, blood, and lungs of mice exposed to chronic hypoxia and in remodeled and occluded pulmonary arteries of CYP2C44-deficient mice. In conclusion, our results demonstrate that CYP2C44-derived 15-HETE plays a critical role in downregulating HSC proliferation and growth, because disruption of the Cyp2c44 gene increased HSCs that potentially contribute to chronic hypoxia-induced pulmonary arterial remodeling and occlusion.NEW & NOTEWORTHY This study demonstrates that cytochrome P-450 2C44 plays a critical role in controlling the phenotype of hematopoietic stem cells and that when this enzyme is knocked out, stem cells are differentiated. These stem cells give rise to increased circulating monocytes and macrophages and contribute to the pathogenesis of chronic hypoxia-induced pulmonary artery remodeling and hypertension.
Collapse
Affiliation(s)
- Ryota Hashimoto
- Department of Pharmacology, and Translation Cardiovascular Institute, School of Medicine, New York Medical College, Valhalla, New York.,Department of Physiology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Sachindra Raj Joshi
- Department of Pharmacology, and Translation Cardiovascular Institute, School of Medicine, New York Medical College, Valhalla, New York
| | - Houli Jiang
- Department of Pharmacology, and Translation Cardiovascular Institute, School of Medicine, New York Medical College, Valhalla, New York
| | - Jorge H Capdevila
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee; and
| | - Ivan F McMurtry
- Department of Pharmacology, University of South Alabama, Mobile, Alabama
| | - Michal Laniado Schwartzman
- Department of Pharmacology, and Translation Cardiovascular Institute, School of Medicine, New York Medical College, Valhalla, New York
| | - Sachin A Gupte
- Department of Pharmacology, and Translation Cardiovascular Institute, School of Medicine, New York Medical College, Valhalla, New York;
| |
Collapse
|
23
|
Joshi SR, Dhagia V, Gairhe S, Edwards JG, McMurtry IF, Gupte SA. MicroRNA-140 is elevated and mitofusin-1 is downregulated in the right ventricle of the Sugen5416/hypoxia/normoxia model of pulmonary arterial hypertension. Am J Physiol Heart Circ Physiol 2016; 311:H689-98. [PMID: 27422986 DOI: 10.1152/ajpheart.00264.2016] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Accepted: 07/13/2016] [Indexed: 01/18/2023]
Abstract
Heart failure, a major cause of morbidity and mortality in patients with pulmonary arterial hypertension (PAH), is an outcome of complex biochemical processes. In this study, we determined changes in microRNAs (miRs) in the right and left ventricles of normal and PAH rats. Using an unbiased quantitative miR microarray analysis, we found 1) miR-21-5p, miR-31-5 and 3p, miR-140-5 and 3p, miR-208b-3p, miR-221-3p, miR-222-3p, miR-702-3p, and miR-1298 were upregulated (>2-fold; P < 0.05) in the right ventricle (RV) of PAH compared with normal rats; 2) miR-31-5 and 3p, and miR-208b-3p were upregulated (>2-fold; P < 0.05) in the left ventricle plus septum (LV+S) of PAH compared with normal rats; 3) miR-187-5p, miR-208a-3p, and miR-877 were downregulated (>2-fold; P < 0.05) in the RV of PAH compared with normal rats; and 4) no miRs were up- or downregulated with >2-fold in LV+S compared with RV of PAH and normal. Upregulation of miR-140 and miR-31 in the hypertrophic RV was further confirmed by quantitative PCR. Interestingly, compared with control rats, expression of mitofusin-1 (MFN1), a mitochondrial fusion protein that regulates apoptosis, and which is a direct target of miR-140, was reduced in the RV relative to LV+S of PAH rats. We found a correlation between increased miR-140 and decreased MFN1 expression in the hypertrophic RV. Our results also demonstrated that upregulation of miR-140 and downregulation of MFN1 correlated with increased RV systolic pressure and hypertrophy. These results suggest that miR-140 and MFN1 play a role in the pathogenesis of PAH-associated RV dysfunction.
Collapse
Affiliation(s)
| | - Vidhi Dhagia
- Department of Pharmacology, New York Medical College, Valhalla, New York
| | - Salina Gairhe
- Department of Pharmacology and Center for Lung Biology, University of South Alabama, College of Medicine, Mobile, Alabama; and
| | - John G Edwards
- Department of Physiology, New York Medical College, Valhalla, New York
| | - Ivan F McMurtry
- Department of Pharmacology and Center for Lung Biology, University of South Alabama, College of Medicine, Mobile, Alabama; and
| | - Sachin A Gupte
- Department of Pharmacology, New York Medical College, Valhalla, New York;
| |
Collapse
|
24
|
Cui J, Zhang F, Wang Y, Liu J, Ming X, Hou J, Lv B, Fang S, Yu B. Macrophage migration inhibitory factor promotes cardiac stem cell proliferation and endothelial differentiation through the activation of the PI3K/Akt/mTOR and AMPK pathways. Int J Mol Med 2016; 37:1299-309. [PMID: 27035848 PMCID: PMC4829139 DOI: 10.3892/ijmm.2016.2542] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 03/16/2016] [Indexed: 01/05/2023] Open
Abstract
Macrophage migration inhibitory factor (MIF) has pleiotropic immune functions in a number of inflammatory diseases. Recent evidence from expression and functional studies has indicated that MIF is involved in various aspects of cardiovascular disease. In this study, we aimed to determine whether MIF supports in vitro c-kit+CD45− cardiac stem cell (CSC) survival, proliferation and differentiation into endothelial cells, as well as the possible mechanisms involved. We observed MIF receptor (CD74) expression in mouse CSCs (mCSCs) using PCR and immunofluorescence staining, and MIF secretion by mCSCs using PCR and ELISA in vitro. Increasing amounts of exogenous MIF did not affect CD74 expression, but promoted mCSC survival, proliferation and endothelial differentiation. By contrast, treatment with an MIF inhibitor (ISO-1) or siRNA targeting CD74 (CD74-siRNA) suppressed the biological changes induced by MIF in the mCSCs. Increasing amounts of MIF increased the phosphorylation of Akt and mammalian target of rapamycin (mTOR), which are known to support cell survival, proliferation and differentiation. These effects of MIF on the mCSCs were abolished by LY294002 [a phosphoinositide 3-kinase (PI3K) inhibitor] and MK-2206 (an Akt inhibitor). Moreover, adenosine monophosphate-activated protein kinase (AMPK) phosphorylation increased following treatment with MIF. The AMPK inhibitor, compound C, partly blocked the pro-proliferative effects of MIF on the mCSCs. In conclusion, our results suggest that MIF promotes mCSC survival, proliferation and endothelial differentiation through the activation of the PI3K/Akt/mTOR and AMPK signaling pathways. Thus, MIF may prove to be a potential therapeutic factor in the treatment of heart failure and myocardial infarction by activating CSCs.
