1
|
Silva CM, Ornellas DS, Ornellas FM, Santos RS, Martini SV, Ferreira D, Muiler C, Cruz FF, Takiya CM, Rocco PRM, Morales MM, Silva PL. Early effects of bone marrow-derived mononuclear cells on lung and kidney in experimental sepsis. Respir Physiol Neurobiol 2023; 309:103999. [PMID: 36460253 DOI: 10.1016/j.resp.2022.103999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 11/11/2022] [Accepted: 11/20/2022] [Indexed: 12/03/2022]
Abstract
BACKGROUND In experimental sepsis, functional and morphological effects of bone marrow-derived mononuclear cell (BMDMC) administration in lung tissue have been evaluated 1 and 7 days after therapy. However, to date no study has evaluated the early effects of BMDMCs in both lung and kidney in experimental polymicrobial sepsis. MATERIAL AND METHODS Twenty-five female C57BL/6 mice were randomly divided into the following groups: 1) cecal ligation and puncture (CLP)-induced sepsis; and 2) Sham (surgical procedure without CLP). After 1 h, CLP animals received saline (NaCl 0.9%) (CLP-Saline) or 106 BMDMCs (CLP-Cell) via the jugular vein. At 6, 12, and 24 h after saline or BMDMC administration, lungs and kidneys were removed for histology and molecular biology analysis. RESULTS In lungs, CLP-Saline, compared to Sham, was associated with increased lung injury score (LIS) and keratinocyte chemoattractant (KC) mRNA expression at 6, 12, and 24 h. BMDMCs were associated with reduced LIS and KC mRNA expression regardless of the time point of analysis. Interleukin (IL)- 10 mRNA content was higher in CLP-Cell than CLP-Saline at 6 and 24 h. In kidney tissue, CLP-Saline, compared to Sham, was associated with tubular cell injury and increased neutrophil gelatinase-associated lipocalin (NGAL) levels, which were reduced after BMDMC therapy at all time points. Surface high-mobility-group-box (HMGB)- 1 levels were higher in CLP-Saline than Sham at 6, 12, and 24 h, whereas nuclear HMGB-1 levels were increased only at 24 h. BMDMCs were associated with decreased surface HMGB-1 and increased nuclear HMGB-1 levels. Kidney injury molecule (KIM)- 1 and IL-18 gene expressions were reduced in CLP-Cell compared to CLP-Saline at 12 and 24 h. CONCLUSION In the present experimental polymicrobial sepsis, early intravenous therapy with BMDMCs was able to reduce lung and kidney damage in a time-dependent manner. BMDMCs thus represent a potential therapy in well-known scenarios of sepsis induction. PURPOSE To evaluate early bone marrow-derived mononuclear cell (BMDMC) therapy on lung and kidney in experimental polymicrobial sepsis. METHODS Twenty-five female C57BL/6 mice were randomly divided into the following groups: cecal ligation and puncture (CLP)-induced sepsis; and sham (surgical procedure without CLP). After 1 h, CLP animals received saline (CLP-saline) or 106 BMDMCs (CLP-cell) via the jugular vein. Lungs and kidneys were evaluated for histology and molecular biology after 6, 12, and 24 h. RESULTS In lungs, BMDMCs reduced the lung injury score and keratinocyte chemoattractant mRNA expression regardless of the time point of analysis; interleukin-10 mRNA content was higher in CLP-cell than CLP-saline at 6 and 24 h. In kidneys, BMDMCs reduced neutrophil gelatinase-associated lipocalin levels at all time points. BMDMCs decreased surface high mobility group box (HMGB)- 1 but increased nuclear HMGB-1 levels. CONCLUSION Early BMDMC therapy reduced lung and kidney damage in a time-dependent manner.
Collapse
Affiliation(s)
- Carla M Silva
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil; Laboratory of Cellular and Molecular Physiology, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil; National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil
| | - Debora S Ornellas
- Laboratory of Cellular and Molecular Physiology, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Felipe M Ornellas
- Laboratory of Cellular and Molecular Physiology, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil; Laboratory of Cellular, Genetic and Molecular Nephrology, Renal Division, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | - Raquel S Santos
- Laboratory of Cellular and Molecular Physiology, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Sabrina V Martini
- Laboratory of Cellular and Molecular Physiology, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Debora Ferreira
- Laboratory of Cellular and Molecular Physiology, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Caroline Muiler
- Laboratory of Cellular and Molecular Physiology, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Fernanda F Cruz
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil; National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil
| | - Christina M Takiya
- Immunopathology Laboratory, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Patricia R M Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil; National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil
| | - Marcelo M Morales
- Laboratory of Cellular and Molecular Physiology, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil; National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil
| | - Pedro L Silva
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil; National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil.
