1
|
Bondeelle L, Clément S, Bergeron A, Tapparel C. Lung stem cells and respiratory epithelial chimerism in transplantation. Eur Respir Rev 2025; 34:240146. [PMID: 39971397 PMCID: PMC11836672 DOI: 10.1183/16000617.0146-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 11/21/2024] [Indexed: 02/21/2025] Open
Abstract
Stem cells are capable of self-renewal and differentiation into specialised types. They range from totipotent cells to multipotent or somatic stem cells and ultimately to unipotent cells. Some adult multipotent stem cells can have the potential to regenerate and colonise diverse tissues. The respiratory airways and lung mucosa, exposed to ambient air, perform vital roles for all human tissues and organs. They serve as barriers against airborne threats and are essential for tissue oxygenation. Despite low steady-state turnover, lungs are vulnerable to injuries and diseases from environmental exposure. Lung stem cells are crucial due to their regenerative potential and ability to replace damaged cells. Lung repair with extrapulmonary stem cells can occur, leading to the coexistence of respiratory cells with different genetic origins, a phenomenon known as airway epithelial chimerism. The impact of such chimerism in lung repair and disease is actively studied. This review explores different stem cell types, focusing on pulmonary stem cells. It discusses airway epithelium models derived from stem cells for studying lung diseases and examines lung chimerism, particularly in lung transplantation and haematopoietic stem cell transplantation, highlighting its significance in understanding tissue repair and chimerism-mediated repair processes in lung pathology.
Collapse
Affiliation(s)
- Louise Bondeelle
- Department of Microbiology and Molecular Medicine, University of Geneva, Geneva, Switzerland
| | - Sophie Clément
- Department of Microbiology and Molecular Medicine, University of Geneva, Geneva, Switzerland
| | - Anne Bergeron
- Pneumology Department, Geneva University Hospitals, Geneva, Switzerland
- Co-last author
| | - Caroline Tapparel
- Department of Microbiology and Molecular Medicine, University of Geneva, Geneva, Switzerland
- Co-last author
| |
Collapse
|
2
|
Baghaie L, Haxho F, Leroy F, Lewis B, Wawer A, Minhas S, Harless WW, Szewczuk MR. Contemporaneous Perioperative Inflammatory and Angiogenic Cytokine Profiles of Surgical Breast, Colorectal, and Prostate Cancer Patients: Clinical Implications. Cells 2023; 12:2767. [PMID: 38067195 PMCID: PMC10706122 DOI: 10.3390/cells12232767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 11/29/2023] [Accepted: 12/02/2023] [Indexed: 12/18/2023] Open
Abstract
Surgery-induced tumor growth acceleration and synchronous metastatic growth promotion have been observed for decades. Surgery-induced wound healing, orchestrated through growth factors, chemokines, and cytokines, can negatively impact patients harboring residual or metastatic disease. We provide detailed clinical evidence of this process in surgical breast, prostate, and colorectal cancer patients. Plasma samples were analyzed from 68 cancer patients who had not received treatment before surgery or adjuvant therapy until at least four weeks post-surgery. The levels of plasma cytokines, chemokines, and growth factors were simultaneously quantified and profiled using multiplexed immunoassays for eight time points sampled per patient. The immunologic processes are induced immediately after surgery in patients, characterized by a drastic short-term shift in the expression levels of pro-inflammatory and angiogenic molecules and cytokines. A rapid and significant spike in circulating plasma levels of hepatocyte growth factor (HGF), interleukin-6 (IL-6), placental growth factor (PLGF), and matrix metalloproteinase-9 (MMP-9) after surgery was noted. The rise in these molecules was concomitant with a significant drop in transforming growth factor-β1 (TGF-β1), platelet-derived growth factor (PDGF-AB/BB), insulin-like growth factor-1 (IGF-1), and monocyte chemoattractant protein-2 (MCP-2). If not earlier, each plasma analyte was normalized to baseline levels within 1-2 weeks after surgery, suggesting that surgical intervention alone was responsible for these effects. The effects of surgical tumor removal on disrupting the pro-inflammatory and angiogenic plasma profiles of cancer patients provide evidence for potentiating malignant progression. Our findings indicate a narrow therapeutic window of opportunity after surgery to prevent disease recurrence.
Collapse
Affiliation(s)
- Leili Baghaie
- Department of Biomedical & Molecular Sciences, Queen’s University, Kingston, ON K7L 3N6, Canada; (L.B.); (F.H.); (F.L.)
| | - Fiona Haxho
- Department of Biomedical & Molecular Sciences, Queen’s University, Kingston, ON K7L 3N6, Canada; (L.B.); (F.H.); (F.L.)
- Dermatology Residency Program, the Cumming School of Medicine, University of Calgary, Calgary, AB T2T 5C7, Canada
| | - Fleur Leroy
- Department of Biomedical & Molecular Sciences, Queen’s University, Kingston, ON K7L 3N6, Canada; (L.B.); (F.H.); (F.L.)
- Faculté de Médecine, Maïeutique et Sciences de la Santé, Université de Strasbourg, F-67000 Strasbourg, France
| | - Beth Lewis
- ENCYT Technologies Inc., Membertou, NS B1S 0H1, Canada; (B.L.); (A.W.); (S.M.)
| | - Alexander Wawer
- ENCYT Technologies Inc., Membertou, NS B1S 0H1, Canada; (B.L.); (A.W.); (S.M.)
| | - Shamano Minhas
- ENCYT Technologies Inc., Membertou, NS B1S 0H1, Canada; (B.L.); (A.W.); (S.M.)
| | - William W. Harless
- ENCYT Technologies Inc., Membertou, NS B1S 0H1, Canada; (B.L.); (A.W.); (S.M.)
| | - Myron R. Szewczuk
- Department of Biomedical & Molecular Sciences, Queen’s University, Kingston, ON K7L 3N6, Canada; (L.B.); (F.H.); (F.L.)
| |
Collapse
|
3
|
Pishel I. Immune system rejuvenation—approaches and real achievements. EXPLORATION OF IMMUNOLOGY 2023:325-340. [DOI: 10.37349/ei.2023.00105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 04/17/2023] [Indexed: 01/03/2025]
Abstract
Interest in the mechanisms of aging of the immune system has not faded over the past 100 years, and it is caused by the immune-mediated development of age-related pathology, including autoimmune organ damage, reduced vaccination efficiency, atherosclerosis, the development of cardiovascular pathology, etc. In contrast to many other organs and systems, the immune system aging begins at an early age and has more pronounced changes that lead to the development of secondary pathology, which significantly affects life expectancy. But an effective strategy to restore immune function has not been developed yet. During this time, the mechanisms of age-related dysfunction of organs and cells of both the adaptive and innate immune systems were studied in detail—thymus involution, a decrease in the potential of hematopoietic stem cells, impaired differentiation and functions of immunocompetent cells, as well as the ways of their interaction. Numerous potential therapeutic targets have been identified and various approaches have been used to implement such therapeutic interventions. The review is devoted to replacement therapy using transplantation of hematopoietic stem cells (HSCs) and young lymphoid cells and tissues, cellular and systemic factor exchange in heterochronic parabiosis, and some other widely used life extension approaches. It has been proven that cell therapy using young cells to rejuvenate the old immune system, unfortunately, often turns out to be ineffective because it does not eliminate the root cause of age-related changes. The phenomenon of inflamm-aging that develops with age can significantly affect both the aging of the organism in general and the functioning of immunocompetent cells in particular. Therefore, the most promising direction in the restoration of immune functions during aging is systemic approaches that have a complex effect on the organism as a whole and can slow down the aging process.
Collapse
Affiliation(s)
- Iryna Pishel
- Lab Applied Pharmacology and Toxicology, Bienta/Enamine Ltd, 02094 Kyiv, Ukraine
| |
Collapse
|
4
|
Quesenberry PJ, Wen S, Goldberg LR, Dooner MS. The universal stem cell. Leukemia 2022; 36:2784-2792. [PMID: 36307485 PMCID: PMC9712109 DOI: 10.1038/s41375-022-01715-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/26/2022] [Accepted: 09/22/2022] [Indexed: 11/08/2022]
Abstract
Current dogma is that there exists a hematopoietic pluripotent stem cell, resident in the marrow, which is quiescent, but with tremendous proliferative and differentiative potential. Furthermore, the hematopoietic system is essentially hierarchical with progressive differentiation from the pluripotent stem cells to different classes of hematopoietic cells. However, results summarized here indicate that the marrow pluripotent hematopoietic stem cell is actively cycling and thus continually changing phenotype. As it progresses through cell cycle differentiation potential changes as illustrated by sequential changes in surface expression of B220 and GR-1 epitopes. Further data indicated that the potential of purified hematopoietic stem cells extends to multiple other non-hematopoietic cells. It appears that marrow stem cells will give rise to epithelial pulmonary cells at certain points in cell cycle. Thus, it appears that the marrow "hematopoietic" stem cell is also a stem cell for other non-hematopoietic tissues. These observations give rise to the concept of a universal stem cell. The marrow stem cell is not limited to hematopoiesis and its differentiation potential continually changes as it transits cell cycle. Thus, there is a universal stem cell in the marrow which alters its differentiation potential as it progresses through cell cycle. This potential is expressed when it resides in tissues compatible with its differentiation potential, at a particular point in cell cycle transit, or when it interacts with vesicles from that tissue.
Collapse
Affiliation(s)
- Peter J Quesenberry
- Division of Hematology/Oncology, Brown University, Rhode Island Hospital, Providence, RI, 02903, USA.
| | - Sicheng Wen
- Division of Hematology/Oncology, Brown University, Rhode Island Hospital, Providence, RI, 02903, USA
| | - Laura R Goldberg
- Division of Hematology/Oncology, Brown University, Rhode Island Hospital, Providence, RI, 02903, USA
- Division of Hematology, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Mark S Dooner
- Division of Hematology/Oncology, Brown University, Rhode Island Hospital, Providence, RI, 02903, USA
| |
Collapse
|
5
|
Liu C, Chi K, Geng X, Hong Q, Mao Z, Huang Q, Liu D, Wang Y, Zhang Y, Zhou F, Cai G, Chen X, Sun X. Exogenous Biological Renal Support Improves Kidney Function in Mice With Rhabdomyolysis-Induced Acute Kidney Injury. Front Med (Lausanne) 2021; 8:655787. [PMID: 34124093 PMCID: PMC8193099 DOI: 10.3389/fmed.2021.655787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 03/16/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Rhabdomyolysis (RM) is a clinical syndrome characterized by breakdown of skeletal muscle fibers and release of their contents into the circulation. Myoglobin-induced acute kidney injury (AKI) is one of the most severe complications of RM. Based on our previous research, exogenous biological renal support alleviates renal ischemia–reperfusion injury in elderly mice. This study aimed to determine whether exogenous biological renal support promotes renal recovery from RM-induced AKI and to preliminarily explore the mechanisms involved. Methods: A parabiosis animal model was established to investigate the effects of exogenous biological renal support on RM-induced AKI. Mice were divided into three groups: the control group (in which mice were injected with sterile saline), the RM group (in which mice were injected with 8 mL/kg glycerol), and the parabiosis + RM group (in which recipient mice were injected with glycerol 3 weeks after parabiosis model establishment). Blood samples and kidney tissue were collected for further processing 48 h after RM induction. Bioinformatics analysis was conducted via Gene Ontology analysis, Kyoto Encyclopedia of Genes and Genomes pathway analysis, functional enrichment analysis, and clustering analysis. Results: No mice died within 48 h after the procedure. Exogenous biological renal support attenuated the histological and functional deterioration in mice with RM-induced AKI. Bioinformatics analysis identified key pathways and proteins involved in this process. We further demonstrated that exogenous biological renal support ameliorated AKI through multiple mechanisms, including by suppressing the complement system; attenuating oxidative stress, inflammation, and cell death; and increasing proliferation. Conclusions: Exogenous biological renal support provided by parabiosis can improve renal function in RM-induced AKI by suppressing the complement system; decreasing oxidative stress, inflammation, and cell death; and promoting tubular cell proliferation. Our study provides basic research evidence for the use of bioartificial kidneys to treat RM-induced AKI.