Collapse
Affiliation(s)
- Jinjin Cui
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Fengyun Zhang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Yongshun Wang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Jingjin Liu
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Xing Ming
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Jingbo Hou
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Bo Lv
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Shaohong Fang
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, Heilongjiang 150081, P.R. China
| | - Bo Yu
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| |
Collapse
|
25
|
Ghazizadeh Z, Vahdat S, Fattahi F, Fonoudi H, Omrani G, Gholampour M, Aghdami N. Isolation and characterization of cardiogenic, stem-like cardiac precursors from heart samples of patients with congenital heart disease. Life Sci 2015; 137:105-15. [PMID: 26165749 DOI: 10.1016/j.lfs.2015.07.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Revised: 06/04/2015] [Accepted: 07/02/2015] [Indexed: 01/14/2023]
Abstract
AIMS Regenerative therapies based on resident human cardiac progenitor cells (hCPCs) are a promising alternative to medical treatments for patients with myocardial infarction. However, hCPCs are rare in human heart and finding efficient source and proper surface marker for isolation of these cells would make them a good candidate for therapy. MAIN METHODS We have isolated 5.34∗10(6)±2.04∗10(5)/g viable cells from 35 heart tissue samples of 23 patients with congenital heart disease obtained during their heart surgery along with 6 samples from 3 normal subjects during cardiac biopsy. KEY FINDINGS According to FACS analysis, younger ages, atrial specimen and disease with increased pulmonary vascular resistance were associated with higher percentage of c-kit(+) (CD117) hCPCs. Analysis for other stemness markers revealed increased CD133(+) cells in the hearts of patients with congenital heart disease. By using both immune-labeling and PCR, we demonstrated that these cells express key cardiac lineage and endothelial transcription factors and structural proteins during in vitro differentiation and do express stemness transcription factors in undifferentiated state. Another novel datum of potentially relevant interest is their ability in promoting greater myocardial regeneration and better survival in rat model of myocardial infarction following transplantation. SIGNIFICANCE Our results could provide evidence for conditions associated with enriched hCPCs in patients with congenital heart disease. Moreover, we showed presence of a significant number of CD133 expressing cardiogenic stem-like cardiac precursors in the heart of patients with congenital heart disease, which could be isolated and stored for future regenerative therapies in these patients.
Collapse
Affiliation(s)
- Zaniar Ghazizadeh
- Department of Stem Cells and Developmental Biology at the Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Sadaf Vahdat
- Department of Stem Cells and Developmental Biology at the Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Faranak Fattahi
- Department of Molecular Systems Biology at the Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Hananeh Fonoudi
- Department of Stem Cells and Developmental Biology at the Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Gholamreza Omrani
- Department of Cardiac Surgery, Rajaei Cardiovascular Medical Research Center, Tehran, Iran
| | - Maziar Gholampour
- Department of Cardiac Surgery, Rajaei Cardiovascular Medical Research Center, Tehran, Iran
| | - Nasser Aghdami
- Department of Stem Cells and Developmental Biology at the Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Department of Regenerative Biomedicine at the Cell Science Research, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| |
Collapse
|
26
|
Bei Y, Wang F, Yang C, Xiao J. Telocytes in regenerative medicine. J Cell Mol Med 2015; 19:1441-54. [PMID: 26059693 PMCID: PMC4511344 DOI: 10.1111/jcmm.12594] [Citation(s) in RCA: 110] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 03/15/2015] [Indexed: 12/13/2022] Open
Abstract
Telocytes (TCs) are a distinct type of interstitial cells characterized by a small cell body and extremely long and thin telopodes (Tps). The presence of TCs has been documented in many tissues and organs (go to http://www.telocytes.com). Functionally, TCs form a three-dimensional (3D) interstitial network by homocellular and heterocellular communication and are involved in the maintenance of tissue homeostasis. As important interstitial cells to guide or nurse putative stem and progenitor cells in stem cell niches in a spectrum of tissues and organs, TCs contribute to tissue repair and regeneration. This review focuses on the latest progresses regarding TCs in the repair and regeneration of different tissues and organs, including heart, lung, skeletal muscle, skin, meninges and choroid plexus, eye, liver, uterus and urinary system. By targeting TCs alone or in tandem with stem cells, we might promote regeneration and prevent the evolution to irreversible tissue damage. Exploring pharmacological or non-pharmacological methods to enhance the growth of TCs would be a novel therapeutic strategy besides exogenous transplantation for many diseased disorders.