| |
Collapse
|
2
|
Han J, Li G, Hou M, Ng J, Kwon MY, Xiong K, Liang X, Taglauer E, Shi Y, Mitsialis SA, Kourembanas S, El-Chemaly S, Lederer JA, Rosas IO, Perrella MA, Liu X. Intratracheal transplantation of trophoblast stem cells attenuates acute lung injury in mice. Stem Cell Res Ther 2021; 12:487. [PMID: 34461993 PMCID: PMC8404310 DOI: 10.1186/s13287-021-02550-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 08/08/2021] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Acute lung injury (ALI) is a common lung disorder that affects millions of people every year. The infiltration of inflammatory cells into the lungs and death of the alveolar epithelial cells are key factors to trigger a pathological cascade. Trophoblast stem cells (TSCs) are immune privileged, and demonstrate the capability of self-renewal and multipotency with differentiation into three germ layers. We hypothesized that intratracheal transplantation of TSCs may alleviate ALI. METHODS ALI was induced by intratracheal delivery of bleomycin (BLM) in mice. After exposure to BLM, pre-labeled TSCs or fibroblasts (FBs) were intratracheally administered into the lungs. Analyses of the lungs were performed for inflammatory infiltrates, cell apoptosis, and engraftment of TSCs. Pro-inflammatory cytokines/chemokines of lung tissue and in bronchoalveolar lavage fluid (BALF) were also assessed. RESULTS The lungs displayed a reduction in cellularity, with decreased CD45+ cells, and less thickening of the alveolar walls in ALI mice that received TSCs compared with ALI mice receiving PBS or FBs. TSCs decreased infiltration of neutrophils and macrophages, and the expression of interleukin (IL) 6, monocyte chemoattractant protein-1 (MCP-1) and keratinocyte-derived chemokine (KC) in the injured lungs. The levels of inflammatory cytokines in BALF, particularly IL-6, were decreased in ALI mice receiving TSCs, compared to ALI mice that received PBS or FBs. TSCs also significantly reduced BLM-induced apoptosis of alveolar epithelial cells in vitro and in vivo. Transplanted TSCs integrated into the alveolar walls and expressed aquaporin 5 and prosurfactant protein C, markers for alveolar epithelial type I and II cells, respectively. CONCLUSION Intratracheal transplantation of TSCs into the lungs of mice after acute exposure to BLM reduced pulmonary inflammation and cell death. Furthermore, TSCs engrafted into the alveolar walls to form alveolar epithelial type I and II cells. These data support the use of TSCs for the treatment of ALI.
Collapse
Affiliation(s)
- Junwen Han
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, 75 Francis Street, Boston, MA, 02115, USA
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Gu Li
- Department of Surgery, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Minmin Hou
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, 75 Francis Street, Boston, MA, 02115, USA
| | - Julie Ng
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, 75 Francis Street, Boston, MA, 02115, USA
| | - Min-Young Kwon
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, 75 Francis Street, Boston, MA, 02115, USA
| | - Kevin Xiong
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, 75 Francis Street, Boston, MA, 02115, USA
| | - Xiaoliang Liang
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, 75 Francis Street, Boston, MA, 02115, USA
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, Baylor College of Medicine, Houston, TX, 77024, USA
| | - Elizabeth Taglauer
- Department of Pediatrics, Division of Newborn Medicine, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Yuanyuan Shi
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - S Alex Mitsialis
- Department of Pediatrics, Division of Newborn Medicine, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Stella Kourembanas
- Department of Pediatrics, Division of Newborn Medicine, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Souheil El-Chemaly
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, 75 Francis Street, Boston, MA, 02115, USA
| | - James A Lederer
- Department of Surgery, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Ivan O Rosas
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, 75 Francis Street, Boston, MA, 02115, USA
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, Baylor College of Medicine, Houston, TX, 77024, USA
| | - Mark A Perrella
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, 75 Francis Street, Boston, MA, 02115, USA
- Department of Pediatric Newborn Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Xiaoli Liu
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, 75 Francis Street, Boston, MA, 02115, USA.
- Department of Pediatric Newborn Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA.
| |
Collapse
|
3
|
Ghafouri-Fard S, Niazi V, Hussen BM, Omrani MD, Taheri M, Basiri A. The Emerging Role of Exosomes in the Treatment of Human Disorders With a Special Focus on Mesenchymal Stem Cells-Derived Exosomes. Front Cell Dev Biol 2021; 9:653296. [PMID: 34307345 PMCID: PMC8293617 DOI: 10.3389/fcell.2021.653296] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 06/10/2021] [Indexed: 12/14/2022] Open
Abstract
Extracellular vesicles (EVs) are produced by diverse eukaryotic and prokaryotic cells. They have prominent roles in the modulation of cell-cell communication, inflammation versus immunomodulation, carcinogenic processes, cell proliferation and differentiation, and tissue regeneration. These acellular vesicles are more promising than cellular methods because of the lower risk of tumor formation, autoimmune responses and toxic effects compared with cell therapy. Moreover, the small size and lower complexity of these vesicles compared with cells have made their production and storage easier than cellular methods. Exosomes originated from mesenchymal stem cells has also been introduced as therapeutic option for a number of human diseases. The current review aims at summarization of the role of EVs in the regenerative medicine with a focus on their therapeutic impacts in liver fibrosis, lung disorders, osteoarthritis, colitis, myocardial injury, spinal cord injury and retinal injury.
Collapse
Affiliation(s)
- Soudeh Ghafouri-Fard
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Vahid Niazi
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Bashdar Mahmud Hussen
- Department of Pharmacognosy, College of Pharmacy, Hawler Medical University, Erbil, Iraq
| | - Mir Davood Omrani
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Taheri
- Skull Base Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abbas Basiri
- Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
4
|
Bone Marrow-Derived Mononuclear Cell Transplantation Can Reduce Systemic Inflammation and Endothelial Glycocalyx Damage in Sepsis. Shock 2020; 56:260-267. [PMID: 33337736 DOI: 10.1097/shk.0000000000001710] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
ABSTRACT Bone marrow-derived mononuclear cells (BMMNCs) secrete anti-inflammatory mediators that protect against acute inflammation. Current evidence suggests that BMMNC transplantation can reduce acute tissue injury caused by systemic inflammation and lung dysfunction. This study evaluated the role of BMMNCs in reducing systemic inflammatory responses to vascular endothelial injury in sepsis. Bone marrow cells were harvested from the tibias and femurs of 12-week-old male Wistar rats; BMMNCs were separated by density centrifugation. Additional rats underwent cecal ligation and puncture (CLP) or similar sham surgery. BMMNCs were injected intravenously 30 min after CLP. The Sham and CLP Control groups were administered PBS. The 7-day survival rate improved markedly in the CLP-BMMNC group compared with that in the Control group. BMMNCs markedly suppressed the serum levels of pro-inflammatory mediators such as tumor necrosis factor-alpha, interleukin-6, and histone H3 at 3, 6, and 12 h after CLP. In the CLP-BMMNC group, the serum levels of syndecan-1, the main component of the vascular endothelial glycocalyx layer, were notably lower than those in the Control group 6 h after CLP. Histological analysis revealed improvement of morphological damages in the CLP-BMMNC group. Ultrastructural analysis revealed that the glycocalyx structure was maintained and the continuity of the vascular endothelial glycocalyx layer was preserved in the BMMNC group, compared with the case for the Control group at 6 and 12 h. Therefore, BMMNC transplantation may provide reduced systemic inflammation and endothelial glycocalyx damage, dramatically improving the survival of rats. These findings provide insights into formulating potential therapeutic strategies against sepsis.