Collapse
Affiliation(s)
- Chao Liu
- Medical School of Chinese People's Liberation Army (PLA), Beijing, China.,State Key Laboratory of Kidney Diseases, Department of Nephrology, National Clinical Research Center for Kidney Diseases, Chinese People's Liberation Army (PLA) Institute of Nephrology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Kun Chi
- Medical School of Chinese People's Liberation Army (PLA), Beijing, China.,State Key Laboratory of Kidney Diseases, Department of Nephrology, National Clinical Research Center for Kidney Diseases, Chinese People's Liberation Army (PLA) Institute of Nephrology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Xiaodong Geng
- Medical School of Chinese People's Liberation Army (PLA), Beijing, China.,State Key Laboratory of Kidney Diseases, Department of Nephrology, National Clinical Research Center for Kidney Diseases, Chinese People's Liberation Army (PLA) Institute of Nephrology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Quan Hong
- Medical School of Chinese People's Liberation Army (PLA), Beijing, China.,State Key Laboratory of Kidney Diseases, Department of Nephrology, National Clinical Research Center for Kidney Diseases, Chinese People's Liberation Army (PLA) Institute of Nephrology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Zhi Mao
- Department of Critical Care Medicine, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Qi Huang
- Department of Nephrology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Dong Liu
- Department of Nephrology, Air Force Medical Center, People's Liberation Army (PLA), Beijing, China
| | - Yiqin Wang
- State Key Laboratory of Kidney Diseases, Department of Nephrology, National Clinical Research Center for Kidney Diseases, Chinese People's Liberation Army (PLA) Institute of Nephrology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Ying Zhang
- Department of Ultrasound, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Feihu Zhou
- Department of Critical Care Medicine, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Guangyan Cai
- State Key Laboratory of Kidney Diseases, Department of Nephrology, National Clinical Research Center for Kidney Diseases, Chinese People's Liberation Army (PLA) Institute of Nephrology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Xiangmei Chen
- State Key Laboratory of Kidney Diseases, Department of Nephrology, National Clinical Research Center for Kidney Diseases, Chinese People's Liberation Army (PLA) Institute of Nephrology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Xuefeng Sun
- State Key Laboratory of Kidney Diseases, Department of Nephrology, National Clinical Research Center for Kidney Diseases, Chinese People's Liberation Army (PLA) Institute of Nephrology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| |
Collapse
|
6
|
Heterogenetic parabiosis between healthy and dystrophic mice improve the histopathology in muscular dystrophy. Sci Rep 2020; 10:7075. [PMID: 32341395 PMCID: PMC7184587 DOI: 10.1038/s41598-020-64042-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 04/09/2020] [Indexed: 11/10/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a progressive muscle disease, characterized by mutations in the X-linked dystrophin, that has several therapeutic options but no curative treatment. Transplantation of muscle progenitor cells for treatment of DMD has been widely investigated; however, its application is hindered by limited cell survival due to the harmful dystrophic microenvironment. An alternative approach to utilize progenitor cells and circulatory factors and to improve the dystrophic muscle pathology and microenvironment is through parabiotic pairing, where mice are surgically sutured to create a joint circulatory system. Parabiotic mice were generated by surgically joining wild type (WT) mice expressing green fluorescent protein (GFP) with mdx mice. These mice developed a common circulation (approximately 50% green cells in the blood of mdx mice) 2-weeks after parabiotic pairing. We observed significantly improved dystrophic muscle pathology, including decreased inflammation, necrotic fibers and fibrosis in heterogenetic parabionts. Importantly, the GFP + cells isolated from the mdx mice (paired with GFP mice) underwent myogenic differentiation in vitro and expressed markers of mesenchymal stem cells and macrophages, which may potentially be involved in the improvement of dystrophic muscle pathology. These observations suggest that changing the dystrophic microenvironment can be a new approach to treat DMD.
Collapse
|
7
|
Bone Marrow-Derived CD44 + Cells Migrate to Tissue-Engineered Constructs via SDF-1/CXCR4-JNK Pathway and Aid Bone Repair. Stem Cells Int 2019; 2019:1513526. [PMID: 31428156 PMCID: PMC6681616 DOI: 10.1155/2019/1513526] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Revised: 06/05/2019] [Accepted: 06/18/2019] [Indexed: 12/22/2022] Open
Abstract
Background and Aims Host-derived cells play crucial roles in the regeneration process of tissue-engineered constructs (TECs) during the treatment of large segmental bone defects (LSBDs). However, their identity, source, and cell recruitment mechanisms remain elusive. Methods A complex model was created using mice by combining methods of GFP+ bone marrow transplantation (GFP-BMT), parabiosis (GFP+-BMT and wild-type mice), and femoral LSBD, followed by implantation of TECs or DBM scaffolds. Postoperatively, the migration of host BM cells was detected by animal imaging and immunofluorescent staining. Bone repair was evaluated by micro-CT. Signaling pathway repressors including AMD3100 and SP600125 associated with the migration of BM CD44+ cells were further investigated. In vitro, transwell migration and western-blotting assays were performed to verify the related signaling pathway. In vivo, the importance of the SDF-1/CXCR4-JNK pathway was validated by ELISA, fluorescence-activated cell sorting (FACS), immunofluorescent staining, and RT-PCR. Results First, we found that host cells recruited to facilitate TEC-mediated bone repair were derived from bone marrow and most of them express CD44, indicating the significance of CD44 in the migration of bone marrow cells towards donor MSCs. Then, the predominant roles of SDF-1/CXCR4 and downstream JNK in the migration of BM CD44+ cells towards TECs were demonstrated. Conclusion Together, we demonstrated that during bone repair promoted by TECs, BM-derived CD44+ cells were essential and their migration towards TECs could be regulated by the SDF-1/CXCR4-JNK signaling pathway.
Collapse
|
8
|
Barwinska D, Oueini H, Poirier C, Albrecht ME, Bogatcheva NV, Justice MJ, Saliba J, Schweitzer KS, Broxmeyer HE, March KL, Petrache I. AMD3100 ameliorates cigarette smoke-induced emphysema-like manifestations in mice. Am J Physiol Lung Cell Mol Physiol 2018; 315:L382-L386. [PMID: 29745251 DOI: 10.1152/ajplung.00185.2018] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
We have shown that cigarette smoke (CS)-induced pulmonary emphysema-like manifestations are preceded by marked suppression of the number and function of bone marrow hematopoietic progenitor cells (HPCs). To investigate whether a limited availability of HPCs may contribute to CS-induced lung injury, we used a Food and Drug Administration-approved antagonist of the interactions of stromal cell-derived factor 1 (SDF-1) with its chemokine receptor CXCR4 to promote intermittent HPC mobilization and tested its ability to limit emphysema-like injury following chronic CS. We administered AMD3100 (5mg/kg) to mice during a chronic CS exposure protocol of up to 24 wk. AMD3100 treatment did not affect either lung SDF-1 levels, which were reduced by CS, or lung inflammatory cell counts. However, AMD3100 markedly improved CS-induced bone marrow HPC suppression and significantly ameliorated emphysema-like end points, such as alveolar airspace size, lung volumes, and lung static compliance. These results suggest that antagonism of SDF-1 binding to CXCR4 is associated with protection of both bone marrow and lungs during chronic CS exposure, thus encouraging future studies of potential therapeutic benefit of AMD3100 in emphysema.
Collapse
Affiliation(s)
- Daria Barwinska
- Department of Cellular and Integrative Physiology, Indiana University , Indianapolis, Indiana.,Indiana Center for Vascular Biology and Medicine, Indiana University , Indianapolis, Indiana.,Vascular and Cardiac Center for Adult Stem Cell Therapy Signature Center, Indiana University, Purdue University , Indianapolis, Indiana.,Roudebush Veterans Affairs Medical Center, Indiana University , Indianapolis, Indiana.,Division of Nephrology, Department of Medicine, Indiana University , Indianapolis, Indiana
| | - Houssam Oueini
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Indiana University , Indianapolis, Indiana
| | - Christophe Poirier
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Indiana University , Indianapolis, Indiana
| | - Marjorie E Albrecht
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Indiana University , Indianapolis, Indiana
| | - Natalia V Bogatcheva
- Indiana Center for Vascular Biology and Medicine, Indiana University , Indianapolis, Indiana.,Vascular and Cardiac Center for Adult Stem Cell Therapy Signature Center, Indiana University, Purdue University , Indianapolis, Indiana.,Roudebush Veterans Affairs Medical Center, Indiana University , Indianapolis, Indiana.,Division of Cardiology, Department of Medicine, Indiana University , Indianapolis, Indiana
| | - Matthew J Justice
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Indiana University , Indianapolis, Indiana.,Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, National Jewish Health, Denver, Colorado
| | - Jacob Saliba
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Indiana University , Indianapolis, Indiana
| | - Kelly S Schweitzer
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Indiana University , Indianapolis, Indiana.,Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, National Jewish Health, Denver, Colorado
| | - Hal E Broxmeyer
- Department of Microbiology and Immunology, Indiana University , Indianapolis, Indiana
| | - Keith L March
- Indiana Center for Vascular Biology and Medicine, Indiana University , Indianapolis, Indiana.,Vascular and Cardiac Center for Adult Stem Cell Therapy Signature Center, Indiana University, Purdue University , Indianapolis, Indiana.,Roudebush Veterans Affairs Medical Center, Indiana University , Indianapolis, Indiana.,Division of Cardiology, Department of Medicine, Indiana University , Indianapolis, Indiana.,Division of Cardiovascular Medicine and Center for Regenerative Medicine, University of Florida , Gainesville, Florida
| | - Irina Petrache
- Indiana Center for Vascular Biology and Medicine, Indiana University , Indianapolis, Indiana.,Vascular and Cardiac Center for Adult Stem Cell Therapy Signature Center, Indiana University, Purdue University , Indianapolis, Indiana.,Roudebush Veterans Affairs Medical Center, Indiana University , Indianapolis, Indiana.,Division of Pulmonary and Critical Care Medicine, Department of Medicine, Indiana University , Indianapolis, Indiana.,Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, National Jewish Health, Denver, Colorado.,Department of Medicine, University of Colorado , Denver, Colorado
| |
Collapse
|
9
|
Firsova AB, Bird AD, Abebe D, Ng J, Mollard R, Cole TJ. Fresh Noncultured Endothelial Progenitor Cells Improve Neonatal Lung Hyperoxia-Induced Alveolar Injury. Stem Cells Transl Med 2017; 6:2094-2105. [PMID: 29027762 PMCID: PMC5702522 DOI: 10.1002/sctm.17-0093] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 09/05/2017] [Indexed: 01/01/2023] Open
Abstract
Treatment of preterm human infants with high oxygen can result in disrupted lung alveolar and vascular development. Local or systemic administration of endothelial progenitor cells (EPCs) is reported to remedy such disruption in animal models. In this study, the effects of both fresh (enriched for KDR) and cultured bone marrow (BM)-derived cell populations with EPC characteristics were examined following hyperoxia in neonatal mouse lungs. Intraperitoneal injection of fresh EPCs into five-day-old mice treated with 90% oxygen resulted in full recovery of hyperoxia-induced alveolar disruption by 56 days of age. Partial recovery in septal number following hyperoxia was observed following injection of short-term cultured EPCs, yet aberrant tissue growths appeared following injection of long-term cultured cells. Fresh and long-term cultured cells had no impact on blood vessel development. Short-term cultured cells increased blood vessel number in normoxic and hyperoxic mice by 28 days but had no impact on day 56. Injection of fresh EPCs into normoxic mice significantly reduced alveolarization compared with phosphate buffered saline-injected normoxic controls. These results indicate that fresh BM EPCs have a higher and safer corrective profile in a hyperoxia-induced lung injury model compared with cultured BM EPCs but may be detrimental to the normoxic lung. The appearance of aberrant tissue growths and other side effects following injection of cultured EPCs warrants further investigation. Stem Cells Translational Medicine 2017;6:2094-2105.
Collapse
Affiliation(s)
- Alexandra B Firsova
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - A Daniel Bird
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Degu Abebe
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Judy Ng
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Richard Mollard
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia.,Department of Veterinary and Agricultural Science, University of Melbourne, Parkville, Victoria, Australia
| | - Timothy J Cole
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
10
|
Mussar K, Pardike S, Hohl TM, Hardiman G, Cirulli V, Crisa L. A CCR2+ myeloid cell niche required for pancreatic β cell growth. JCI Insight 2017; 2:93834. [PMID: 28768911 DOI: 10.1172/jci.insight.93834] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 06/27/2017] [Indexed: 12/11/2022] Open
Abstract
Organ-specific patterns of myeloid cells may contribute tissue-specific growth and/or regenerative potentials. The perinatal stage of pancreas development marks a time characterized by maximal proliferation of pancreatic islets, ensuring the maintenance of glucose homeostasis throughout life. Ontogenically distinct CX3CR1+ and CCR2+ macrophage populations have been reported in the adult pancreas, but their functional contribution to islet cell growth at birth remains unknown. Here, we uncovered a temporally restricted requirement for CCR2+ myeloid cells in the perinatal proliferation of the endocrine pancreatic epithelium. CCR2+ macrophages are transiently enriched over CX3CR1+ subsets in the neonatal pancreas through both local expansion and recruitment of immature precursors. Using CCR2-specific depletion models, we show that loss of this myeloid population leads to a striking reduction in β cell proliferation, dysfunctional islet phenotypes, and glucose intolerance in newborns. Replenishment of pancreatic CCR2+ myeloid compartments by adoptive transfer rescues these defects. Gene profiling identifies pancreatic CCR2+ myeloid cells as a prominent source of IGF2, which contributes to IGF1R-mediated islet proliferation. These findings uncover proproliferative functions of CCR2+ myeloid subsets and identify myeloid-dependent regulation of IGF signaling as a local cue supporting pancreatic proliferation.