Collapse
Affiliation(s)
- Yihua Bei
- Regeneration and Ageing Lab, Experimental Center of Life Sciences, School of Life Science, Shanghai UniversityShanghai, China
| | - Fei Wang
- Division of Gastroenterology and Hepatology, Digestive Disease Institute, Shanghai Tongji Hospital, Tongji University School of MedicineShanghai, China
| | - Changqing Yang
- Division of Gastroenterology and Hepatology, Digestive Disease Institute, Shanghai Tongji Hospital, Tongji University School of MedicineShanghai, China
| | - Junjie Xiao
- Regeneration and Ageing Lab, Experimental Center of Life Sciences, School of Life Science, Shanghai UniversityShanghai, China
| |
Collapse
|
27
|
Boucek RJ, Steele J, Jacobs JP, Steele P, Asante-Korang A, Quintessenza J, Steele A. Ex vivo paracrine properties of cardiac tissue: Effects of chronic heart failure. J Heart Lung Transplant 2015; 34:839-48. [DOI: 10.1016/j.healun.2014.07.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Revised: 06/11/2014] [Accepted: 07/10/2014] [Indexed: 12/15/2022] Open
|
28
|
Rusu MC, Vrapciu AD, Hostiuc S, Hariga CS. Brown adipocytes, cardiac protection and a common adipo- and myogenic stem precursor in aged human hearts. Med Hypotheses 2015; 85:212-4. [PMID: 25956736 DOI: 10.1016/j.mehy.2015.04.027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Revised: 04/18/2015] [Accepted: 04/25/2015] [Indexed: 12/25/2022]
Abstract
New data on adult stem cells (ASCs) are continuously added by research for use in regenerative medicine. However organ-specific ASC markers are incompletely explored. It was demonstrated that in non-cardiac brown adipose tissue (BAT) CD133+ cells differentiate in cardiomyocytes, and such BAT-derived cells induce bone marrow-derived cells into cardiomyocytes, thus being a promising source for cardiac stem cell therapy. During embryogenesis the subepicardial fat derives from BAT. Although it was not specifically investigated in human adult or aged hearts, it is actually known that metabolically active BAT can be found in many adult humans, is related to antiobesity effects, and it may derive from stem/progenitor cells. Stro-1 can safely identify in situ cardiac stem cells (CSCs) with myogenic and adipogenic potential. It was therefore raised the hypothesis of subepicardial differentiation of CSCs in BAT in adult/aged hearts, which could be viewed, such as in infants, as a mechanism of protection. This could be determined by the reactivation of an embryologic differentiation pattern in which brown adipocytes and muscle cells derive from a common stem ancestor. Such quiescent common stem ancestors could be suggested in adult, or aged, human hearts, when subepicardial BAT is found, and if a Stro-1+/CD133+/Isl-1+ phenotype of CSCs is determined.
Collapse
Affiliation(s)
- M C Rusu
- Division of Anatomy, Faculty of Dental Medicine, "Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania; MEDCENTER, Center of Excellence in Laboratory Medicine and Pathology, Bucharest, Romania; International Society of Regenerative Medicine and Surgery (ISRMS), Romania.
| | - A D Vrapciu
- Division of Anatomy, Faculty of Dental Medicine, "Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania
| | - S Hostiuc
- Division of Legal Medicine and Bioethics, Department 2 Morphological Sciences, Faculty of Medicine, "Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania; National Institute of Legal Medicine, Bucharest, Romania
| | - C S Hariga
- Department 11 Surgery, Faculty of Medicine, "Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania
| |
Collapse
|
29
|
Bei Y, Zhou Q, Fu S, Lv D, Chen P, Chen Y, Wang F, Xiao J. Cardiac telocytes and fibroblasts in primary culture: different morphologies and immunophenotypes. PLoS One 2015; 10:e0115991. [PMID: 25693182 PMCID: PMC4333820 DOI: 10.1371/journal.pone.0115991] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Accepted: 12/03/2014] [Indexed: 01/30/2023] Open
Abstract
Telocytes (TCs) are a peculiar type of interstitial cells with very long prolongations termed telopodes. TCs have previously been identified in different anatomic structures of the heart, and have also been isolated and cultured from heart tissues in vitro. TCs and fibroblasts, both located in the interstitial spaces of the heart, have different morphologies and functionality. However, other than microscopic observation, a reliable means to make differential diagnosis of cardiac TCs from fibroblasts remains unclear. In the present study, we isolated and cultured cardiac TCs and fibroblasts from heart tissues, and observed their different morphological features and immunophenotypes in primary culture. Morphologically, TCs had extremely long and thin telopodes with moniliform aspect, stretched away from cell bodies, while cell processes of fibroblasts were short, thick and cone shaped. Furthermore, cardiac TCs were positive for CD34/c-kit, CD34/vimentin, and CD34/PDGFR-β, while fibroblasts were only vimentin and PDGFR-β positive. In addition, TCs were also different from pericytes as TCs were CD34 positive and α-SMA weak positive while pericytes were CD34 negative but α-SMA positive. Besides that, we also showed cardiac TCs were homogenously positive for mesenchymal marker CD29 but negative for hematopoietic marker CD45, indicating that TCs could be a source of cardiac mesenchymal cells. The differences in morphological features and immunophenotypes between TCs and fibroblasts will provide more compelling evidence to differentiate cardiac TCs from fibroblasts.