Collapse
|
5
|
Lima MN, Oliveira HA, Fagundes PM, Estato V, Silva AYO, Freitas RJRX, Passos BABR, Oliveira KS, Batista CN, Vallochi AL, Rocco PRM, Castro-Faria-Neto HC, Maron-Gutierrez T. Mesenchymal stromal cells protect against vascular damage and depression-like behavior in mice surviving cerebral malaria. Stem Cell Res Ther 2020; 11:367. [PMID: 32843073 PMCID: PMC7448996 DOI: 10.1186/s13287-020-01874-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 07/16/2020] [Accepted: 08/04/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Malaria is one of the most critical global infectious diseases. Severe systemic inflammatory diseases, such as cerebral malaria, lead to the development of cognitive and behavioral alterations, such as learning disabilities and loss of memory capacity, as well as increased anxiety and depression. The consequences are profound and usually contribute to reduce the patient's quality of life. There are no therapies to treat the neurological sequelae of cerebral malaria. Mesenchymal stromal cells (MSCs) may be an alternative, since they have been used as therapy for neurodegenerative diseases and traumatic lesions of the central nervous system. So far, no study has investigated the effects of MSC therapy on the blood-brain barrier, leukocyte rolling and adherence in the brain, and depression like-behavior in experimental cerebral malaria. METHODS Male C57BL/6 mice were infected with Plasmodium berghei ANKA (PbA, 1 × 106 PbA-parasitized red blood cells, intraperitoneally). At day 6, PbA-infected animals received chloroquine (25 mg/kg orally for seven consecutive days) as the antimalarial treatment and were then randomized to receive MSCs (1 × 105 cells in 0.05 ml of saline/mouse) or saline (0.05 ml) intravenously. Parasitemia, clinical score, and survival rate were analyzed throughout the experiments. Evans blue assay was performed at 6, 7, and 15 days post-infection (dpi). Behavioral tests were performed at 5 and 15 dpi. Intravital microscopy experiments and brain-derived neurotrophic factor (BDNF) protein expression analyses were performed at 7 dpi, whereas inflammatory mediators were measured at 15 dpi. In vitro, endothelial cells were used to evaluate the effects of conditioned media derived from MSCs (CMMSC) on cell viability by lactate dehydrogenase (LDH) release. RESULTS PbA-infected mice presented increased parasitemia, adherent leukocytes, blood-brain barrier permeability, and reduced BDNF protein levels, as well as depression-like behavior. MSCs mitigated behavioral alterations, restored BDNF and transforming growth factor (TGF)-β protein levels, and reduced blood-brain barrier dysfunction and leukocyte adhesion in the brain microvasculature. In a cultured endothelial cell line stimulated with heme, CMMSC reduced LDH release, suggesting a paracrine mechanism of action. CONCLUSION A single dose of MSCs as adjuvant therapy protected against vascular damage and improved depression-like behavior in mice that survived experimental cerebral malaria.
Collapse
Affiliation(s)
- Maiara N Lima
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Fiocruz, Av. Brasil, 4365, Pavilhão 108, sala 45, Manguinhos, Rio de Janeiro, RJ, 21040-360, Brazil
| | - Helena A Oliveira
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Fiocruz, Av. Brasil, 4365, Pavilhão 108, sala 45, Manguinhos, Rio de Janeiro, RJ, 21040-360, Brazil
| | - Paula M Fagundes
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Fiocruz, Av. Brasil, 4365, Pavilhão 108, sala 45, Manguinhos, Rio de Janeiro, RJ, 21040-360, Brazil
| | - Vanessa Estato
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Fiocruz, Av. Brasil, 4365, Pavilhão 108, sala 45, Manguinhos, Rio de Janeiro, RJ, 21040-360, Brazil
| | - Adriano Y O Silva
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Fiocruz, Av. Brasil, 4365, Pavilhão 108, sala 45, Manguinhos, Rio de Janeiro, RJ, 21040-360, Brazil
| | - Rodrigo J R X Freitas
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Fiocruz, Av. Brasil, 4365, Pavilhão 108, sala 45, Manguinhos, Rio de Janeiro, RJ, 21040-360, Brazil
| | - Beatriz A B R Passos
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Fiocruz, Av. Brasil, 4365, Pavilhão 108, sala 45, Manguinhos, Rio de Janeiro, RJ, 21040-360, Brazil
| | - Karina S Oliveira
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Fiocruz, Av. Brasil, 4365, Pavilhão 108, sala 45, Manguinhos, Rio de Janeiro, RJ, 21040-360, Brazil
| | - Camila N Batista
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Fiocruz, Av. Brasil, 4365, Pavilhão 108, sala 45, Manguinhos, Rio de Janeiro, RJ, 21040-360, Brazil
| | - Adriana L Vallochi
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Fiocruz, Av. Brasil, 4365, Pavilhão 108, sala 45, Manguinhos, Rio de Janeiro, RJ, 21040-360, Brazil
| | - Patricia R M Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, RJ, Brazil
| | - Hugo C Castro-Faria-Neto
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Fiocruz, Av. Brasil, 4365, Pavilhão 108, sala 45, Manguinhos, Rio de Janeiro, RJ, 21040-360, Brazil
- National Institute of Science and Technology on Neuroimmunomodulation, Rio de Janeiro, RJ, Brazil
| | - Tatiana Maron-Gutierrez
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Fiocruz, Av. Brasil, 4365, Pavilhão 108, sala 45, Manguinhos, Rio de Janeiro, RJ, 21040-360, Brazil.