Collapse
Affiliation(s)
- Kristin Mussar
- Department of Medicine and Institute of Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington, USA
| | - Stephanie Pardike
- Department of Medicine and Institute of Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington, USA
| | - Tobias M Hohl
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Gary Hardiman
- Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Vincenzo Cirulli
- Department of Medicine and Institute of Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington, USA
| | - Laura Crisa
- Department of Medicine and Institute of Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington, USA
| |
Collapse
|
11
|
Qiu M, Chen Y, Ye Q. Risk factors for acute exacerbation of idiopathic pulmonary fibrosis: A systematic review and meta-analysis. CLINICAL RESPIRATORY JOURNAL 2017; 12:1084-1092. [PMID: 28332341 DOI: 10.1111/crj.12631] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2016] [Revised: 01/15/2017] [Accepted: 03/07/2017] [Indexed: 11/29/2022]
Affiliation(s)
- Meihua Qiu
- Department of Occupational Medicine and Toxicology; Beijing Institute of Respiratory Medicine, Beijing Chao-Yang Hospital, Capital Medical University; Beijing China
| | - Yuqing Chen
- Department of Occupational Medicine and Toxicology; Beijing Institute of Respiratory Medicine, Beijing Chao-Yang Hospital, Capital Medical University; Beijing China
| | - Qiao Ye
- Department of Occupational Medicine and Toxicology; Beijing Institute of Respiratory Medicine, Beijing Chao-Yang Hospital, Capital Medical University; Beijing China
| |
Collapse
|
12
|
Ahmadi M, Rahbarghazi R, Soltani S, Aslani MR, Keyhanmanesh R. Contributory Anti-Inflammatory Effects of Mesenchymal Stem Cells, Not Conditioned Media, On Ovalbumin-Induced Asthmatic Changes in Male Rats. Inflammation 2017; 39:1960-1971. [PMID: 27590236 DOI: 10.1007/s10753-016-0431-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Our aim in selecting an appropriate cell fraction and conditioned media (CM) was to achieve the suitable candidate for ameliorating long-term chronic asthmatic changes of respiratory tract. Thirty-six rats were classified into healthy and sensitized groups, which were further divided into three subgroups; rats received systemically 50 μl volume of PBS, CM, or 2 × 106 rat bone marrow-derived mesenchymal stem cells (rBMMSCs). Tracheal responsiveness (TR), immunologic responses, and recruitment of rBMMSCs into the lungs were evaluated. A high degree of TR and total WBC and percentages of eosinophils and neutrophils was significantly recorded in all sensitized groups rather than of controls (p < 0.001 to p < 0.05). Concurrently, a significant improvement of TR and eosinophil and neutrophil return toward normal levels was evident in sensitized rats receiving cells as compared to parallel asthmatic animals. Flow cytometric monitoring of lymphocyte subpopulation revealed a decrease in the number of CD3+CD4+ and concurrent increase in CD3+CD8+ in all sensitized rats as compared to control (p < 0.001 to p < 0.05). Noticeably, no significant modulatory effects of either cell or CM administration were achieved on the CD3+CD4+ and CD3+CD8+ populations in non-asthmatic rats. Corroborating our results, the number of CD3+CD4+ tended to increase (p < 0.05) which coincided with a decreased manner of CD3+CD8+ populations as compared to other asthmatic groups (p < 0.01 to p < 0.05). Moreover, stem cells could efficiently transmigrate to the lung parenchyma, albeit the dynamic of asthmatic changes stimulated the rate of recruited cells. Our study shed light on superior effects of mesenchymal stem cells, but not CM, in attenuating chronic asthmatic changes in the model of rat.
Collapse
Affiliation(s)
- Mahdi Ahmadi
- Department of Physiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sina Soltani
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Reza Aslani
- Department of Physiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Rana Keyhanmanesh
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
13
|
Maria OM, Maria AM, Ybarra N, Jeyaseelan K, Lee S, Perez J, Shalaby MY, Lehnert S, Faria S, Serban M, Seuntjens J, El Naqa I. Mesenchymal Stem Cells Adopt Lung Cell Phenotype in Normal and Radiation-induced Lung Injury Conditions. Appl Immunohistochem Mol Morphol 2016. [PMID: 26200842 DOI: 10.1097/pai.0000000000000180] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Lung tissue exposure to ionizing irradiation can invariably occur during the treatment of a variety of cancers leading to increased risk of radiation-induced lung disease (RILD). Mesenchymal stem cells (MSCs) possess the potential to differentiate into epithelial cells. However, cell culture methods of primary type II pneumocytes are slow and cannot provide a sufficient number of cells to regenerate damaged lungs. Moreover, effects of ablative radiation doses on the ability of MSCs to differentiate in vitro into lung cells have not been investigated yet. Therefore, an in vitro coculture system was used, where MSCs were physically separated from dissociated lung tissue obtained from either healthy or high ablative doses of 16 or 20 Gy whole thorax irradiated rats. Around 10±5% and 20±3% of cocultured MSCs demonstrated a change into lung-specific Clara and type II pneumocyte cells when MSCs were cocultured with healthy lung tissue. Interestingly, in cocultures with irradiated lung biopsies, the percentage of MSCs changed into Clara and type II pneumocytes cells increased to 40±7% and 50±6% at 16 Gy irradiation dose and 30±5% and 40±8% at 20 Gy irradiation dose, respectively. These data suggest that MSCs to lung cell differentiation is possible without cell fusion. In addition, 16 and 20 Gy whole thorax irradiation doses that can cause varying levels of RILD, induced different percentages of MSCs to adopt lung cell phenotype compared with healthy lung tissue, providing encouraging outlook for RILD therapeutic intervention for ablative radiotherapy prescriptions.
Collapse
Affiliation(s)
- Ola M Maria
- *Medical Physics Unit, Department of Oncology, Radiation Oncology Division, McGill University, Montreal General Hospital ‡Department of Oncology, Radiation Oncology Division, McGill University Health Centre ∥International Baccalaureate, Marymount Academy, Montreal, QC, Canada †Department of Oral Biology, Faculty of Dentistry, Mansoura University, Mansoura §Faculty of Medicine, Tanta University, Tanta, Egypt
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Bone marrow mesenchymal stem cells and their conditioned media could potentially ameliorate ovalbumin-induced asthmatic changes. Biomed Pharmacother 2016; 85:28-40. [PMID: 27930984 DOI: 10.1016/j.biopha.2016.11.127] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 11/20/2016] [Accepted: 11/27/2016] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND The major feature of asthma is governed by chronic airway inflammation. This investigation was proposed to achieve the suitable candidate for ameliorating long-term chronic asthmatic changes of respiratory tract. METHODS 36 rats were classified into healthy (C) and ovalbumin (OVA)-sensitized animals (S). To sensitize, the rats were exposed to OVA over a course of 32±1days. One day after sensitization, equal six different groups were subjected to experimental procedure (n=6); Rats only received intratracheally 50ml PBS (CPT and SPT groups), 50μl conditioned medium (CM) (CST and SST groups) and 50μl PBS containing 2×106 rat bone marrow-derived mesenchymal stem cells (rBMMSCs) (CCT and SCT groups). Two weeks after treatment, tracheal responsiveness, immunologic responses and recruitment of rBMMSCs into the lung as well as pathological changes were evaluated. RESULTS A high degree of tracheal responsiveness, total white blood cell and percentages of eosinophil and neutrophil was significantly recorded in all sensitized groups rather than of controls (p<0.001 to p<0.05). Of interest, all above-mentioned parameters decreased significantly in SST and notably SCT groups as compared to S group (p<0.001 to p<0.05). The results revealed decrease number of blood CD3+CD4+ and concurrent increase in CD3+CD8+ in all sensitized rats as compared to control (p<0.001 to p<0.05). Noticeably, no significant modulatory effects of either cell or CM administration were achieved on the CD3+CD4+ and CD3+CD8+ populations in non-asthmatic rats. Moreover, the number of CD3+CD4+ in SST and SCT groups tended to increase, which coincided with a decreased manner of CD3+CD8+ populations as compared with S group (p<0.001 to p<0.05). However, the CD3+CD4+ cells in SCT rats were significantly higher than the group SST (p<0.01) whereas CD3+CD8+ cells diminished simultaneously (p<0.001). Real-time PCR analysis further showed that both CM and particularly MSCs changed the expression of interleukin (IL)-4 and IL-10 in the asthmatic groups to the near level of control rats (p<0.001 to p<0.05). Histopathological analysis revealed a profound reduction of lungs injuries in asthmatic rats when received CM and peculiarly mesenchymal stem cells (p<0.01 to p<0.05). CONCLUSION Our study shed light on the superior effects of rBMMSCs, rather than CM, in attenuating of chronic asthmatic changes in the rat model.
Collapse
|
15
|
Siavashi V, Asadian S, Taheri-Asl M, Babaei H, Keshavarz S, Bazaei M, Nassiri SM. The Improvement of Respiratory Performance After Phototherapy-Induced EPC Mobilization in Preterm Infants With RDS. J Cell Biochem 2016; 118:594-604. [DOI: 10.1002/jcb.25745] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 09/19/2016] [Indexed: 01/03/2023]
Affiliation(s)
- Vahid Siavashi
- Department of Clinical Pathology; Faculty of Veterinary Medicine; University of Tehran; Tehran Iran
| | - Simin Asadian
- Imam Reza Hospitals; Kermanshah University of Medical Sciences; Kermanshah Iran
| | - Masoud Taheri-Asl
- Departments of Radiology; Faculty of Paramedicine; AJA University of Medical Sciences; Tehran Iran
| | - Homa Babaei
- Department of Pediatrics; Imam Reza Hospital; Kermanshah University of Medical Science; Kermanshah Iran
| | | | - Mohammad Bazaei
- Department of Clinical Pathology; Faculty of Veterinary Medicine; University of Tehran; Tehran Iran
| | - Seyed Mahdi Nassiri
- Department of Clinical Pathology; Faculty of Veterinary Medicine; University of Tehran; Tehran Iran
| |
Collapse
|
16
|
Xiang MSW, Kikuchi K. Endogenous Mechanisms of Cardiac Regeneration. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2016; 326:67-131. [PMID: 27572127 DOI: 10.1016/bs.ircmb.2016.04.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Zebrafish possess a remarkable capacity for cardiac regeneration throughout their lifetime, providing a model for investigating endogenous cellular and molecular mechanisms regulating myocardial regeneration. By contrast, adult mammals have an extremely limited capacity for cardiac regeneration, contributing to mortality and morbidity from cardiac diseases such as myocardial infarction and heart failure. However, the viewpoint of the mammalian heart as a postmitotic organ was recently revised based on findings that the mammalian heart contains multiple undifferentiated cell types with cardiogenic potential as well as a robust regenerative capacity during a short period early in life. Although it occurs at an extremely low level, continuous cardiomyocyte turnover has been detected in adult mouse and human hearts, which could potentially be enhanced to restore lost myocardium in damaged human hearts. This review summarizes and discusses recent advances in the understanding of endogenous mechanisms of cardiac regeneration.
Collapse
Affiliation(s)
- M S W Xiang
- Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst NSW, Australia
| | - K Kikuchi
- Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst NSW, Australia; St. Vincent's Clinical School, University of New South Wales, Kensington NSW, Australia.
| |
Collapse
|
17
|
Yang C, Jiang J, Yang X, Wang H, Du J. Stem/progenitor cells in endogenous repairing responses: new toolbox for the treatment of acute lung injury. J Transl Med 2016; 14:47. [PMID: 26865361 PMCID: PMC4750219 DOI: 10.1186/s12967-016-0804-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 01/27/2016] [Indexed: 02/07/2023] Open
Abstract
The repair of organs and tissues has stepped into a prospective era of regenerative medicine. However, basic research and clinical practice in the lung regeneration remains crawling. Owing to the complicated three dimensional structures and above 40 types of pulmonary cells, the regeneration of lung tissues becomes a great challenge. Compelling evidence has showed that distinct populations of intrapulmonary and extrapulmonary stem/progenitor cells can regenerate epithelia as well as endothelia in various parts of the respiratory tract. Recently, the discovery of human lung stem cells and their relevant studies has opened the door of hope again, which might put us on the path to repair our injured body parts, lungs on demand. Herein, we emphasized the role of endogenous and exogenous stem/progenitor cells in lungs as well as artificial tissue repair for the injured lungs, which constitute a marvelous toolbox for the treatment of acute lung injury. Finally, we further discussed the potential problems in the pulmonary remodeling and regeneration.