Collapse
Affiliation(s)
- Yihua Bei
- Regeneration and Ageing Lab, Experimental Center of Life Sciences and Innovative Drug Research Center, School of Life Science, Shanghai University, Shanghai 200444, China
- Shanghai Key Laboratory of Bio-Energy Crops, School of Life Science, Shanghai University, Shanghai 200444, China
| | - Qiulian Zhou
- Regeneration and Ageing Lab, Experimental Center of Life Sciences and Innovative Drug Research Center, School of Life Science, Shanghai University, Shanghai 200444, China
| | - Siyi Fu
- Regeneration and Ageing Lab, Experimental Center of Life Sciences and Innovative Drug Research Center, School of Life Science, Shanghai University, Shanghai 200444, China
| | - Dongchao Lv
- Regeneration and Ageing Lab, Experimental Center of Life Sciences and Innovative Drug Research Center, School of Life Science, Shanghai University, Shanghai 200444, China
| | - Ping Chen
- Regeneration and Ageing Lab, Experimental Center of Life Sciences and Innovative Drug Research Center, School of Life Science, Shanghai University, Shanghai 200444, China
| | - Yuanyuan Chen
- Regeneration and Ageing Lab, Experimental Center of Life Sciences and Innovative Drug Research Center, School of Life Science, Shanghai University, Shanghai 200444, China
| | - Fei Wang
- Division of Gastroenterology and Hepatology, Digestive Disease Institute, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China
| | - Junjie Xiao
- Regeneration and Ageing Lab, Experimental Center of Life Sciences and Innovative Drug Research Center, School of Life Science, Shanghai University, Shanghai 200444, China
- Shanghai Key Laboratory of Bio-Energy Crops, School of Life Science, Shanghai University, Shanghai 200444, China
- Division of Gastroenterology and Hepatology, Digestive Disease Institute, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China
| |
Collapse
|
30
|
Abstract
Although the adult mammalian heart was once believed to be a post-mitotic organ without any capacity for regeneration, recent findings have challenged this dogma. A modified view assigns to the mammalian heart a measurable capacity for regeneration throughout life. The ultimate goals of the cardiac regeneration field have been pursued by multiple strategies, including understanding the developmental biology of cardiomyocytes and cardiac stem and progenitor cells, applying chemical genetics, and engineering biomaterials and delivery methods that facilitate cell transplantation. Successful stimulation of endogenous regenerative capacity in injured adult mammalian hearts can benefit from studies of natural cardiac regeneration.
Collapse
Affiliation(s)
- Aurora Bernal
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), C/Melchor Fernández Almagro, 3, 28029 Madrid, Spain
| | - Beatriz G. Gálvez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), C/Melchor Fernández Almagro, 3, 28029 Madrid, Spain
| |
Collapse
|
31
|
DONATO GIUSEPPE, CONFORTI FRANCESCO, CAMASTRA CATERINA, AMMENDOLA MICHELE, DONATO ANNALIDIA, RENZULLI ATTILIO. The role of mast cell tryptases in cardiac myxoma: Histogenesis and development of a challenging tumor. Oncol Lett 2014; 8:379-383. [PMID: 24959280 PMCID: PMC4063662 DOI: 10.3892/ol.2014.2104] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Accepted: 02/26/2014] [Indexed: 12/17/2022] Open
Abstract
A number of available studies have focused on the role of mastocytes and their angiogenic factors, such as tryptase expression, in cancer growth as a major research objective. Cardiac myxoma is a rare neoplasia and is the most common primary tumor of the heart. The cellular elements of cardiac myxoma have an endothelial phenotype; however, its histogenesis remains unclear. Currently, no available studies have correlated the pathological characteristics of cardiac myxomas, such as cell differentiation and vascularization, with the angiogenic factors of mast cells. The aim of the present study was to investigate the role of mast cell tryptases on the development of cardiac myxomas and examine the histogenesis of tumoral cells. A series of 10 cardiac myxomas were examined by immunohistochemical analysis for the presence of tryptase-positive mast cells. Statistical analysis of our data demonstrated that angiogenesis and the development of pseudovascular structures were correlated with the number of tryptase-positive mast cells. Therefore, we hypothesize that cardiac myxoma cells are endothelial precursors which are able to generate mature vascular structures. Further morphological and immunophenotypic analyses of tumoral cells may corroborate such a hypothesis.