- National Institute of Science and Technology on Neuroimmunomodulation, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
6
|
Deconstructing tissue engineered trachea: Assessing the role of synthetic scaffolds, segmental replacement and cell seeding on graft performance. Acta Biomater 2020; 102:181-191. [PMID: 31707085 DOI: 10.1016/j.actbio.2019.11.008] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 11/04/2019] [Accepted: 11/05/2019] [Indexed: 01/05/2023]
Abstract
The ideal construct for tracheal replacement remains elusive in the management of long segment airway defects. Tissue engineered tracheal grafts (TETG) have been limited by the development of graft stenosis or collapse, infection, or lack of an epithelial lining. We applied a mouse model of orthotopic airway surgery to assess the impact of three critical barriers encountered in clinical applications: the scaffold, the extent of intervention, and the impact of cell seeding and characterized their impact on graft performance. First, synthetic tracheal scaffolds electrospun from polyethylene terephthalate / polyurethane (PET/PU) were orthotopically implanted in anterior tracheal defects of C57BL/6 mice. Scaffolds demonstrated complete coverage with ciliated respiratory epithelium by 2 weeks. Epithelial migration was accompanied by macrophage infiltration which persisted at long term (>6 weeks) time points. We then assessed the impact of segmental tracheal implantation using syngeneic trachea as a surrogate for the ideal tracheal replacement. Graft recovery involved local upregulation of epithelial progenitor populations and there was no evidence of graft stenosis or necrosis. Implantation of electrospun synthetic tracheal scaffold for segmental replacement resulted in respiratory distress and required euthanasia at an early time point. There was limited epithelial coverage of the scaffold with and without seeded bone marrow-derived mononuclear cells (BM-MNCs). We conclude that synthetic scaffolds support re-epithelialization in orthotopic patch implantation, syngeneic graft integration occurs with focal repair mechanisms, however epithelialization in segmental synthetic scaffolds is limited and is not influenced by cell seeding. STATEMENT OF SIGNIFICANCE: The life-threatening nature of long-segment tracheal defects has led to clinical use of tissue engineered tracheal grafts in the last decade for cases of compassionate use. However, the ideal tracheal reconstruction using tissue-engineered tracheal grafts (TETG) has not been clarified. We addressed the core challenges in tissue engineered tracheal replacement (re-epithelialization and graft patency) by defining the role of cell seeding with autologous bone marrow-derived mononuclear cells, the mechanism of respiratory epithelialization and proliferation, and the role of the inflammatory immune response in regeneration. This research will facilitate comprehensive understanding of cellular regeneration and neotissue formation on TETG, which will permit targeted therapies for accelerating re-epithelialization and attenuating stenosis in tissue engineered airway replacement.
Collapse
|
7
|
Liu A, Zhang X, He H, Zhou L, Naito Y, Sugita S, Lee JW. Therapeutic potential of mesenchymal stem/stromal cell-derived secretome and vesicles for lung injury and disease. Expert Opin Biol Ther 2019; 20:125-140. [PMID: 31701782 DOI: 10.1080/14712598.2020.1689954] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Introduction: The acute respiratory distress syndrome (ARDS) is a devastating clinical condition common in patients with respiratory failure. Based largely on numerous preclinical studies and recent Phase I/II clinical trials, administration of stem cells, specifically mesenchymal stem or stromal cells (MSC), as a therapeutic for acute lung injury (ALI) holds great promise. However, concern for the use of stem cells, specifically the risk of iatrogenic tumor formation, remains unresolved. Accumulating evidence now suggest that stem cell-derived conditioned medium (CM) and/or extracellular vesicles (EV) might constitute compelling alternatives.Areas covered: The current review focuses on the preclinical studies testing MSC CM and/or EV as treatment for ALI and other inflammatory lung diseases.Expert opinion: Clinical application of MSC or their secreted CM may be limited by the cost of growing enough cells, the logistic of MSC storage, and the lack of standardization of what constitutes MSC CM. However, the clinical application of MSC EV remains promising, primarily due to the ability of EV to maintain the functional phenotype of the parent cell as a therapeutic. However, utilization of MSC EV will also require large-scale production, the cost of which may be prohibitive unless the potency of the EV can be increased.