Collapse
Affiliation(s)
- Ce Yang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Changjiang Zhilu, Daping, 400042, Chongqing, China.
| | - Jianxin Jiang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Changjiang Zhilu, Daping, 400042, Chongqing, China.
| | - Xuetao Yang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Changjiang Zhilu, Daping, 400042, Chongqing, China.
| | - Haiyan Wang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Changjiang Zhilu, Daping, 400042, Chongqing, China.
| | - Juan Du
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Changjiang Zhilu, Daping, 400042, Chongqing, China.
| |
Collapse
|
18
|
Abstract
The understanding of bone marrow stem cell plasticity and contribution of bone marrow stem cells to pathophysiology is evolving with the advent of innovative technologies. Recent data has led to new mechanistic insights in the field of mesenchymal stem cell (MSC) research, and an increased appreciation for the plasticity of the hematopoietic stem cell (HSC). In this review, we discuss current research examining the origin of pulmonary cell types from endogenous lung stem and progenitor cells as well as bone marrow-derived stem cells (MSCs and HSCs) and their contributions to lung homeostasis and pathology. We specifically highlight recent findings from our laboratory that demonstrate an HSC origin for pulmonary fibroblasts based on transplantation of a clonal population of cells derived from a single HSC. These findings demonstrate the importance of developing an understanding of the sources of effector cells in disease state. Finally, a perspective is given on the potential clinical implications of these studies and others addressing stem cell contributions to lung tissue homeostasis and pathology.
Collapse
Affiliation(s)
- Lindsay T McDonald
- Research Services, Ralph H Johnson VAMC, Charleston, SC, USA; Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Amanda C LaRue
- Research Services, Ralph H Johnson VAMC, Charleston, SC, USA; Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, USA; Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
19
|
Yao W, Liu S, Li J, Hao C. Silica promotes the transdifferentiation of rat circulating fibrocytes in vitro. Mol Med Rep 2015; 12:5828-36. [PMID: 26299717 PMCID: PMC4581811 DOI: 10.3892/mmr.2015.4212] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 07/21/2015] [Indexed: 01/18/2023] Open
Abstract
To investigate the effects of silica on circulating fibrocytes (cFbs), the present study established a primary culture model of rat alveolar macrophages and cFbs in vitro. Macrophages were treated with free silica, and their supernatant was used to stimulate cFbs. The mRNA expression levels of collagen I, collagen III and α-smooth muscle actin (SMA) in cFbs were analyzed by reverse transcription-quantitative polymerase chain reaction. The intracellular and extracellular protein expression levels of collagen I, collagen III and α-SMA were detected by ELISA and immunofluorescence staining. The results indicated that in the cell model, the free silica effectively increased the protein and mRNA expression levels of collagen-I, collagen-III and α-SMA. The free silica significantly promoted the transdifferentiation of cFbs into myofibroblasts in a dose-and time-dependent manner.
Collapse
Affiliation(s)
- Wu Yao
- Department of Public Health, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Suna Liu
- Department of Public Health, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Ju Li
- Department of Public Health, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Changfu Hao
- Department of Public Health, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| |
Collapse
|
20
|
Leite CF, Lopes CS, Alves AC, Fuzaro CSC, Silva MV, Oliveira LFD, Garcia LP, Farnesi TS, Cuba MBD, Rocha LB, Rodrigues V, Oliveira CJFD, Dias da Silva VJ. Endogenous resident c-Kit cardiac stem cells increase in mice with an exercise-induced, physiologically hypertrophied heart. Stem Cell Res 2015; 15:151-64. [PMID: 26070113 DOI: 10.1016/j.scr.2015.05.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Revised: 05/13/2015] [Accepted: 05/20/2015] [Indexed: 02/08/2023] Open
Abstract
Physical activity evokes well-known adaptations in the cardiovascular system. Although exercise training induces cardiac remodeling, whether multipotent stem cells play a functional role in the hypertrophic process remains unknown. To evaluate this possibility, C57BL/6 mice were subjected to swimming training aimed at achieving cardiac hypertrophy, which was morphologically and electrocardiographically characterized. Subsequently, c-Kit(+)Lin(-) and Sca-1(+)Lin(-) cardiac stem cells (CSCs) were quantified using flow cytometry while cardiac muscle-derived stromal cells (CMSCs, also known as cardiac-derived mesenchymal stem cells) were assessed using in vitro colony-forming unit fibroblast assay (CFU-F). Only the number of c-Kit(+)Lin(-) cells increased in the hypertrophied heart. To investigate a possible extracardiac origin of these cells, a parabiotic eGFP transgenic/wild-type mouse model was used. The parabiotic pairs were subjected to swimming, and the wild-type heart in particular was tested for eGFP(+) stem cells. The results revealed a negligible number of extracardiac stem cells in the heart, allowing us to infer a cardiac origin for the increased amount of detected c-Kit(+) cells. In conclusion, the number of resident Sca-1(+)Lin(-) cells and CMSCs was not changed, whereas the number of c-Kit(+)Lin(-) cells was increased during physiological cardiac hypertrophy. These c-Kit(+)Lin(-) CSCs may contribute to the physiological cardiac remodeling that result from exercise training.
Collapse
Affiliation(s)
- Camila Ferreira Leite
- Department of Biochemistry, Pharmacology, Physiology and Molecular Biology, Triângulo Mineiro Federal University, Praça Manoel Terra, 330, Centro, 38025-015 Uberaba, MG, Brazil
| | - Carolina Salomão Lopes
- Department of Biochemistry, Pharmacology, Physiology and Molecular Biology, Triângulo Mineiro Federal University, Praça Manoel Terra, 330, Centro, 38025-015 Uberaba, MG, Brazil
| | - Angélica Cristina Alves
- Department of Biochemistry, Pharmacology, Physiology and Molecular Biology, Triângulo Mineiro Federal University, Praça Manoel Terra, 330, Centro, 38025-015 Uberaba, MG, Brazil
| | - Caroline Santos Capitelli Fuzaro
- Department of Biochemistry, Pharmacology, Physiology and Molecular Biology, Triângulo Mineiro Federal University, Praça Manoel Terra, 330, Centro, 38025-015 Uberaba, MG, Brazil
| | - Marcos Vinícius Silva
- Department of Microbiology, Immunology and Parasitology, Triângulo Mineiro Federal University, Praça Manoel Terra, 330, Centro, 38025-015 Uberaba, MG, Brazil
| | - Lucas Felipe de Oliveira
- Department of Biochemistry, Pharmacology, Physiology and Molecular Biology, Triângulo Mineiro Federal University, Praça Manoel Terra, 330, Centro, 38025-015 Uberaba, MG, Brazil
| | - Lidiane Pereira Garcia
- Department of Biochemistry, Pharmacology, Physiology and Molecular Biology, Triângulo Mineiro Federal University, Praça Manoel Terra, 330, Centro, 38025-015 Uberaba, MG, Brazil
| | - Thaís Soares Farnesi
- Department of Biochemistry, Pharmacology, Physiology and Molecular Biology, Triângulo Mineiro Federal University, Praça Manoel Terra, 330, Centro, 38025-015 Uberaba, MG, Brazil
| | - Marília Beatriz de Cuba
- Department of Biochemistry, Pharmacology, Physiology and Molecular Biology, Triângulo Mineiro Federal University, Praça Manoel Terra, 330, Centro, 38025-015 Uberaba, MG, Brazil
| | - Lenaldo Branco Rocha
- Department of Morphology, Institute for Biological and Natural Sciences, Triângulo Mineiro Federal University, Praça Manoel Terra, 330, Centro, 38025-015 Uberaba, MG, Brazil
| | - Virmondes Rodrigues
- Department of Microbiology, Immunology and Parasitology, Triângulo Mineiro Federal University, Praça Manoel Terra, 330, Centro, 38025-015 Uberaba, MG, Brazil
| | - Carlo José Freire de Oliveira
- Department of Microbiology, Immunology and Parasitology, Triângulo Mineiro Federal University, Praça Manoel Terra, 330, Centro, 38025-015 Uberaba, MG, Brazil
| | - Valdo José Dias da Silva
- Department of Biochemistry, Pharmacology, Physiology and Molecular Biology, Triângulo Mineiro Federal University, Praça Manoel Terra, 330, Centro, 38025-015 Uberaba, MG, Brazil.
| |
Collapse
|
21
|
Paun A, Kunwar A, Haston CK. Acute adaptive immune response correlates with late radiation-induced pulmonary fibrosis in mice. Radiat Oncol 2015; 10:45. [PMID: 25889053 PMCID: PMC4342202 DOI: 10.1186/s13014-015-0359-y] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Accepted: 02/16/2015] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND The lung response to radiation exposure can involve an immediate or early reaction to the radiation challenge, including cell death and an initial immune reaction, and can be followed by a tissue injury response, of pneumonitis or fibrosis, to this acute reaction. Herein, we aimed to determine whether markers of the initial immune response, measured within days of radiation exposure, are correlated with the lung tissue injury responses occurring weeks later. METHODS Inbred strains of mice known to be susceptible (KK/HIJ, C57BL/6J, 129S1/SvImJ) or resistant (C3H/HeJ, A/J, AKR/J) to radiation-induced pulmonary fibrosis and to vary in time to onset of respiratory distress post thoracic irradiation (from 10-23 weeks) were studied. Mice were untreated (controls) or received 18 Gy whole thorax irradiation and were euthanized at 6 h, 1d or 7 d after radiation treatment. Pulmonary CD4+ lymphocytes, bronchoalveolar cell profile & cytokine level, and serum cytokine levels were assayed. RESULTS Thoracic irradiation and inbred strain background significantly affected the numbers of CD4+ cells in the lungs and the bronchoalveolar lavage cell differential of exposed mice. At the 7 day timepoint greater numbers of pulmonary Th1 and Th17 lymphocytes and reduced lavage interleukin17 and interferonγ levels were significant predictors of late stage fibrosis. Lavage levels of interleukin-10, measured at the 7 day timepoint, were inversely correlated with fibrosis score (R=-0.80, p=0.05), while serum levels of interleukin-17 in control mice significantly correlated with post irradiation survival time (R=0.81, p=0.04). Lavage macrophage, lymphocyte or neutrophil counts were not significantly correlated with either of fibrosis score or time to respiratory distress in the six mouse strains. CONCLUSION Specific cytokine and lymphocyte levels, but not strain dependent lavage cell profiles, were predictive of later radiation-induced lung injury in this panel of inbred strains.
Collapse
Affiliation(s)
- Alexandra Paun
- Department of Human Genetics, Meakins-Christie Laboratories, McGill University, Montreal, QC, Canada.
| | - Amit Kunwar
- Department of Human Genetics, Meakins-Christie Laboratories, McGill University, Montreal, QC, Canada.
| | - Christina K Haston
- Department of Human Genetics, Meakins-Christie Laboratories, McGill University, Montreal, QC, Canada. .,Department of Medicine, Meakins-Christie Laboratories, McGill University, 3626 St. Urbain, H2X 2P2, Montreal, QC, Canada.
| |
Collapse
|
22
|
Wu JMF, Hsueh YC, Ch'ang HJ, Luo CY, Wu LW, Nakauchi H, Hsieh PCH. Circulating cells contribute to cardiomyocyte regeneration after injury. Circ Res 2015; 116:633-41. [PMID: 25398235 DOI: 10.1161/circresaha.116.304564] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
RATIONALE The contribution of bone marrow-borne hematopoietic cells to the ischemic myocardium has been documented. However, a pivotal study reported no evidence of myocardial regeneration from hematopoietic-derived cells. The study did not take into account the possible effect of early injury-induced signaling as the test mice were parabiotically paired to partners immediately after surgery-induced myocardial injury when cross-circulation has not yet developed. OBJECTIVE To re-evaluate the role of circulating cells in the injured myocardium. METHODS AND RESULTS By combining pulse-chase labeling and parabiosis model, we show that circulating cells derived from the parabiont expressed cardiac-specific markers in the injured myocardium. Genetic fate mapping also revealed that circulating hematopoietic cells acquired cardiac cell fate by means of cell fusion and transdifferentiation. CONCLUSIONS These results suggest that circulating cells participate in cardiomyocyte regeneration in a mouse model of parabiosis when the circulatory system is fully developed before surgery-induced heart injury.