Collapse
Affiliation(s)
- GIUSEPPE DONATO
- Department of Pathology, School of Medicine, University Magna Graecia, Catanzaro I-88100, Italy
| | - FRANCESCO CONFORTI
- Department of Pathology, School of Medicine, University Magna Graecia, Catanzaro I-88100, Italy
| | - CATERINA CAMASTRA
- Department of Pathology, School of Medicine, University Magna Graecia, Catanzaro I-88100, Italy
| | - MICHELE AMMENDOLA
- Department of Pharmacology, School of Medicine, University Magna Graecia, Catanzaro I-88100, Italy
| | - ANNALIDIA DONATO
- Department of Pathology, School of Medicine, University Magna Graecia, Catanzaro I-88100, Italy
| | - ATTILIO RENZULLI
- Department of Cardiac Surgery, School of Medicine, University Magna Graecia, Catanzaro I-88100, Italy
| |
Collapse
|
32
|
Matuszczak S, Czapla J, Jarosz-Biej M, Wiśniewska E, Cichoń T, Smolarczyk R, Kobusińska M, Gajda K, Wilczek P, Sliwka J, Zembala M, Zembala M, Szala S. Characteristic of c-Kit+ progenitor cells in explanted human hearts. Clin Res Cardiol 2014; 103:711-8. [PMID: 24722830 PMCID: PMC4129222 DOI: 10.1007/s00392-014-0705-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2013] [Accepted: 03/20/2014] [Indexed: 11/29/2022]
Abstract
According to literature data, self-renewing, multipotent, and clonogenic cardiac c-Kit+ progenitor cells occur within human myocardium. The aim of this study was to isolate and characterize c-Kit+ progenitor cells from explanted human hearts. Experimental material was obtained from 19 adult and 7 pediatric patients. Successful isolation and culture was achieved for 95 samples (84.1 %) derived from five different regions of the heart: right and left ventricles, atrium, intraventricular septum, and apex. The average percentage of c-Kit+ cells, as assessed by FACS, ranged between 0.7 and 0.9 %. In contrast to published data we do not observed statistically significant differences in the number of c-Kit+ cells between disease-specific groups, parts of the heart or sexes. Nevertheless, c-Kit+ cells were present in significant numbers (11–24 %) in samples derived from three explanted pediatric hearts. c-Kit+ cells were also positive for CD105 and a majority of them was positive for CD31 and CD34 (83.7 ± 8.6 and 75.7 ± 11.4 %, respectively). Immunohistochemical analysis of the heart tissue revealed that most cells possessing the c-Kit antigen were also positive for tryptase, a specific mast cell marker. However, flow cytometry analysis has shown cultured c-Kit+ cells to be negative for hematopoietic marker CD45 and mast cell marker CD33. Isolated c-Kit+ cells display mesenchymal stem cell features and are thought to differentiate into endothelial cells.
Collapse
Affiliation(s)
- Sybilla Matuszczak
- Center for Translational Research and Molecular Biology of Cancer, Maria Skłodowska-Curie Memorial Cancer Center and Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej Street 15, 44-101, Gliwice, Poland
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Stem cell marker-positive stellate cells and mast cells are reduced in benign-appearing bladder tissue in patients with urothelial carcinoma. Virchows Arch 2014; 464:473-88. [PMID: 24570393 DOI: 10.1007/s00428-014-1561-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Revised: 02/07/2014] [Accepted: 02/13/2014] [Indexed: 01/16/2023]
Abstract
Survival after invasive bladder cancer has improved less than that of other common non-skin cancers. In many types of malignancy, treatment failure has been attributed to therapy-resistant stem-like cancer cells. Our aim was therefore to determine identities of stem cell marker-positive cells in bladder cancer tissue and to investigate possible associations between these cells and different forms of bladder neoplasia. We investigated tissue from 52 patients with bladder neoplasia and 18 patients with benign bladder conditions, from a cohort that had been previously described with regard to diagnosis and outcome. The samples were analysed immunohistologically for the stem cell markers aldehyde dehydrogenase 1 A1 (ALDH1) and CD44, and markers of cell differentiation. The majority of stem cell marker-positive cells were located in connective tissue, and a smaller fraction in epithelial tissue. Stem cell marker-positive cells exhibiting possible stem cell characteristics included cells in deeper locations of benign and malignant epithelium, and sub-endothelial cells in patients with or without neoplasia. Stem cell marker-positive cells with non-stem cell character included stellate cells, mast cells, endothelial cells, foamy histiocytes, and neurons. Significantly, ALDH1+ stellate cells and ALDH1+ mast cells were reduced in number in stroma of benign-appearing mucosa of bladder cancer patients. The stem cell markers ALDH1 and CD44 label several types of differentiated cells in bladder tissue. ALDH1+ stellate cells and mast cells appear to be reduced in stroma of normal-appearing mucosa of bladder cancer patients, and may be part of a "field effect" in cancer-near areas.
Collapse
|
34
|
Tang YL, Wang YJ, Chen LJ, Pan YH, Zhang L, Weintraub NL. Cardiac-derived stem cell-based therapy for heart failure: progress and clinical applications. Exp Biol Med (Maywood) 2013; 238:294-300. [PMID: 23598975 DOI: 10.1177/1535370213477982] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Stem cell-based therapy is emerging as a promising strategy to treat end-stage heart failure, a leading cause of morbidity and mortality. Stem cells can be isolated from a variety of sources and exhibit unique characteristics that impact their potential therapeutic utility. The adult heart contains small populations of committed, multipotent cardiac stem cells (CSC), which are adapted to the cardiac microenvironment and participate in postnatal physiological and pathological cardiac renewal or repair. These cells can be isolated, expanded in culture, and administered therapeutically to improve cardiac function in the setting of heart failure. CSC can be differentiated into three distinct cardiovascular lineages and exhibit enhanced paracrine factor production and engraftment as compared with other types of mesenchymal stem cells, which in turn may translate into improved therapeutic efficacy. The cell surface marker expression and phenotype of these CSC, however, depends on the method of isolation, selection and propagation, which likely explains the variable experimental results obtained to date. Moreover, invasive procedures are required to obtain CSC from humans. Early trials using autologous CSC in patients with ischemic cardiomyopathy have demonstrated feasibility and safety, along with variable degrees of therapeutic efficacy in terms of enhancing myocardial viability and cardiac function. Further studies are needed to optimize methods of CSC isolation, manipulation and delivery. If fully realized, the potential of CSC therapy could fundamentally change the approach to the treatment of end-stage heart failure.