Collapse
Affiliation(s)
- Airan Liu
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Xiwen Zhang
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Hongli He
- Departments of Anesthesiology and Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| | - Li Zhou
- Departments of Anesthesiology and Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| | - Yoshifumi Naito
- Departments of Anesthesiology and Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| | - Shinji Sugita
- Departments of Anesthesiology and Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| | - Jae-Woo Lee
- Departments of Anesthesiology and Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| |
Collapse
|
8
|
Eicosapentaenoic acid potentiates the therapeutic effects of adipose tissue-derived mesenchymal stromal cells on lung and distal organ injury in experimental sepsis. Stem Cell Res Ther 2019; 10:264. [PMID: 31443678 PMCID: PMC6708232 DOI: 10.1186/s13287-019-1365-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 07/07/2019] [Accepted: 07/29/2019] [Indexed: 12/14/2022] Open
Abstract
Background Even though mesenchymal stromal cells (MSCs) mitigate lung and distal organ damage in experimental polymicrobial sepsis, mortality remains high. We investigated whether preconditioning with eicosapentaenoic acid (EPA) would potentiate MSC actions in experimental sepsis by further decreasing lung and distal organ injury, thereby improving survival. Methods In C57BL/6 mice, sepsis was induced by cecal hligation and puncture (CLP); sham-operated animals were used as control. Twenty-four hours after surgery, CLP mice were further randomized to receive saline, adipose tissue-derived (AD)-MSCs (105, nonpreconditioned), or AD-MSCs preconditioned with EPA for 6 h (105, EPA-preconditioned MSCs) intravenously. After 24 h, survival rate, sepsis severity score, lung mechanics and histology, protein level of selected biomarkers in lung tissue, cellularity in blood, distal organ damage, and MSC distribution (by technetium-99m tagging) were analyzed. Additionally, the effects of EPA on the secretion of resolvin-D1 (RvD1), prostaglandin E2 (PGE2), interleukin (IL)-10, and transforming growth factor (TGF)-β1 by MSCs were evaluated in vitro. Results Nonpreconditioned and EPA-preconditioned AD-MSCs exhibited similar viability and differentiation capacity, accumulated mainly in the lungs and kidneys following systemic administration. Compared to nonpreconditioned AD-MSCs, EPA-preconditioned AD-MSCs further reduced static lung elastance, alveolar collapse, interstitial edema, alveolar septal inflammation, collagen fiber content, neutrophil cell count as well as protein levels of interleukin-1β and keratinocyte chemoattractant in lung tissue, and morphological abnormalities in the heart (cardiac myocyte architecture), liver (hepatocyte disarrangement and Kupffer cell hyperplasia), kidney (acute tubular necrosis), spleen (increased number of megakaryocytes and lymphocytes), and small bowel (villi architecture disorganization). EPA preconditioning of MSCs resulted in increased secretion of pro-resolution and anti-inflammatory mediators (RvD1, PGE2, IL-10, and TGF-β). Conclusions Compared to nonpreconditioned cells, EPA-preconditioned AD-MSCs yielded further reductions in the lung and distal organ injury, resulting in greater improvement in sepsis severity score and higher survival rate in CLP-induced experimental sepsis. This may be a promising therapeutic approach to improve outcome in septic patients. Electronic supplementary material The online version of this article (10.1186/s13287-019-1365-z) contains supplementary material, which is available to authorized users.
Collapse
|
9
|
Mesenchymal Stromal Cells Are More Effective Than Their Extracellular Vesicles at Reducing Lung Injury Regardless of Acute Respiratory Distress Syndrome Etiology. Stem Cells Int 2019; 2019:8262849. [PMID: 31531026 PMCID: PMC6720722 DOI: 10.1155/2019/8262849] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 07/01/2019] [Accepted: 07/21/2019] [Indexed: 02/07/2023] Open
Abstract
Although mesenchymal stromal cells (MSCs) have demonstrated beneficial effects on experimental acute respiratory distress syndrome (ARDS), preconditioning may be required to potentiate their therapeutic effects. Additionally, administration of cell-free products, such as extracellular vesicles (EVs) obtained from MSC-conditioned media, might be as effective as MSCs. In this study, we comparatively evaluated the effects of MSCs, preconditioned or not with serum collected from mice with pulmonary or extrapulmonary ARDS (ARDSp and ARDSexp, respectively), and the EVs derived from these cells on lung inflammation and remodeling in ARDSp and ARDSexp mice. Administration of MSCs (preconditioned or not), but not their EVs, reduced static lung elastance, interstitial edema, and collagen fiber content in both ARDSp and ARDSexp. Although MSCs and EVs reduced alveolar collapse and neutrophil cell counts in lung tissue, therapeutic responses were superior in mice receiving MSCs, regardless of preconditioning. Despite higher total cell, macrophage, and neutrophil counts in bronchoalveolar lavage fluid in ARDSp than ARDSexp, MSCs and EVs (preconditioned or not) led to a similar decrease. In ARDSp, both MSCs and EVs, regardless of preconditioning, reduced levels of tumor necrosis factor- (TNF-) α, interleukin-6, keratinocyte chemoattractant (KC), vascular endothelial growth factor (VEGF), and transforming growth factor- (TGF-) β in lung homogenates. In ARDSexp, TNF-α, interleukin-6, and KC levels were reduced by MSCs and EVs, preconditioned or not; only MSCs reduced VEGF levels, while TGF-β levels were similarly increased in ARDSexp treated either with saline, MSCs, or EVs, regardless of preconditioning. In conclusion, MSCs yielded greater overall improvement in ARDS in comparison to EVs derived from the same number of cells and regardless of the preconditioning status. However, the effects of MSCs and EVs differed according to ARDS etiology.
Collapse
|
10
|
Felix RG, Bovolato ALC, Cotrim OS, Leão PDS, Batah SS, Golim MDA, Velosa AP, Teodoro W, Martins V, Cruz FF, Deffune E, Fabro AT, Capelozzi VL. Adipose-derived stem cells and adipose-derived stem cell-conditioned medium modulate in situ imbalance between collagen I- and collagen V-mediated IL-17 immune response recovering bleomycin pulmonary fibrosis. Histol Histopathol 2019; 35:289-301. [PMID: 31318036 DOI: 10.14670/hh-18-152] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The immunogenic collagen V (Col V) and the proinflammatory cytokine interleukin (IL)-17 have been implicated in the pathogenesis of multiple autoimmune diseases. Col V is also up-regulated during adipogenesis and can stimulate adipocyte differentiation in vitro. Conditioned medium (CM) generated from adipose-derived mesenchymal stem cells (MSCs) reduces bleomycin (BLM)-induced lung injury in rats, suggesting a crucial role in situ of immunomodulatory factors secreted by MSCs in these beneficial effects. In the present work, we investigated this hypothesis, analyzing levels of plasma inflammatory mediators and inflammatory and fibrotic mediators in the lung tissue of BLM-injured rats after treatment with MSCs and CM. Pulmonary fibrosis was intratracheally induced by BLM. After 10 days, BLM animals were further randomized into subgroups receiving saline, MSCs, or CM intravenously. On days 14 and 21, the animals were euthanized, and the lungs were examined through protein expression of nitric oxide synthase (NOS), IL-17, transforming growth factor-β (TGF-β), vascular endothelial growth factor, endothelin-1, and the immunogenic Col V through histological quantitative evaluation and plasma levels of fibrinogen, Von Willebrand factor, and platelet-derived growth factor (PDGF). Rats that had been injected with MSCs and CM showed a significant increase in weight and significant improvements at 14 and 21 days after intravenous injection at both time points of analysis of plasma fibrinogen, PDGF, and Von Willebrand factor and NOS-2 expression, supporting an early anti-inflammatory action, thus reducing TGF-β and collagen I fibers. In contrast, intravenous injection of CM was able to significantly increase the deposition of Col V fibers and IL-17 on both day 14 and day 21 as compared with the amount observed in rats from the BLM group and MSC groups. In conclusion, this study reinforces previous observations on the therapeutic properties of MSCs and CM and is the first report to demonstrate the association of its actions with immunomodulatory biomarkers on lung tissue. We concluded that adipose-derived stem cells and adipose-derived stem cells-CM modulate an in situ imbalance between collagen I- and Col V-mediated IL-17 immune response, emerging as a promising therapeutic option for recovering from BLM pulmonary fibrosis.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Ana Paula Velosa
- Rheumatology Division, Faculdade de Medicina, Universidade de São Paulo, Brazil
| | - Walcy Teodoro
- Rheumatology Division, Faculdade de Medicina, Universidade de São Paulo, Brazil
| | - Vanessa Martins
- Department of Pathology, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | - Fernanda Ferreira Cruz
- Laboratory of Pulmonary Investigation, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Brazil
| | | | | | - Vera Luiza Capelozzi
- Department of Pathology, Faculty of Medicine, University of São Paulo, São Paulo, Brazil.