Collapse
Affiliation(s)
- Jasmine M F Wu
- From the Institute of Basic Medical Sciences (J.M.F.W., Y.-C.H.) and Institute of Molecular Medicine (L.-W.W.), College of Medicine, National Cheng Kung University, Tainan, Taiwan; Department of Surgery (C.-Y.L.) and Department of Radiation Oncology (H.-J.C.), National Cheng Kung University Hospital, Tainan, Taiwan; National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan (H.-J.C.); Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Tokyo, Japan (H.N.); and Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan (P.C. H.H.)
| | - Ying-Chang Hsueh
- From the Institute of Basic Medical Sciences (J.M.F.W., Y.-C.H.) and Institute of Molecular Medicine (L.-W.W.), College of Medicine, National Cheng Kung University, Tainan, Taiwan; Department of Surgery (C.-Y.L.) and Department of Radiation Oncology (H.-J.C.), National Cheng Kung University Hospital, Tainan, Taiwan; National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan (H.-J.C.); Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Tokyo, Japan (H.N.); and Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan (P.C. H.H.)
| | - Hui-Ju Ch'ang
- From the Institute of Basic Medical Sciences (J.M.F.W., Y.-C.H.) and Institute of Molecular Medicine (L.-W.W.), College of Medicine, National Cheng Kung University, Tainan, Taiwan; Department of Surgery (C.-Y.L.) and Department of Radiation Oncology (H.-J.C.), National Cheng Kung University Hospital, Tainan, Taiwan; National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan (H.-J.C.); Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Tokyo, Japan (H.N.); and Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan (P.C. H.H.)
| | - Chwan-Yau Luo
- From the Institute of Basic Medical Sciences (J.M.F.W., Y.-C.H.) and Institute of Molecular Medicine (L.-W.W.), College of Medicine, National Cheng Kung University, Tainan, Taiwan; Department of Surgery (C.-Y.L.) and Department of Radiation Oncology (H.-J.C.), National Cheng Kung University Hospital, Tainan, Taiwan; National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan (H.-J.C.); Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Tokyo, Japan (H.N.); and Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan (P.C. H.H.)
| | - Li-Wha Wu
- From the Institute of Basic Medical Sciences (J.M.F.W., Y.-C.H.) and Institute of Molecular Medicine (L.-W.W.), College of Medicine, National Cheng Kung University, Tainan, Taiwan; Department of Surgery (C.-Y.L.) and Department of Radiation Oncology (H.-J.C.), National Cheng Kung University Hospital, Tainan, Taiwan; National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan (H.-J.C.); Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Tokyo, Japan (H.N.); and Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan (P.C. H.H.)
| | - Hiromitsu Nakauchi
- From the Institute of Basic Medical Sciences (J.M.F.W., Y.-C.H.) and Institute of Molecular Medicine (L.-W.W.), College of Medicine, National Cheng Kung University, Tainan, Taiwan; Department of Surgery (C.-Y.L.) and Department of Radiation Oncology (H.-J.C.), National Cheng Kung University Hospital, Tainan, Taiwan; National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan (H.-J.C.); Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Tokyo, Japan (H.N.); and Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan (P.C. H.H.)
| | - Patrick C H Hsieh
- From the Institute of Basic Medical Sciences (J.M.F.W., Y.-C.H.) and Institute of Molecular Medicine (L.-W.W.), College of Medicine, National Cheng Kung University, Tainan, Taiwan; Department of Surgery (C.-Y.L.) and Department of Radiation Oncology (H.-J.C.), National Cheng Kung University Hospital, Tainan, Taiwan; National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan (H.-J.C.); Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Tokyo, Japan (H.N.); and Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan (P.C. H.H.).
| |
Collapse
|
23
|
Torres M, Rojas M, Campillo N, Cardenes N, Montserrat JM, Navajas D, Farré R. Parabiotic model for differentiating local and systemic effects of continuous and intermittent hypoxia. J Appl Physiol (1985) 2014; 118:42-7. [PMID: 25377885 DOI: 10.1152/japplphysiol.00858.2014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Hypoxia can be damaging either because cells are directly sensitive to low oxygen pressure in their local microenvironment and/or because they are exposed to circulating factors systemically secreted in response to hypoxia. The conventional hypoxia model, breathing hypoxic air, does not allow one to distinguish between these local and systemic effects. Here we propose and validate a model for differentially applying local and systemic hypoxic challenges in an animal. We used parabiosis, two mice sharing circulation by surgical union through the skin, and tested the hypothesis that when one of the parabionts breathes room air and the other one is subjected to hypoxic air, both mice share systemic circulation but remain normoxic and hypoxic, respectively. We tested two common hypoxic paradigms in 10 parabiotic pairs: continuous hypoxia (10% O2) mimicking chronic lung diseases, and intermittent hypoxia (40 s, 21% O2; 20 s, 5% O2) simulating sleep apnea. Arterial oxygen saturation and oxygen partial pressure at muscle tissue were measured in both parabionts. Effective cross-circulation was assessed by intraperitoneally injecting a dye in one of the parabionts and measuring blood dye concentration in both animals after 2 h. The results confirmed the hypothesis that tissues of the parabiont under room air were perfused with normally oxygenated blood and, at the same time, were exposed to all of the systemic mediators secreted by the other parabiont actually subjected to hypoxia. In conclusion, combination of parabiosis and hypoxic/normoxic air breathing is a novel approach to investigate the effects of local and systemic hypoxia in respiratory diseases.
Collapse
Affiliation(s)
- Marta Torres
- CIBER de Enfermedades Respiratorias, Bunyola, Spain; Sleep Laboratory, Hospital Clinic, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain
| | - Mauricio Rojas
- Dorothy P. & Richard P. Simmons Center for Interstitial Lung Disease, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Noelia Campillo
- CIBER de Enfermedades Respiratorias, Bunyola, Spain; Unitat de Biofísica i Bioenginyeria, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain
| | - Nayra Cardenes
- Dorothy P. & Richard P. Simmons Center for Interstitial Lung Disease, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Josep M Montserrat
- CIBER de Enfermedades Respiratorias, Bunyola, Spain; Sleep Laboratory, Hospital Clinic, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain; Institut Investigacions Biomediques August Pi Sunyer, Barcelona, Spain; and
| | - Daniel Navajas
- CIBER de Enfermedades Respiratorias, Bunyola, Spain; Unitat de Biofísica i Bioenginyeria, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain; Institut de Bioenginyeria de Catalunya, Barcelona, Spain
| | - Ramon Farré
- CIBER de Enfermedades Respiratorias, Bunyola, Spain; Unitat de Biofísica i Bioenginyeria, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain; Institut Investigacions Biomediques August Pi Sunyer, Barcelona, Spain; and
| |
Collapse
|
24
|
Baroke E, Gauldie J, Kolb M. New treatment and markers of prognosis for idiopathic pulmonary fibrosis: lessons learned from translational research. Expert Rev Respir Med 2014; 7:465-78. [PMID: 24138691 DOI: 10.1586/17476348.2013.838015] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic progressive interstitial lung disease with increasing prevalence, high mortality rates and poor treatment options. The diagnostic process is complex and often requires an interdisciplinary approach between different specialists. Information gained over the past 10 years of intense research resulted in improved diagnostic algorithms, a better understanding of the underlying pathogenesis and the development of new therapeutic options. Specifically, the change from the traditional concept that viewed IPF as a chronic inflammatory disorder to the current belief that is primarily resulting from aberrant wound healing enabled the identification of novel treatment targets. This increased the clinical trial activity dramatically and resulted in the approval of the first IPF-specific therapy in many countries. Still, the natural history and intrinsic behavior of IPF are very difficult to predict. There is an urgent need for new therapies and also for development and validation of prognostic markers that predict disease progression, survival and also response to antifibrotic drugs. This review provides an up to date summary of the most relevant clinical trials, novel therapeutic drug targets and outlines a spectrum of potential prognostic biomarkers for IPF.
Collapse
Affiliation(s)
- Eva Baroke
- Department of Medicine, McMaster University, ON, Canada, L8S4L8 and Department of Pathology & Molecular Medicine, McMaster University, Ontario ON, Canada, L8S4L8
| | | | | |
Collapse
|
25
|
Kaddah S, Selim S, Rashed L, Noaman M. Circulating fibrocytes are an indicator of severity and exacerbation in chronic obstructive pulmonary disease. EGYPTIAN JOURNAL OF CHEST DISEASES AND TUBERCULOSIS 2014. [DOI: 10.1016/j.ejcdt.2014.07.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
|
26
|
Mussar K, Tucker A, McLennan L, Gearhart A, Jimenez-Caliani AJ, Cirulli V, Crisa L. Macrophage/epithelium cross-talk regulates cell cycle progression and migration in pancreatic progenitors. PLoS One 2014; 9:e89492. [PMID: 24586821 PMCID: PMC3929706 DOI: 10.1371/journal.pone.0089492] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2013] [Accepted: 01/21/2014] [Indexed: 01/06/2023] Open
Abstract
Macrophages populate the mesenchymal compartment of all organs during embryogenesis and have been shown to support tissue organogenesis and regeneration by regulating remodeling of the extracellular microenvironment. Whether this mesenchymal component can also dictate select developmental decisions in epithelia is unknown. Here, using the embryonic pancreatic epithelium as model system, we show that macrophages drive the epithelium to execute two developmentally important choices, i.e. the exit from cell cycle and the acquisition of a migratory phenotype. We demonstrate that these developmental decisions are effectively imparted by macrophages activated toward an M2 fetal-like functional state, and involve modulation of the adhesion receptor NCAM and an uncommon "paired-less" isoform of the transcription factor PAX6 in the epithelium. Over-expression of this PAX6 variant in pancreatic epithelia controls both cell motility and cell cycle progression in a gene-dosage dependent fashion. Importantly, induction of these phenotypes in embryonic pancreatic transplants by M2 macrophages in vivo is associated with an increased frequency of endocrine-committed cells emerging from ductal progenitor pools. These results identify M2 macrophages as key effectors capable of coordinating epithelial cell cycle withdrawal and cell migration, two events critical to pancreatic progenitors' delamination and progression toward their differentiated fates.
Collapse
Affiliation(s)
- Kristin Mussar
- Department of Medicine, Institute of Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington, United States of America
| | - Andrew Tucker
- Department of Medicine, Institute of Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington, United States of America
| | - Linsey McLennan
- Department of Medicine, Institute of Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington, United States of America
| | - Addie Gearhart
- Department of Medicine, Institute of Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington, United States of America
| | - Antonio J. Jimenez-Caliani
- Department of Medicine, Institute of Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington, United States of America
| | - Vincenzo Cirulli
- Department of Medicine, Institute of Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington, United States of America
| | - Laura Crisa
- Department of Medicine, Institute of Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington, United States of America
| |
Collapse
|
27
|
Hematopoietic and mesenchymal stem cells for the treatment of chronic respiratory diseases: role of plasticity and heterogeneity. ScientificWorldJournal 2014; 2014:859817. [PMID: 24563632 PMCID: PMC3916026 DOI: 10.1155/2014/859817] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Accepted: 10/20/2013] [Indexed: 12/21/2022] Open
Abstract
Chronic lung diseases, such as cystic fibrosis (CF), asthma, and chronic obstructive pulmonary disease (COPD) are incurable and represent a very high social burden. Stem cell-based treatment may represent a hope for the cure of these diseases. In this paper, we revise the overall knowledge about the plasticity and engraftment of exogenous marrow-derived stem cells into the lung, as well as their usefulness in lung repair and therapy of chronic lung diseases. The lung is easily accessible and the pathophysiology of these diseases is characterized by injury, inflammation, and eventually by remodeling of the airways. Bone marrow-derived stem cells, including hematopoietic stem/progenitor cells (HSPCs) and mesenchymal stromal (stem) cells (MSCs), encompass a wide array of cell subsets with different capacities of engraftment and injured tissue regenerating potential. Proof-of-principle that marrow cells administered locally may engraft and give rise to specialized epithelial cells has been given, but the efficiency of this conversion is too limited to give a therapeutic effect. Besides the identification of plasticity mechanisms, the characterization/isolation of the stem cell subpopulations represents a major challenge to improving the efficacy of transplantation protocols used in regenerative medicine for lung diseases.
Collapse
|
28
|
Huang H, Weaver A, Wu E, Li Y, Gao H, Fan W, Wu M. Lipid-based signaling modulates DNA repair response and survival against Klebsiella pneumoniae infection in host cells and in mice. Am J Respir Cell Mol Biol 2013; 49:798-807. [PMID: 23742126 DOI: 10.1165/rcmb.2013-0069oc] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Klebsiella pneumoniae causes serious infections in the urinary tract, respiratory tract, and blood. Lipid rafts, also known as membrane microdomains, have been linked to the pathogenesis of bacterial infection. However, whether lipid rafts affect K. pneumoniae internalization into host cells remains unknown. Here, we show for the first time that K. pneumoniae was internalized into lung cells by activating lipid rafts. Disrupting lipid rafts by methyl-β-cyclodextrin inhibited pathogen internalization, impairing host defense. A deficient mutant of capsule polysaccharide (CPS) showed a higher internalization rate than a wild-type strain, indicating that CPS may inhibit bacterial entry to host cells. Furthermore, lipid rafts may affect the function of extracellular regulated kinase (ERK)-1/2, and knocking down ERK1/2 via short, interfering RNA increased apoptosis in both alveolar macrophages and epithelial cells after infection. To gain insights into bacterial pathogenesis, we evaluated the impact of lipid rafts on DNA integrity, and showed that raft aggregates also affect DNA damage and DNA repair responses (i.e., 8-oxoguanine DNA glycosylase [Ogg1]) through the regulation of reactive oxygen species. Importantly, cells overexpressing Ogg1 demonstrated reduced cytotoxicity during bacterial infection. Taken together, these results suggest that lipid rafts may modulate bacterial internalization, thereby affecting DNA damage and repair, which is critical to host defense against K. pneumoniae.