Collapse
Affiliation(s)
- Yaoliang L Tang
- Division of Cardiovascular Disease, Department of Internal Medicine, College of Medicine, University of Cincinnati, Ohio 45267, USA.
| | | | | | | | | | | |
Collapse
|
35
|
Itinteang T, Tan ST, Jia J, Steel R, Laing EL, Brasch HD, Day DJ. Mast cells in infantile haemangioma possess a primitive myeloid phenotype. J Clin Pathol 2013; 66:597-600. [PMID: 23559352 DOI: 10.1136/jclinpath-2012-201096] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
AIMS Recent reports on infantile haemangioma (IH) have demonstrated a primitive population of interstitial cells expressing the embryonic transcription factor, Nanog, with decreasing abundance during involution. In this report we investigated the expression of Nanog on mast cells in all three phases of IH progression. METHODS Paraffin-embedded sections of six proliferating, six involuting and six involuted IH lesions were used to investigate the expression of tryptase, Nanog, CD45, CD34 and GLUT-1 by immunostaining. RESULTS Mast cells, identified by their expression of tryptase, were located in the interstitium of IH lesions. 93%, 42% and 0% of these tryptase(+) cells also expressed Nanog, in proliferating, involuting and involuted IH, respectively. CONCLUSIONS The identification of an abundant population of tryptase(+)/Nanog(+) cells in IH is novel. The relative loss of Nanog expression as IH involutes may be a result of maturation and/or proliferation of these cells. This report supports the primitive nature of IH.
Collapse
|
36
|
Heart cells with regenerative potential from pediatric patients with end stage heart failure: a translatable method to enrich and propagate. Stem Cells Int 2012; 2012:452102. [PMID: 22936950 PMCID: PMC3425869 DOI: 10.1155/2012/452102] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2012] [Revised: 06/22/2012] [Accepted: 06/29/2012] [Indexed: 11/17/2022] Open
Abstract
Background. Human cardiac-derived progenitor cells (hCPCs) have shown promise in treating heart failure (HF) in adults. The purpose of this study was to describe derivation of hCPCs from pediatric patients with end-stage HF. Methods. At surgery, discarded right atrial tissues (hAA) were obtained from HF patients (n = 25; hAA-CHF). Minced tissues were suspended in complete (serum-containing) DMEM. Cells were selected for their tissue migration and expression of stem cell factor receptor (hc-kit). Characterization of hc-kit(positive) cells included immunohistochemical screening with a panel of monoclonal antibodies. Results. Cells, including phase-bright cells identified as hc-kit(positive), spontaneously emigrated from hAA-CHF in suspended explant cultures (SEC) after Day 7. When cocultured with tissue, emigrated hc-kit(positive) cells proliferated, first as loosely attached clones and later as multicellular clusters. At Day 21~5% of cells were hc-kit(positive). Between Days 14 and 28 hc-kit(positive) cells exhibited mesodermal commitment (GATA-4(positive) and NKX2.5(positive)); then after Day 28 cardiac lineages (flk-1(positive), smooth muscle actin(positive), troponin-I(positive), and myosin light chain(positive)). Conclusions. C-kit(positive) hCPCs can be derived from atrial tissue of pediatric patients with end-stage HF. SEC is a novel culture method for derivation of migratory hc-kit(positive) cells that favors clinical translation by reducing the need for exogenously added factors to expand hCPCs in vitro.
Collapse
|
37
|
Mastri M, Shah Z, McLaughlin T, Greene CJ, Baum L, Suzuki G, Lee T. Activation of Toll-like receptor 3 amplifies mesenchymal stem cell trophic factors and enhances therapeutic potency. Am J Physiol Cell Physiol 2012; 303:C1021-33. [PMID: 22843797 DOI: 10.1152/ajpcell.00191.2012] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Clinical trials of bone marrow mesenchymal stem cell (MSC) therapy have thus far demonstrated moderate and inconsistent benefits, indicating an urgent need to improve therapeutic efficacy. Although administration of sufficient cells is necessary to achieve maximal therapeutic benefits, documented MSC clinical trials have largely relied on injections of ∼1 × 10(6) cells/kg, which appears too low to elicit a robust therapeutic response according to published preclinical studies. However, repeated cell passaging necessary for large-scale expansion of MSC causes cellular senescence and reduces stem cell potency. Using the RNA mimetic polyinosinic-polycytidylic acid [poly(I:C)] to engage MSC Toll-like receptor 3 (TLR3), we found that poly(I:C), signaling through multiple mitogen-activated protein kinase pathways, induced therapeutically relevant trophic factors such as interleukin-6-type cytokines, stromal-derived factor 1, hepatocyte growth factor, and vascular endothelial growth factor while slightly inhibiting the proliferation and migration potentials of MSC. At the suboptimal injection dose of 1 × 10(6) cells/kg, poly(I:C)-treated MSC, but not untreated MSC, effectively stimulated regeneration of the failing hamster heart 1 mo after cell administration. The regenerating heart exhibited increased CD34(+)/Ki67(+) and CD34(+)/GATA4(+) progenitor cells in the presence of decreased inflammatory cells and cytokines. Cardiac functional improvement was associated with a ∼50% reduction in fibrosis, a ∼40% reduction in apoptosis, and a ∼55% increase in angiogenesis, culminating in prominent cardiomyogenesis evidenced by abundant distribution of small myocytes and a ∼90% increase in wall thickening. These functional, histological, and molecular characterizations thus establish the utility of TLR3 engagement for enabling the low-dose MSC therapy that may be translated to more efficacious clinical applications.