| |
Collapse
|
11
|
de Oliveira MV, Rocha NDN, Santos RS, Rocco MRM, de Magalhães RF, Silva JD, Souza SAL, Capelozzi VL, Pelosi P, Silva PL, Rocco PRM. Endotoxin-Induced Emphysema Exacerbation: A Novel Model of Chronic Obstructive Pulmonary Disease Exacerbations Causing Cardiopulmonary Impairment and Diaphragm Dysfunction. Front Physiol 2019; 10:664. [PMID: 31191356 PMCID: PMC6546905 DOI: 10.3389/fphys.2019.00664] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 05/09/2019] [Indexed: 12/26/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a progressive disorder of the lung parenchyma which also involves extrapulmonary manifestations, such as cardiovascular impairment, diaphragm dysfunction, and frequent exacerbations. The development of animal models is important to elucidate the pathophysiology of COPD exacerbations and enable analysis of possible therapeutic approaches. We aimed to characterize a model of acute emphysema exacerbation and evaluate its consequences on the lung, heart, and diaphragm. Twenty-four Wistar rats were randomly assigned into one of two groups: control (C) or emphysema (ELA). In ELA group, animals received four intratracheal instillations of pancreatic porcine elastase (PPE) at 1-week intervals. The C group received saline under the same protocol. Five weeks after the last instillation, C and ELA animals received saline (SAL) or E. coli lipopolysaccharide (LPS) (200 μg in 200 μl) intratracheally. Twenty-four hours after saline or endotoxin administration, arterial blood gases, lung inflammation and morphometry, collagen fiber content, and lung mechanics were analyzed. Echocardiography, diaphragm ultrasonography (US), and computed tomography (CT) of the chest were done. ELA-LPS animals, compared to ELA-SAL, exhibited decreased arterial oxygenation; increases in alveolar collapse (p < 0.0001), relative neutrophil counts (p = 0.007), levels of cytokine-induced neutrophil chemoattractant-1, interleukin (IL)-1β, tumor necrosis factor-α, IL-6, and vascular endothelial growth factor in lung tissue, collagen fiber deposition in alveolar septa, airways, and pulmonary vessel walls, and dynamic lung elastance (p < 0.0001); reduced pulmonary acceleration time/ejection time ratio, (an indirect index of pulmonary arterial hypertension); decreased diaphragm thickening fraction and excursion; and areas of emphysema associated with heterogeneous alveolar opacities on chest CT. In conclusion, we developed a model of endotoxin-induced emphysema exacerbation that affected not only the lungs but also the heart and diaphragm, thus resembling several features of human disease. This model of emphysema should allow preclinical testing of novel therapies with potential for translation into clinical practice.
Collapse
Affiliation(s)
- Milena Vasconcellos de Oliveira
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Nazareth de Novaes Rocha
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,Department of Physiology and Pharmacology, Biomedical Institute, Fluminense Federal University, Niterói, Brazil
| | - Raquel Souza Santos
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcella Rieken Macedo Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Raquel Ferreira de Magalhães
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Johnatas Dutra Silva
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Sergio Augusto Lopes Souza
- Department of Radiology, Faculty of Medicine, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Vera Luiza Capelozzi
- Department of Pathology, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | - Paolo Pelosi
- Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa, Genoa, Italy.,San Martino Policlinico Hospital, IRCCS for Oncology and Neurosciences, Genoa, Italy
| | - Pedro Leme Silva
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Patricia Rieken Macedo Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
12
|
Umemura Y, Ogura H, Matsuura H, Ebihara T, Shimizu K, Shimazu T. Bone marrow-derived mononuclear cell therapy can attenuate systemic inflammation in rat heatstroke. Scand J Trauma Resusc Emerg Med 2018; 26:97. [PMID: 30445981 PMCID: PMC6240199 DOI: 10.1186/s13049-018-0566-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 11/02/2018] [Indexed: 12/30/2022] Open
Abstract
Background This study was performed to gain insights into novel therapeutic approaches for acute systemic inflammation in heatstroke. Bone marrow-derived mononuclear cells (BMMNCs) secrete anti-inflammatory proteins and have protective effects against acute inflammation. Recent evidence suggested that transplantation of BMMNCs can reduce the acute tissue injury caused by regional myocardial reperfusion and the lung dysfunction induced by lipopolysaccharides. We evaluated whether BMMNCs attenuate systemic inflammatory response induced by severe heatstroke. Material and methods Anesthetized 12-week-old male Wistar rats were subjected to heat stress (41.8 °C for 30 min) with/without transplantation of BMMNCs. Bone marrow cells were harvested from the femur and tibia of other Wistar rats. BMMNCs were separated by density centrifugation, dissolved in phosphate-buffered saline (PBS), and injected intravenously immediately after heat stress (HS-BMMNCs group). The control group was administered an equal volume of PBS, and the sham group underwent the same procedure without heat stress. Results Seven-day survival improved significantly in the HS-BMMNCs group versus control group (83.3% vs 41.7%). Transplantation of BMMNCs significantly suppressed serum levels of pro-inflammatory mediators, such as tumor necrosis factor-alpha, interleukin-6 and histone H3 at 3, 6, and 12 h after heat stress. Besides, the elevation of serum syndecan-1, a main component of the vascular endothelial glycocalyx layer, in the BMMNCs group was significantly suppressed compared to that in the control group at 6 and 12 h after heat stress. Histological analysis revealed that edema of the alveolar septum and vascular endothelial injury in the lung were evident in the control group 6 h after heat stress, whereas the morphological alteration was ameliorated in the HS-BMMNCs group. Also, histological analysis using BMMNCs derived from green fluorescent protein transgenic rats showed that the transplanted BMMNCs migrated into lung, kidney, and spleen at 24 h after heat stress but did not engraft to host tissues. Conclusion Transplantation of BMMNCs attenuated acute systemic inflammation and vascular endothelial injury, reduced organ dysfunction, and improved survival in a rat heatstroke model. These findings provide a possible therapeutic strategy against critical heatstroke.