Collapse
Affiliation(s)
- Huang Huang
- 1 Department of Biochemistry and Molecular Biology, University of North Dakota, Grand Forks, North Dakota
| | | | | | | | | | | | | |
Collapse
|
29
|
Kamran P, Sereti KI, Zhao P, Ali SR, Weissman IL, Ardehali R. Parabiosis in mice: a detailed protocol. J Vis Exp 2013. [PMID: 24145664 DOI: 10.3791/50556] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Parabiosis is a surgical union of two organisms allowing sharing of the blood circulation. Attaching the skin of two animals promotes formation of microvasculature at the site of inflammation. Parabiotic partners share their circulating antigens and thus are free of adverse immune reaction. First described by Paul Bert in 1864(1), the parabiosis surgery was refined by Bunster and Meyer in 1933 to improve animal survival(2). In the current protocol, two mice are surgically joined following a modification of the Bunster and Meyer technique. Animals are connected through the elbow and knee joints followed by attachment of the skin allowing firm support that prevents strain on the sutured skin. Herein, we describe in detail the parabiotic joining of a ubiquitous GFP expressing mouse to a wild type (WT) mouse. Two weeks after the procedure, the pair is separated and GFP positive cells can be detected by flow cytometric analysis in the blood circulation of the WT mouse. The blood chimerism allows one to examine the contribution of the circulating cells from one animal in the other.
Collapse
Affiliation(s)
- Paniz Kamran
- Department of Medicine-Division of Cardiology, University of California, Los Angeles
| | | | | | | | | | | |
Collapse
|
30
|
Amniotic fluid stem cells inhibit the progression of bleomycin-induced pulmonary fibrosis via CCL2 modulation in bronchoalveolar lavage. PLoS One 2013; 8:e71679. [PMID: 23967234 PMCID: PMC3742516 DOI: 10.1371/journal.pone.0071679] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2013] [Accepted: 07/02/2013] [Indexed: 02/06/2023] Open
Abstract
The potential for amniotic fluid stem cell (AFSC) treatment to inhibit the progression of fibrotic lung injury has not been described. We have previously demonstrated that AFSC can attenuate both acute and chronic-fibrotic kidney injury through modification of the cytokine environment. Fibrotic lung injury, such as in Idiopathic Pulmonary Fibrosis (IPF), is mediated through pro-fibrotic and pro-inflammatory cytokine activity. Thus, we hypothesized that AFSC treatment might inhibit the progression of bleomycin-induced pulmonary fibrosis through cytokine modulation. In particular, we aimed to investigate the effect of AFSC treatment on the modulation of the pro-fibrotic cytokine CCL2, which is increased in human IPF patients and is correlated with poor prognoses, advanced disease states and worse fibrotic outcomes. The impacts of intravenous murine AFSC given at acute (day 0) or chronic (day 14) intervention time-points after bleomycin injury were analyzed at either day 3 or day 28 post-injury. Murine AFSC treatment at either day 0 or day 14 post-bleomycin injury significantly inhibited collagen deposition and preserved pulmonary function. CCL2 expression increased in bleomycin-injured bronchoalveolar lavage (BAL), but significantly decreased following AFSC treatment at either day 0 or at day 14. AFSC were observed to localize within fibrotic lesions in the lung, showing preferential targeting of AFSC to the area of fibrosis. We also observed that MMP-2 was transiently increased in BAL following AFSC treatment. Increased MMP-2 activity was further associated with cleavage of CCL2, rendering it a putative antagonist for CCL2/CCR2 signaling, which we surmise is a potential mechanism for CCL2 reduction in BAL following AFSC treatment. Based on this data, we concluded that AFSC have the potential to inhibit the development or progression of fibrosis in a bleomycin injury model during both acute and chronic remodeling events.
Collapse
|
31
|
Mobilization of endothelial progenitor cells from bone marrow is impaired in a piglet model of acute respiratory distress syndrome. Pediatr Crit Care Med 2013; 14:e233-42. [PMID: 23867444 DOI: 10.1097/pcc.0b013e31828a7242] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
OBJECTIVES To characterize the endothelial progenitor cell mobilization in the models of moderate and severe lung injury, we hypothesized that there were differences in endothelial progenitor cell levels and mobilizing cytokines between moderate and severe lung injury. DESIGN Prospective, randomized, and controlled experimental study. SETTING University research laboratory center. SUBJECTS Fifteen healthy piglets. INTERVENTIONS Piglets were randomly allocated to control, moderate lung injury (acute lung injury), and severe lung injury (acute respiratory distress syndrome) groups. Lung injury was established by intravenous infusion of oleic acid. Animals were mechanically ventilated for 24-48 hours, and then animals were weaned from ventilation and cared for until day 7. MEASUREMENTS AND MAIN RESULTS Endothelial progenitor cells were quantified by flow cytometry. After 24 hours, the number of endothelial progenitor cells in peripheral blood increased in the acute lung injury group but was not altered in the acute respiratory distress syndrome group compared to the control group. The number of CD34KDR, KDRCD133, and CD34KDRCD133 cells was higher in the acute lung injury group than in the acute respiratory distress syndrome group. In bone marrow, the number of CD34KDR and KDRCD133 cells was greater in acute respiratory distress syndrome animals but not altered in acute lung injury animals at 24 hours. Furthermore, plasma stromal cell-derived factor-1 and vascular endothelial growth factor concentrations were higher in acute lung injury than in acute respiratory distress syndrome at 24 hours. Matrix metalloproteinase-9 and soluble kit ligand levels in bone marrow were reduced in acute respiratory distress syndrome compared with acute lung injury. Lung CD34, KDR, and lung stromal cell-derived factor-1 messenger RNA expression were higher in the acute lung injury group than in the acute respiratory distress syndrome group. Furthermore, the expression of CD34, KDR, and CD133 messenger RNA in lung tissue was correlated with stromal cell-derived factor-1 in the lung. CONCLUSIONS There was a rapid release of endothelial progenitor cells from bone marrow into circulation in moderate acute lung injury, and endothelial progenitor cell mobilization was impaired in acute respiratory distress syndrome.
Collapse
|
32
|
Conese M, Carbone A, Castellani S, Di Gioia S. Paracrine effects and heterogeneity of marrow-derived stem/progenitor cells: relevance for the treatment of respiratory diseases. Cells Tissues Organs 2013; 197:445-73. [PMID: 23652321 DOI: 10.1159/000348831] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/12/2013] [Indexed: 11/19/2022] Open
Abstract
Stem cell-based treatment may represent a hope for the treatment of acute lung injury and pulmonary fibrosis, and other chronic lung diseases, such as cystic fibrosis, asthma and chronic obstructive pulmonary disease (COPD). It is well established in preclinical models that bone marrow-derived stem and progenitor cells exert beneficial effects on inflammation, immune responses and repairing of damage in virtually all lung-borne diseases. While it was initially thought that the positive outcome was due to a direct engraftment of these cells into the lung as endothelial and epithelial cells, paracrine factors are now considered the main mechanism through which stem and progenitor cells exert their therapeutic effect. This knowledge has led to the clinical use of marrow cells in pulmonary hypertension with endothelial progenitor cells (EPCs) and in COPD with mesenchymal stromal (stem) cells (MSCs). Bone marrow-derived stem cells, including hematopoietic stem/progenitor cells, MSCs, EPCs and fibrocytes, encompass a wide array of cell subsets with different capacities of engraftment and injured tissue-regenerating potential. The characterization/isolation of the stem cell subpopulations represents a major challenge to improve the efficacy of transplantation protocols used in regenerative medicine and applied to lung disorders.
Collapse
Affiliation(s)
- Massimo Conese
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy.
| | | | | | | |
Collapse
|
33
|
Kim EK, Lee JH, Jeong HC, Oh D, Hwang SG, Cho YW, Lee SJ, Oh YM, Lee SD. Impaired colony-forming capacity of circulating endothelial progenitor cells in patients with emphysema. TOHOKU J EXP MED 2013; 227:321-31. [PMID: 22850689 DOI: 10.1620/tjem.227.321] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is classified into emphysema and chronic bronchitis, which are thought to result from different pathophysiological pathways. Smoking-induced lung parenchymal destruction and inadequate repair are involved in the pathogenesis of emphysema. In addition, decreased expression of vascular endothelial growth factor and increased endothelial cell apoptosis in the lung may participate in emphysema pathogenesis. As stem cells, circulating endothelial progenitor cells (EPCs) may play a key role in the maintenance of vascular integrity by replacing and repairing the damaged endothelial cells in the tissues. To determine whether the lack of appropriate repair by circulating EPCs in cases of smoking-induced endothelial cell injury participates in emphysema pathogenesis, we determined the association between the colony-forming or migratory capacity of circulating EPCs and the presence of emphysema in 51 patients with COPD. The patients were divided into emphysema (n = 23) and non-emphysema groups (n = 28) based on high-resolution computed tomography. Twenty-two smokers with normal lung function and 14 normal non-smokers served as controls. Circulating EPCs isolated from patients with emphysema showed significantly lower colony-forming units (CFUs) than those from patients with non-emphysema group, smokers with normal lung function, and normal non-smokers. EPCs from patients with emphysema showed significantly lower migratory capacity than those from normal non-smoking controls (p < 0.05). On multivariate analysis, the EPC-CFU was independently associated with emphysema (OR 0.944, 95% CI = 0.903-0.987, p = 0.011). Thus, impaired functions of circulating EPCs may contribute to the development of emphysema.
Collapse
Affiliation(s)
- Eun-Kyung Kim
- Department of Internal Medicine, CHA Bundang Medical Center, College of Medicine, CHA University, Seongnam, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Lung. Regen Med 2013. [DOI: 10.1007/978-94-007-5690-8_34] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
35
|
Abreu SC, Antunes MA, Maron-Gutierrez T, Cruz FF, Ornellas DS, Silva AL, Diaz BL, Ab'Saber AM, Capelozzi VL, Xisto DG, Morales MM, Rocco PRM. Bone marrow mononuclear cell therapy in experimental allergic asthma: intratracheal versus intravenous administration. Respir Physiol Neurobiol 2012; 185:615-24. [PMID: 23164835 DOI: 10.1016/j.resp.2012.11.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2012] [Revised: 11/08/2012] [Accepted: 11/09/2012] [Indexed: 12/14/2022]
Abstract
We hypothesized that the route of administration would impact the beneficial effects of bone marrow-derived mononuclear cell (BMDMC) therapy on the remodelling process of asthma. C57BL/6 mice were randomly assigned to two main groups. In the OVA group, mice were sensitized and challenged with ovalbumin, while the control group received saline using the same protocol. Twenty-four hours before the first challenge, control and OVA animals were further randomized into three subgroups to receive saline (SAL), BMDMCs intravenously (2×10(6)), or BMDMCs intratracheally (2×10(6)). The following changes were induced by BMDMC therapy in OVA mice regardless of administration route: reduction in resistive and viscoelastic pressures, static elastance, eosinophil infiltration, collagen fibre content in airways and lung parenchyma; and reduction in the levels of interleukin (IL)-4, IL-13, transforming growth factor-β and vascular endothelial growth factor. In conclusion, BMDMC modulated inflammatory and remodelling processes regardless of administration route in this experimental model of allergic asthma.
Collapse
Affiliation(s)
- Soraia C Abreu
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Kolosova IA, Angelini D, Fan C, Skinner J, Cheadle C, Johns RA. Resistin-like molecule α stimulates proliferation of mesenchymal stem cells while maintaining their multipotency. Stem Cells Dev 2012; 22:239-47. [PMID: 22891677 DOI: 10.1089/scd.2012.0192] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Resistin-like molecule α (RELMα) is highly upregulated in the lungs of mice subjected to hypoxia. It is secreted from pulmonary epithelium and causes potent mitogenic, angiogenic, and vasoconstrictive effects in the lung vasculature. By using bone marrow transplantation in mice, we previously showed that RELMα is able to increase the number of bone marrow-derived cells in lung tissue, especially in the remodeling pulmonary vasculature. The current study investigated the effect of RELMα on progenitor stem cell content in mouse lung. Hypoxia, while stimulating RELMα expression, caused an increase in the number of Sca1(+)/CD45(-) progenitor cells in lungs of wild-type mice, but not in lungs of RELMα knockout mice. An in vitro study with cultured mesenchymal stem cells (MSCs) showed that RELMα induced a robust proliferative response that was dependent on Phosphatidylinositol 3-kinase/Akt and Erk activation. RELMα treatment of MSCs caused upregulation of a large number of genes involved in cell cycle, mitosis, organelle, and cytoskeleton biogenesis, and DNA metabolism. MSCs cultured in RELMα-supplemented media were able to maintain their differentiation potential into adipogenic, osteogenic, or mesenchymal phenotypes, although adipogenic differentiation was partially inhibited. These results demonstrate that RELMα may be involved in stem cell proliferation in the lung, without affecting differentiation potential.