Collapse
Affiliation(s)
- Michalis Mastri
- Department of Biochemistry, Center for Research in Cardiovascular Medicine, University at Buffalo, 3435 Main St., Buffalo, NY 14214, USA
| | | | | | | | | | | | | |
Collapse
|
38
|
Abstract
A heart attack kills off many cells in the heart. Parts of the heart become thin and fail to contract properly following the replacement of lost cells by scar tissue. However, the notion that the same adult cardiomyocytes beat throughout the lifespan of the organ and organism, without the need for a minimum turnover, gives way to a fascinating investigations. Since the late 1800s, scientists and cardiologists wanted to demonstrate that the cardiomyocytes cannot be generated after the perinatal period in human beings. This curiosity has been passed down in subsequent years and has motivated more and more accurate studies in an attempt to exclude the presence of renewed cardiomyocytes in the tissue bordering the ischaemic area, and then to confirm the dogma of the heart as terminally differentiated organ. Conversely, peri-lesional mitosis of cardiomyocytes were discovered initially by light microscopy and subsequently confirmed by more sophisticated technologies. Controversial evidence of mechanisms underlying myocardial regeneration has shown that adult cardiomyocytes are renewed through a slow turnover, even in the absence of damage. This turnover is ensured by the activation of rare clusters of progenitor cells interspersed among the cardiac cells functionally mature. Cardiac progenitor cells continuously interact with each other, with the cells circulating in the vessels of the coronary microcirculation and myocardial cells in auto-/paracrine manner. Much remains to be understood; however, the limited functional recovery in human beings after myocardial injury clearly demonstrates weak regenerative potential of cardiomyocytes and encourages the development of new approaches to stimulate this process.
Collapse
Affiliation(s)
- Lucio Barile
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
| | | |
Collapse
|
39
|
Genead R, Fischer H, Hussain A, Jaksch M, Andersson AB, Ljung K, Bulatovic I, Franco-Cereceda A, Elsheikh E, Corbascio M, Smith CIE, Sylvén C, Grinnemo KH. Ischemia-reperfusion injury and pregnancy initiate time-dependent and robust signs of up-regulation of cardiac progenitor cells. PLoS One 2012; 7:e36804. [PMID: 22590612 PMCID: PMC3348899 DOI: 10.1371/journal.pone.0036804] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2011] [Accepted: 04/07/2012] [Indexed: 01/10/2023] Open
Abstract
To explore how cardiac regeneration and cell turnover adapts to disease, different forms of stress were studied for their effects on the cardiac progenitor cell markers c-Kit and Isl1, the early cardiomyocyte marker Nkx2.5, and mast cells. Adult female rats were examined during pregnancy, after myocardial infarction and ischemia-reperfusion injury with/out insulin like growth factor-1(IGF-1) and hepatocyte growth factor (HGF). Different cardiac sub-domains were analyzed at one and two weeks post-intervention, both at the mRNA and protein levels. While pregnancy and myocardial infarction up-regulated Nkx2.5 and c-Kit (adjusted for mast cell activation), ischemia-reperfusion injury induced the strongest up-regulation which occurred globally throughout the entire heart and not just around the site of injury. This response seems to be partly mediated by increased endogenous production of IGF-1 and HGF. Contrary to c-Kit, Isl1 was not up-regulated by pregnancy or myocardial infarction while ischemia-reperfusion injury induced not a global but a focal up-regulation in the outflow tract and also in the peri-ischemic region, correlating with the up-regulation of endogenous IGF-1. The addition of IGF-1 and HGF did boost the endogenous expression of IGF and HGF correlating to focal up-regulation of Isl1. c-Kit expression was not further influenced by the exogenous growth factors. This indicates that there is a spatial mismatch between on one hand c-Kit and Nkx2.5 expression and on the other hand Isl1 expression. In conclusion, ischemia-reperfusion injury was the strongest stimulus with both global and focal cardiomyocyte progenitor cell marker up-regulations, correlating to the endogenous up-regulation of the growth factors IGF-1 and HGF. Also pregnancy induced a general up-regulation of c-Kit and early Nkx2.5+ cardiomyocytes throughout the heart. Utilization of these pathways could provide new strategies for the treatment of cardiac disease.
Collapse
Affiliation(s)
- Rami Genead
- Division of Cardiology, Department of Medicine, Karolinska Institutet, Stockholm, Sweden.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Pressure overload leads to an increase of cardiac resident stem cells. Basic Res Cardiol 2012; 107:252. [PMID: 22361741 DOI: 10.1007/s00395-012-0252-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2011] [Revised: 12/29/2011] [Accepted: 02/06/2012] [Indexed: 01/13/2023]
Abstract
Recent studies suggest that the mammalian heart possesses some capacity for cardiac regeneration. This regenerative capacity is primarily documented postnatally and after myocardial infarction or pressure overload. Although the cell type that mediates endogenous regeneration is unclear, cardiac stem cells might be considered as potential candidates. To determine the number of c-kit + cardiac resident cells under conditions of pressure overload, we evaluated specimens derived from n = 8 patients with pressure overloaded single right ventricles in comparison to n = 4 explanted hearts from patients with dilated cardiomyopathy and n = 14 biopsies from children after heart transplantation. The age of the patients ranged from 16 days to 19 years. For quantification of cardiac stem cells, c-kit+/mast cell tryptase-/CD45- cells were counted and expressed as percent of the total nuclei. In specimens from patients with dilated cardiomyopathy, 0.13 ± 0.09% c-kit +/mast cell tryptase-/CD45- cells were detected. However, in specimens from patients with pressure overloaded single right ventricles, the numbers of c-kit+/mast cell tryptase-/CD45- cells were significantly higher (0.41 ±0.24%, p < 0.05). Under conditions of pressure overload, the right ventricle shows an approximately three-fold increase in c-kit+/mast cell tryptase-/CD45- cardiac resident cells. Despite the fact that this increased number of c-kit+ cells is not sufficient to prevent the failing heart from congestive heart failure, understanding the mechanism that leads to an increase of presumably cardiac resident stem cells under conditions of pressure overload might help to develop new strategies to enhance endogenous repair.