Collapse
Affiliation(s)
- Yutaka Umemura
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, 2-15 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Hiroshi Ogura
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, 2-15 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Hiroshi Matsuura
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, 2-15 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Takeshi Ebihara
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, 2-15 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Kentaro Shimizu
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, 2-15 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Takeshi Shimazu
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, 2-15 Yamadaoka, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
13
|
Zhu Y, Chen X, Yang X, El-Hashash A. Stem cells in lung repair and regeneration: Current applications and future promise. J Cell Physiol 2018; 233:6414-6424. [PMID: 29271480 DOI: 10.1002/jcp.26414] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 12/19/2017] [Indexed: 12/18/2022]
Abstract
Lung diseases are major cause of morbidity and mortality worldwide. The progress in regenerative medicine and stem cell research in the lung are currently a fast-growing research topic that can provide solutions to these major health problems. Under normal conditions, the rate of cellular proliferation is relatively low in the lung in vivo, compared to other major organ systems. Lung injury leads to the activation of stem/progenitor cell populations that re-enter the cell cycle. Yet, little is known about stem cells in the lung, despite common thoughts that these cells could play a critical role in the repair of lung injuries. Nor do we fully understand the cellular and architectural complexity of the respiratory tract, and the diverse stem/progenitor cells that are involved in the lung repair and regeneration. In this review, we discuss the conceptual framework of lung stem/progenitor cell biology, and describe lung diseases, in which stem cell manipulations may be physiologically significant. In addition, we highlight the challenges of lung stem cell-based therapy.
Collapse
Affiliation(s)
- Yuqing Zhu
- Centre of Stem cell and Regenerative Medicine, Schools of Medicine and Basic Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xiao Chen
- Centre of Stem cell and Regenerative Medicine, Schools of Medicine and Basic Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xu Yang
- Section of Environmental Biomedicine, School of Life Science, Central China Normal University, Wuhan, Hubei, China
| | - Ahmed El-Hashash
- Centre of Stem cell and Regenerative Medicine, Schools of Medicine and Basic Medicine, Zhejiang University, Hangzhou, Zhejiang, China.,University of Edinburgh-Zhejiang University Institute (UoE-ZJU Institute), Haining, Zhejiang, China.,Edinburgh Medical School, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
14
|
Zheng Y, Liu SQ, Sun Q, Xie JF, Xu JY, Li Q, Pan C, Liu L, Huang YZ. Plasma microRNAs levels are different between pulmonary and extrapulmonary ARDS patients: a clinical observational study. Ann Intensive Care 2018; 8:23. [PMID: 29442256 PMCID: PMC5811418 DOI: 10.1186/s13613-018-0370-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 02/05/2018] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSC) obviously alleviate the damage of the structure and function of pulmonary vascular endothelial cells (VEC). The therapeutic effects of MSC are significantly different between pulmonary ARDS (ARDSp) and extrapulmonary ARDS (ARDSexp). MicroRNAs (miRNAs), as important media of MSC regulating VEC, are not studied between ARDSp and ARDSexp. We aimed to explore the plasma levels difference of miRNAs that regulate VEC function and are associated with MSC (MSC-VEC-miRNAs) between ARDSp and ARDSexp patients. METHODS MSC-VEC-miRNAs were obtained through reviewing relevant literatures screened in PubMed database. We enrolled 57 ARDS patients within 24 h of admission to the ICU and then collected blood samples, extracted plasma supernatant. Patients' clinical data were collected. Then, plasma expression of MSC-VEC-miRNAs was measured by real-time fluorescence quantitative PCR. Simultaneously, plasma endothelial injury markers VCAM-1, vWF and inflammatory factors TNF-α, IL-10 were detected by ELISA method. RESULTS Fourteen miRNAs were picked out after screening. A total of 57 ARDS patients were included in this study, among which 43 cases pertained to ARDSp group and 14 cases pertained to ARDSexp group. Plasma miR-221 and miR-27b levels in ARDSexp group exhibited significantly lower than that in ARDSp group (miR-221, 0.22 [0.12-0.49] vs. 0.57 [0.22-1.57], P = 0.008, miR-27b, 0.34 [0.10-0.46] vs. 0.60 [0.20-1.46], P = 0.025). Plasma vWF concentration in ARDSexp group exhibited significantly lower than that in ARDSp group (0.77 [0.29-1.54] vs. 1.80 [0.95-3.51], P = 0.048). Significant positive correlation was found between miR-221 and vWF in plasma levels (r = 0.688, P = 0.022). Plasma miR-26a and miR-27a levels in non-survival group exhibited significantly lower than that in survival group (miR-26a, 0.17 [0.08-0.20] vs. 0.69 [0.24-2.33] P = 0.018, miR-27a, 0.23 [0.16-0.58] vs. 1.45 [0.38-3.63], P = 0.021) in ARDSp patients. CONCLUSION Plasma miR-221, miR-27b and vWF levels in ARDSexp group are significantly lower than that in ARDSp group. Plasma miR-26a and miR-27a levels in non-survival group are significantly lower than that in survival group in ARDSp patients.