Collapse
Affiliation(s)
- Irina A Kolosova
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | | | | | | | | | | |
Collapse
|
37
|
[Mice cope with parabiosis − assessment of their physiological changes of life]. DONG WU XUE YAN JIU = ZOOLOGICAL RESEARCH 2012; 33:493-7. [PMID: 23019031 DOI: 10.3724/sp.j.1141.2012.05493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The purpose of this study was to establish a parabiotic mice model and assess the physiological changes of the mice under the parabiotic state. Thirteen pairs of isogenic partners were studied. The model was created by preparing a bridge of skin and subcutaneous tissues between the two mice starting distal of the elbow joint along the humerus along the lateral costal region until the end of the waist line. Physiological, social and affective qualities of life were studied in the mice through behavioural observations for 120 days following the parabiotic surgery. During the first 2-3 days following the operation, the animals suffered from severe pain and distress. During the following days and weeks, the physiological system began to recover and the animals displayed behavioral adaptations to the parabiotic condition. All animals survived at day 120. At three days post operation, the body weight began to decrease. Following this, the animals experienced a continual body weight recovery and reached pre-surgical measures at about 30 days post op. Forty-eight h post op., faecal corticosterone-metabolites were extremely elevated, but their levels returned to two to four times of levels in control females within 72 hours post op. The faecal corticosterone-metabolite levels decreased near to control values on day 75. Out of the 13 pairs, the blood exchange rate of three parabiotic partners was tested, with the result being normal post op. After 12 weeks, the total blood exchange between both partners needed 63 or 46 or 107 min, respectively. These results demonstrated that the animals could adapt behaviourally to the parabiotic situation. Therefore, this parabiosis mouse model may provide useful insights in many research areas, such as transplantation immunity, hematological system and metabolism, etc.
Collapse
|
38
|
Pishel I, Shytikov D, Orlova T, Peregudov A, Artyuhov I, Butenko G. Accelerated aging versus rejuvenation of the immune system in heterochronic parabiosis. Rejuvenation Res 2012; 15:239-48. [PMID: 22533440 DOI: 10.1089/rej.2012.1331] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The emergence of immune disorders in aging is explained by many factors, including thymus dysfunction, decrease in the proportion and function of naïve T cells, and so forth. There are several approaches to preventing these changes, such as thymus rejuvenation, stem cells recovery, modulation of hormone production, and others. Our investigations of heterochronic parabiosis have shown that benefits of a young immune system, e.g., actively working thymus and regular migration of young hematopoietic stem cells between parabiotic partners, appeared unable to restore the immune system of the old partner. At the same time, we have established a progressive immune impairment in the young heterochronic partners. The mechanism of age changes in the immune system in this model, which may lead to reduced life expectancy, has not been fully understood. The first age-related manifestation in the young partners observed 3 weeks after the surgery was a dramatic increase of CD8(+)44(+) cells population in the spleen. A detailed analysis of further changes revealed a progressive decline of most immunological functions observable for up to 3 months after the surgery. This article reviews possible mechanisms of induction of age-related changes in the immune system of young heterochronic partners. The data obtained suggest the existence of certain factors in the old organisms that trigger aging, thus preventing the rejuvenation process.
Collapse
Affiliation(s)
- Iryna Pishel
- Institute of Gerontology NAMS of Ukraine, Kyiv, Ukraine.
| | | | | | | | | | | |
Collapse
|
39
|
Haston CK. Mouse genetic approaches applied to the normal tissue radiation response. Front Oncol 2012; 2:94. [PMID: 22891164 PMCID: PMC3413016 DOI: 10.3389/fonc.2012.00094] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2012] [Accepted: 07/22/2012] [Indexed: 01/20/2023] Open
Abstract
The varying responses of inbred mouse models to radiation exposure present a unique opportunity to dissect the genetic basis of radiation sensitivity and tissue injury. Such studies are complementary to human association studies as they permit both the analysis of clinical features of disease, and of specific variants associated with its presentation, in a controlled environment. Herein I review how animal models are studied to identify specific genetic variants influencing predisposition to radiation-induced traits. Among these radiation-induced responses are documented strain differences in repair of DNA damage and in extent of tissue injury (in the lung, skin, and intestine) which form the base for genetic investigations. For example, radiation-induced DNA damage is consistently greater in tissues from BALB/cJ mice, than the levels in C57BL/6J mice, suggesting there may be an inherent DNA damage level per strain. Regarding tissue injury, strain specific inflammatory and fibrotic phenotypes have been documented for principally, C57BL/6 C3H and A/J mice but a correlation among responses such that knowledge of the radiation injury in one tissue informs of the response in another is not evident. Strategies to identify genetic differences contributing to a trait based on inbred strain differences, which include linkage analysis and the evaluation of recombinant congenic (RC) strains, are presented, with a focus on the lung response to irradiation which is the only radiation-induced tissue injury mapped to date. Such approaches are needed to reveal genetic differences in susceptibility to radiation injury, and also to provide a context for the effects of specific genetic variation uncovered in anticipated clinical association studies. In summary, mouse models can be studied to uncover heritable variation predisposing to specific radiation responses, and such variations may point to pathways of importance to phenotype development in the clinic.
Collapse
Affiliation(s)
- Christina K Haston
- Meakins-Christie Laboratories and the Department of Medicine, McGill University Montreal, QC, Canada
| |
Collapse
|
40
|
Hannoush EJ, Elhassan I, Sifri ZC, Mohr AA, Alzate WD, Livingston DH. Role of bone marrow and mesenchymal stem cells in healing after traumatic injury. Surgery 2012; 153:44-51. [PMID: 22862904 DOI: 10.1016/j.surg.2012.06.020] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2011] [Accepted: 06/08/2012] [Indexed: 12/26/2022]
Abstract
BACKGROUND The role of bone marrow-derived cells (BMDCs) and mesenchymal stem cells (MSC) in healing of traumatic-induced injury remains poorly understood. Mesenteric lymph duct ligation (LDL) results in decreased BMDC mobilization and impaired healing. We hypothesized that LDL-mediated impaired healing would be abrogated by reinjection of BMDC or MSC. METHODS Sprague-Dawley rats were subjected to LDL + lung contusion (LC+LDL) with or without injection of BMDCs or MSCs. Unmanipulated control (UC) and lung contusion alone (LC) served as controls. BMDC and MSC homing was assessed by hematopoietic progenitor cell (HPC [granulocyte-, erythrocyte-, monocyte-, and megakaryocyte colony-forming units; erythroid burst-forming units; and erythroid colony-forming units]) colony growth and immunofluorescent microscopic tracking of tagged MSC, respectively. Histologic lung injury score (LIS) was used to grade injury. Data are mean ± SD. *P < .05/Student t test. RESULTS Lung HPC growth was decreased in LC+LDL versus LC alone (HPC colonies: 2 ± 2, 4 ± 3, 4 ± 2 vs. 11 ± 2, 20 ± 6, 22 ± 9. *P < .05). LC+LDL had greater degree of lung injury on days 5 and 7 LC alone (LIS: 5 ± 1, 4 ± 1 vs. 3 ± 1, 1 ± 0.4. *P < .05). BMDC injection into rats with LC + LDL increased lung HPC growth to LC level (HPC colonies: 12 ± 2, 19 ± 5, 17 ± 4 vs 11 ± 2, 20 ± 6, 22 ± 9. P > .05). Injected MSCs into LC+LDL rats homed preferentially to contused versus noncontused lung (MSC/high-powered field: 6 ± 4 vs. 2 ± 2 *P < .05). Either BMDC or MSC injection into LC+LDL rats returned lung injury to LC level on day 7 (LIS: 1 ± 0.4 and 1 ± 1 vs. 1 ± 0.4. P > .05). CONCLUSION LDL-mediated impaired tissue healing is abrogated by either whole BMDC or MSC injection. This highlights the critical role of BMDC and MSC on healing of trauma-induced injury.
Collapse
Affiliation(s)
- Edward J Hannoush
- University of Medicine and Dentistry of New Jersey-New Jersey Medical School, 150 Bergen Street, Newark, NJ 07103, USA
| | | | | | | | | | | |
Collapse
|
41
|
In vivo molecular imaging of murine embryonic stem cells delivered to a burn wound surface via Integra® scaffolding. J Burn Care Res 2012; 33:e49-54. [PMID: 22540138 DOI: 10.1097/bcr.0b013e3182331d1c] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
It has been demonstrated that restoration of function to compromised tissue can be accomplished by transplantation of bone marrow stem cells and/or embryonic stem cells (ESCs). One limitation to this approach has been the lack of noninvasive techniques to longitudinally monitor stem cell attachment and proliferation. Recently, murine ESC lines that express green fluorescent protein (GFP), luciferase (LV), and herpes simplex thymidine kinase (HVTK) were developed for detection of actively growing cells in vivo by imaging. In this study, the authors investigated the use of these ESC lines in a burned mouse model using Integra® as a delivery scaffolding/matrix. Two different cell lines were used: one expressing GFP and LV and the other expressing GFP, LV, and HVTK. Burn wounds were produced by application of a brass block (2 × 2 cm kept in boiling water before application) to the dorsal surface of SV129 mice for 10 seconds. Twenty-four hours after injury, Integra® with adherent stem cells was engrafted onto a burn wound immediately after excision of eschar. The stem cells were monitored in vivo by measuring bioluminescence with a charge-coupled device camera and immunocytochemistry of excised tissue. Bioluminescence progressively increased in intensity over the time course of the study, and GFP-positive cells growing into the Integra® were detected. These studies demonstrate the feasibility of using Integra® as a scaffolding, or matrix, for the delivery of stem cells to burn wounds as well as the utility of bioluminescence for monitoring in vivo cellular tracking of stably transfected ESC cells.
Collapse
|
42
|
Abstract
Fibrotic lung injury is often attributed to a myriad of factors, including environmental exposure, age, genetic predisposition, epigenetics, coexisting conditions, acute lung injury, and viral infection. No effective therapies, other than lung transplantation, have proven effective against lung fibrosis. Loss of cellular homeostasis mechanisms in alveolar epithelial type I cells and any inability of type II progenitor cells to resist and repair epithelial injury are indicators that impaired response to injury and regeneration is a critical component of this disorder. The alveolar epithelium has a limited repertoire of responses to injury, which are dictated by the alveolar milieu, a repository of cytokines and growth factors that affect recruitment of other cells to the site of injury, or the proliferation of resident cells at the site of injury. The identification and characterization of the cytokines, growth factors, and other biomarkers that dictate the response to disease is key to understanding, diagnosing, treating, and determining the trajectory of various lung disorders. Corrective therapy of the alveolar milieu may therefore prove to be beneficial in many presently serious and incurable lung diseases that likely begin and progress with injury to the alveolar epithelium.
Collapse
|
43
|
Mesenchymal stem cell therapy and lung diseases. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2012; 130:105-29. [PMID: 22772131 DOI: 10.1007/10_2012_140] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Mesenchymal stem cells (MSCs), a distinct population of adult stem cells, have amassed significant interest from both medical and scientific communities. An inherent multipotent differentiation potential offers a cell therapy option for various diseases, including those of the musculoskeletal, neuronal, cardiovascular and pulmonary systems. MSCs also secrete an array of paracrine factors implicated in the mitigation of pathological conditions through anti-inflammatory, anti-apoptotic and immunomodulatory mechanisms. The safety and efficacy of MSCs in human application have been confirmed through small- and large-scale clinical trials. However, achieving the optimal clinical benefit from MSC-mediated regenerative therapy approaches is entirely dependent upon adequate understanding of their healing/regeneration mechanisms and selection of appropriate clinical conditions. MSC-mediated acute alveolar injury repair. A cartoon depiction of an injured alveolus with associated inflammation and AEC apoptosis. Proposed routes of MSC delivery into injured alveoli could be by either intratracheal or intravenous routes, for instance. Following delivery a proposed mechanism of MSC action is to inhibit/reduce alveolar inflammation by abrogation of IL-1_-depenedent Tlymphocyte proliferation and suppression of TNF-_ secretion via macrophage activation following on from stimulation by MSC-secreted IL-1 receptor antagonist (IL-1RN). The inflammatory environment also stimulates MSC to secrete prostaglandin-E2 (PGE2) which can stimulate activated macrophages to secrete the anti-inflammatory cytokine IL-10. Inhibition of AEC apoptosis following injury can also be promoted via MSC stimulated up-regulation of the anti-apoptotic Bcl-2 gene. MSC-secreted KGF can stimulate AECII proliferation and migration propagating alveolar epithelial restitution. Alveolar structural engraftment of MSC is a rare event.