Collapse
|
41
|
Ressel L, Ricci E, Bonassi P, Parolini O. Simultaneous histochemical and immunohistochemical staining as a simple tool to identify mast cells within CD117-positive cell populations. Histopathology 2012; 60:655-7. [PMID: 22260311 DOI: 10.1111/j.1365-2559.2011.03957.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
42
|
He JQ, Vu DM, Hunt G, Chugh A, Bhatnagar A, Bolli R. Human cardiac stem cells isolated from atrial appendages stably express c-kit. PLoS One 2011; 6:e27719. [PMID: 22140461 PMCID: PMC3225366 DOI: 10.1371/journal.pone.0027719] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2011] [Accepted: 10/23/2011] [Indexed: 12/15/2022] Open
Abstract
The in vivo studies of myocardial infarct using c-kit+/Lin− cardiac stem cells (CSCs) are still in the early stage with margin or no beneficial effects for cardiac function. One of the potential reasons may be related to the absence of fully understanding the properties of these cells both in vitro and in vivo. In the present study, we aimed to systematically examine how CSCs adapted to in vitro cell processes and whether there is any cell contamination after long-term culture. Human CSCs were enzymatically isolated from the atrial appendages of patients. The fixed tissue sections, freshly isolated or cultured CSCs were then used for identification of c-kit+/Lin− cells, detection of cell contamination, or differentiation of cardiac lineages. By specific antibody staining, we demonstrated that tissue sections from atrial appendages contained less than 0.036% c-kit+/Lin− cells. For the first time, we noted that without magnetic activated cell sorting (MACS), the percentages of c-kit+/Lin− cells gradually increased up to ∼40% during continuously culture between passage 2 to 8, but could not exceed >80% unless c-kit MACS was carried out. The resulting c-kit+/Lin− cells were negative for CD34, CD45, CD133, and Lin markers, but positive for KDR and CD31 in few patients after c-kit MACS. Lin depletion seemed unnecessary for enrichment of c-kit+/Lin− cell population. Following induced differentiation, c-kit+/Lin− CSCs demonstrated strong differentiation towards cardiomyocytes but less towards smooth and endothelial cells. We concluded that by using an enzymatic dissociation method, a large number, or higher percentage, of relative pure human CSCs with stable expression of c-kit+ could be obtained from atrial appendage specimens within ∼4 weeks following c-kit MACS without Lin depletion. This simple but cost-effective approach can be used to obtain enough numbers of stably-expressed c-kit+/Lin− cells for clinical trials in repairing myocardial infarction.
Collapse
Affiliation(s)
- Jia-Qiang He
- Institute of Molecular Cardiology, University of Louisville, Louisville, Kentucky, United States of America
| | - Duc Minh Vu
- Institute of Molecular Cardiology, University of Louisville, Louisville, Kentucky, United States of America
| | - Greg Hunt
- Institute of Molecular Cardiology, University of Louisville, Louisville, Kentucky, United States of America
| | - Atul Chugh
- Institute of Molecular Cardiology, University of Louisville, Louisville, Kentucky, United States of America
| | - Aruni Bhatnagar
- Institute of Molecular Cardiology, University of Louisville, Louisville, Kentucky, United States of America
| | - Roberto Bolli
- Institute of Molecular Cardiology, University of Louisville, Louisville, Kentucky, United States of America
- * E-mail:
| |
Collapse
|
43
|
Zhou J, Zhang Y, Wen X, Cao J, Li D, Lin Q, Wang H, Liu Z, Duan C, Wu K, Wang C. Telocytes accompanying cardiomyocyte in primary culture: two- and three-dimensional culture environment. J Cell Mol Med 2011; 14:2641-5. [PMID: 21158014 PMCID: PMC4373485 DOI: 10.1111/j.1582-4934.2010.01186.x] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Recently, the presence of telocytes was demonstrated in human and mammalian tissues and organs (digestive and extra-digestive organs, genitourinary organs, heart, placenta, lungs, pleura, striated muscle). Noteworthy, telocytes seem to play a significant role in the normal function and regeneration of myocardium. By cultures of telocytes in two- and three-dimensional environment we aimed to study the typical morphological features as well as functionality of telocytes, which will provide important support to understand their in vivo roles. Neonatal rat cardiomyocytes were isolated and cultured as seeding cells in vitro in two-dimensional environment. Furthermore, engineered myocardium tissue was constructed from isolated cells in three-dimensional collagen/Matrigel scaffolds. The identification of telocytes was performed by using histological and immunohistochemical methods. The results showed that typical telocytes are distributed among cardiomyocytes, connecting them by long telopodes. Telocytes have a typical fusiform cell body with two or three long moniliform telopodes, as main characteristics. The vital methylene blue staining showed the existence of telocytes in primary culture. Immunohistochemistry demonstrated that some c-kit or CD34 immuno-positive cells in engineered heart tissue had the morphology of telocytes, with a typical fusiform cell body and long moniliform telopodes. Also, a significant number of vimentin+ telocytes were present within engineered heart tissue. We suggest that the model of three-dimensional engineered heart tissue could be useful for the ongoing research on the functional relationships of telocytes with cardiomyocytes. Because the heart has the necessary potential of changing the muscle and non-muscle cells during the lifetime, telocytes might play an active role in the heart regeneration process. Moreover, telocytes might be a useful tool for cardiac tissue engineering.
Collapse
Affiliation(s)
- Jin Zhou
- Department of Tissue Engineering, Institute of Basic Medical Sciences and Tissue Engineering Research Center, Academy of Military Medical Sciences, Beijing, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|