Collapse
|
44
|
Smoking and idiopathic pulmonary fibrosis. Pulm Med 2012; 2012:808260. [PMID: 22448328 PMCID: PMC3289849 DOI: 10.1155/2012/808260] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2011] [Revised: 08/09/2011] [Accepted: 12/07/2011] [Indexed: 12/27/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a disease of unknown etiology with considerable morbidity and mortality. Cigarette smoking is one of the most recognized risk factors for development of IPF. Furthermore, recent work suggests that smoking may have a detrimental effect on survival of patients with IPF. The mechanism by which smoking may contribute to the pathogenesis of IPF is largely unknown. However, accumulating evidence suggests that increased oxidative stress might promote disease progression in IPF patients who are current and former smokers. In this review, potential mechanisms by which cigarette smoking affects IPF, the effects of cigarette smoking on accelerated loss of lung function in patients with IPF, key genetic studies evaluating the potential candidate genes and gene-environment (smoking) interaction, diagnosis, and treatment with emphasis on recently closed and ongoing clinical trials are presented.
Collapse
|
45
|
Gibney B, Chamoto K, Lee GS, Simpson DC, Miele L, Tsuda A, Konerding MA, Wagers A, Mentzer SJ. Cross-circulation and cell distribution kinetics in parabiotic mice. J Cell Physiol 2012; 227:821-8. [PMID: 21503883 PMCID: PMC3160515 DOI: 10.1002/jcp.22796] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Blood-borne nucleated cells participate not only in inflammation, but in tissue repair and regeneration. Because progenitor and stem cell populations have a low concentration in the blood, the circulation kinetics and tissue distribution of these cells is largely unknown. An important approach to tracking cell lineage is the use of fluorescent tracers and parabiotic models of cross-circulation. Here, we investigated the cross-circulation and cell distribution kinetics of C57/B6 GFP(+)/wild-type parabionts. Flow cytometry analysis of the peripheral blood after parabiosis demonstrated no evidence for a "parabiotic barrier" based on cell size or surface characterstics; all peripheral blood cell subpopulations in this study reached equilibrium within 14 days. Whole blood fluorescence analysis indicated that the mean exchange flow rate was 16 µl/h or 0.66% of the circulating blood volume per hour. Studies of peripheral lymphoid organs indicated differential cell distribution kinetics. Some subpopulations, such as CD8(+) and CD11c(+), equilibrated in both lymph nodes and spleen indicating a residence time <28 days; in contrast, other lymphocyte subpopulations, such as B220(+) and CD4(+) cells, had not yet reached equilibrium at 28 days. We conclude that parabiosis can provide important insights into defining tissue distribution, residence times, and recirculating pools using fluorochrome markers of cell lineage.
Collapse
Affiliation(s)
- Barry Gibney
- Laboratory of Adaptive and Regenerative Biology, Brigham & Women’s Hospital, Harvard Medical School, Boston MA
| | - Kenji Chamoto
- Laboratory of Adaptive and Regenerative Biology, Brigham & Women’s Hospital, Harvard Medical School, Boston MA
| | - Grace S. Lee
- Laboratory of Adaptive and Regenerative Biology, Brigham & Women’s Hospital, Harvard Medical School, Boston MA
| | - Dinee C. Simpson
- Laboratory of Adaptive and Regenerative Biology, Brigham & Women’s Hospital, Harvard Medical School, Boston MA
| | - Lino Miele
- Laboratory of Adaptive and Regenerative Biology, Brigham & Women’s Hospital, Harvard Medical School, Boston MA
| | - Akira Tsuda
- Molecular and Integrative Physiological Sciences, Harvard School of Public Health, Boston, MA
| | | | - Amy Wagers
- Department of Pathology, Joslin Diabetes Center, Boston, MA
| | - Steven J. Mentzer
- Laboratory of Adaptive and Regenerative Biology, Brigham & Women’s Hospital, Harvard Medical School, Boston MA
| |
Collapse
|
46
|
Ekert JE, Murray LA, Das AM, Sheng H, Giles-Komar J, Rycyzyn MA. Chemokine (C-C motif) ligand 2 mediates direct and indirect fibrotic responses in human and murine cultured fibrocytes. FIBROGENESIS & TISSUE REPAIR 2011; 4:23. [PMID: 22011363 PMCID: PMC3206835 DOI: 10.1186/1755-1536-4-23] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2011] [Accepted: 10/19/2011] [Indexed: 12/02/2022]
Abstract
Background Fibrocytes are a population of circulating bone-marrow-derived cells that express surface markers for leukocytes and mesenchymal cells, and are capable of differentiating into myofibroblasts. They have been observed at sites of active fibrosis and increased circulating numbers correlate with mortality in idiopathic pulmonary fibrosis (IPF). Inhibition of chemokine (C-C motif) receptor 2 (CCR2) during experimental models of lung fibrosis reduces lung collagen deposition, as well as reducing lung fibrocyte accumulation. The aim of the present study was to determine whether human and mouse fibrocytes express functional CCR2. Results Following optimized and identical human and murine fibrocyte isolation, both cell sources were shown to be positive for CCR2 by flow cytometry and this expression colocalized with collagen I and CD45. Human blood fibrocytes stimulated with the CCR2 ligand chemokine (C-C motif) ligand 2 (CCL2), demonstrated increased proliferation (P < 0.005) and differentiation into myofibroblasts (P < 0.001), as well as a chemotactic response (P < 0.05). Murine fibrocytes also responded to CCR2 stimulation, with CCL12 being more potent than CCL2. Conclusions This study directly compares the functional responses of human and murine fibrocytes to CCR2 ligands, and following comparable isolation techniques. We have shown comparable biological effects, strengthening the translatability of the murine models to human disease with respect to targeting the CCR2 axis to ameliorate disease in IPF patients.
Collapse
Affiliation(s)
- Jason E Ekert
- Departments of Cell Biology and Assay Technologies, Centocor R&D, a division of Johnson & Johnson Pharmaceutical Research & Development, LLC, Radnor, PA, USA
| | - Lynne A Murray
- Department of Immunology, Centocor R&D, a division of Johnson & Johnson Pharmaceutical Research & Development, LLC, Radnor, PA, USA
| | - Anuk M Das
- Department of Immunology, Centocor R&D, a division of Johnson & Johnson Pharmaceutical Research & Development, LLC, Radnor, PA, USA
| | - Hai Sheng
- Departments of Cell Biology and Assay Technologies, Centocor R&D, a division of Johnson & Johnson Pharmaceutical Research & Development, LLC, Radnor, PA, USA
| | - Jill Giles-Komar
- Departments of Cell Biology and Assay Technologies, Centocor R&D, a division of Johnson & Johnson Pharmaceutical Research & Development, LLC, Radnor, PA, USA
| | - Michael A Rycyzyn
- Departments of Cell Biology and Assay Technologies, Centocor R&D, a division of Johnson & Johnson Pharmaceutical Research & Development, LLC, Radnor, PA, USA
| |
Collapse
|
47
|
Murray LA. Commonalities between the pro-fibrotic mechanisms in COPD and IPF. Pulm Pharmacol Ther 2011; 25:276-80. [PMID: 21983244 DOI: 10.1016/j.pupt.2011.08.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2011] [Revised: 08/03/2011] [Accepted: 08/25/2011] [Indexed: 01/06/2023]
Abstract
COPD and IPF are two chronic lung diseases which are characterized by a decline in lung function, resulting in significant morbidity and mortality. Both of these diseases are more commonly associated with an aging population and the duration for which the disease has been underlying is often unknown. Significant matrix deposition occurs, resulting in either non-reversible airways obstruction in the case of COPD and impaired gas exchange and parenchymal consolidation in IPF. There are no approved therapies that have been demonstrated to target these underlying fibrotic changes in the lung. This may in part be due to the challenges of quantitating lung fibrosis in a temporal manner in specific regions of the lung. However, this may also be due to our understanding of aberrant and pathogenic collagen deposition being somewhat limited. The core processes associated with lung fibrosis are often observed in normal wound healing. Moreover, in the extreme fibrotic setting of IPF, the remodelling is sometimes associated with uncontrolled wound healing responses. As wound healing is a critical aspect to maintaining tissue function and homeostasis, targeting this process directly may result in safety concerns. This review therefore describes some of the recent advances in ascertaining pathways promoting lung fibrosis that may be amenable to therapeutic intervention in both COPD and IPF.
Collapse
Affiliation(s)
- Lynne A Murray
- Respiratory, Inflammation and Autoimmunity, MedImmune Ltd, Granta Park, Cambridge CB21 6GH, UK.
| |
Collapse
|
48
|
Yahaya B, Baker A, Tennant P, Smith SH, Shaw DJ, McLachlan G, Collie DDS. Analysis of airway epithelial regeneration and repair following endobronchial brush biopsy in sheep. Exp Lung Res 2011; 37:519-35. [PMID: 21895444 DOI: 10.3109/01902148.2011.605513] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Understanding the fundamental processes involved in repairing the airway wall following injury is fundamental to understanding the way in which these processes are perturbed during disease pathology. Indeed complex diseases such as asthma and chronic obstructive pulmonary disease (COPD) have at their core evidence of airway wall remodeling processes that play a crucial functional role in these diseases. The authors sought to understand the dynamic cellular events that occur during bronchial airway epithelial repair in sheep. The injury was induced by endobronchial brush biopsy (BBr), a process that causes epithelial débridement and induces a consequential repair process. In addition, the current experimental protocol allowed for the time-dependent changes in airway wall morphology to be studied both within and between animals. The initial débridement was followed by evidence of dedifferentiation in the intact epithelium at the wound margins, followed by proliferation of cells both within the epithelium and in the deeper wall structures, notably in association with the submucosal glands and smooth muscle bundles. Seven days after injury, although the airway wall was thickened at the site of damage, the epithelial layer was intact, with evidence of redifferentiation. These studies, in demonstrating broad agreement with previous studies in small animals, indicate the wider relevance of this system as a comparative model and should provide a solid basis upon which to further characterize the critical cellular and molecular interactions that underlie both effective restitution and pathological repair.
Collapse
Affiliation(s)
- B Yahaya
- Human Genome Centre, School of Medical Sciences, Health Campus, Universiti Sains Malaysia, Kubang Kerian Kelantan, Malaysia.
| | | | | | | | | | | | | |
Collapse
|
49
|
|
50
|
Zhang F, Zhang L, Jiang HS, Chen XY, Zhang Y, Li HP, Zhang RX, Zheng H, Chu JX, Chen XJ. Mobilization of bone marrow cells by CSF3 protects mice from bleomycin-induced lung injury. ACTA ACUST UNITED AC 2011; 82:358-68. [PMID: 21778693 DOI: 10.1159/000328762] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2010] [Accepted: 04/26/2011] [Indexed: 01/09/2023]
Abstract
BACKGROUND Bone marrow-derived cells may play a role in tissue injury and repair. Growth factors facilitate the mobilization of bone marrow-derived cells to the site of injury. OBJECTIVES The aim of this study was to determine the effect of the mobilization of autologous bone marrow-derived cells by granulocyte colony-stimulating factor (CSF3) on bleomycin-induced lung injury in mice. METHODS The bone marrow from male green fluorescent protein transgenic (C57Bl/6J) mice was transplanted into irradiated female C57Bl/6J mice. Bleomycin lung injury was induced in these bone marrow-reconstituted mice and unreconstituted C57Bl/6J mice, and some mice were treated with recombinant CSF3. Lung histology, survival, cytokine expression and matrix metalloproteinase (MMP) expression were evaluated to determine the effect of CSF3 after bleomycin-induced lung injury. RESULTS Histology and flow cytometry analysis showed successful mobilization of bone marrow-derived cells by CSF3 treatment in the recipient lungs. Importantly, CSF3 attenuated bleomycin-induced lung injury and improved survival. Furthermore, CSF3 administration regulated transforming growth factor-β, interferon-γ, MMP9 and tissue inhibitors of MMP1 expression during bleomycin injury. CONCLUSIONS These data demonstrated that the mobilization of bone marrow-derived cells by CSF3 has a protective effect against bleomycin-induced lung injury and fibrosis.
Collapse
Affiliation(s)
- Fen Zhang
- The First Hospital, Suzhou University, Suzhou, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|