Collapse
Affiliation(s)
- Yi Zheng
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, No. 87, Dingjiaqiao Road, Gulou District, Nanjing, 210009, China.,Department of Critical Care Medicine, The First Affiliated Hospital of Medical School of Zhejiang University, 79 Qingchun Road, Shangcheng District, Hangzhou, 310003, China
| | - Song-Qiao Liu
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, No. 87, Dingjiaqiao Road, Gulou District, Nanjing, 210009, China
| | - Qin Sun
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, No. 87, Dingjiaqiao Road, Gulou District, Nanjing, 210009, China
| | - Jian-Feng Xie
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, No. 87, Dingjiaqiao Road, Gulou District, Nanjing, 210009, China
| | - Jing-Yuan Xu
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, No. 87, Dingjiaqiao Road, Gulou District, Nanjing, 210009, China
| | - Qing Li
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, No. 87, Dingjiaqiao Road, Gulou District, Nanjing, 210009, China
| | - Chun Pan
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, No. 87, Dingjiaqiao Road, Gulou District, Nanjing, 210009, China
| | - Ling Liu
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, No. 87, Dingjiaqiao Road, Gulou District, Nanjing, 210009, China
| | - Ying-Zi Huang
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, No. 87, Dingjiaqiao Road, Gulou District, Nanjing, 210009, China.
| |
Collapse
|
15
|
An Official American Thoracic Society Workshop Report 2015. Stem Cells and Cell Therapies in Lung Biology and Diseases. Ann Am Thorac Soc 2018; 13:S259-78. [PMID: 27509163 DOI: 10.1513/annalsats.201606-466st] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The University of Vermont College of Medicine, in collaboration with the NHLBI, Alpha-1 Foundation, American Thoracic Society, Cystic Fibrosis Foundation, European Respiratory Society, International Society for Cellular Therapy, and the Pulmonary Fibrosis Foundation, convened a workshop, "Stem Cells and Cell Therapies in Lung Biology and Lung Diseases," held July 27 to 30, 2015, at the University of Vermont. The conference objectives were to review the current understanding of the role of stem and progenitor cells in lung repair after injury and to review the current status of cell therapy and ex vivo bioengineering approaches for lung diseases. These are all rapidly expanding areas of study that both provide further insight into and challenge traditional views of mechanisms of lung repair after injury and pathogenesis of several lung diseases. The goals of the conference were to summarize the current state of the field, discuss and debate current controversies, and identify future research directions and opportunities for both basic and translational research in cell-based therapies for lung diseases. This 10th anniversary conference was a follow up to five previous biennial conferences held at the University of Vermont in 2005, 2007, 2009, 2011, and 2013. Each of those conferences, also sponsored by the National Institutes of Health, American Thoracic Society, and respiratory disease foundations, has been important in helping guide research and funding priorities. The major conference recommendations are summarized at the end of the report and highlight both the significant progress and major challenges in these rapidly progressing fields.
Collapse
|
16
|
Capelozzi VL, Allen TC, Beasley MB, Cagle PT, Guinee D, Hariri LP, Husain AN, Jain D, Lantuejoul S, Larsen BT, Miller R, Mino-Kenudson M, Mehrad M, Raparia K, Roden A, Schneider F, Sholl LM, Smith ML. Molecular and Immune Biomarkers in Acute Respiratory Distress Syndrome: A Perspective From Members of the Pulmonary Pathology Society. Arch Pathol Lab Med 2017; 141:1719-1727. [DOI: 10.5858/arpa.2017-0115-sa] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Acute respiratory distress syndrome (ARDS) is a multifactorial syndrome with high morbidity and mortality rates, characterized by deficiency in gas exchange and lung mechanics that lead to hypoxemia, dyspnea, and respiratory failure. Histologically, ARDS is characterized by an acute, exudative phase, combining diffuse alveolar damage and noncardiogenic edema, followed by a later fibroproliferative phase. Despite an enhanced understanding of ARDS pathogenesis, the capacity to predict the development of ARDS and to risk-stratify patients with the disease remains limited. Biomarkers may help to identify patients at the greatest risk of developing ARDS, to evaluate response to therapy, to predict outcome, and to improve clinical trials. The ARDS pathogenesis is presented in this article, as well as concepts and information on biomarkers that are currently used clinically or are available for laboratory use by academic and practicing pathologists and the developing and validating of new assays, focusing on the assays' major biologic roles in lung injury and/or repair and to ultimately suggest innovative, therapeutic approaches.
Collapse
|
17
|
Mei SHJ, Dos Santos CC, Stewart DJ. Advances in Stem Cell and Cell-Based Gene Therapy Approaches for Experimental Acute Lung Injury: A Review of Preclinical Studies. Hum Gene Ther 2017; 27:802-812. [PMID: 27531647 DOI: 10.1089/hum.2016.063] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Given the failure of pharmacological interventions in acute respiratory distress syndrome (ARDS), researchers have been actively pursuing novel strategies to treat this devastating, life-threatening condition commonly seen in the intensive care unit. There has been considerable research on harnessing the reparative properties of stem and progenitor cells to develop more effective therapeutic approaches for respiratory diseases with limited treatment options, such as ARDS. This review discusses the preclinical literature on the use of stem and progenitor cell therapy and cell-based gene therapy for the treatment of preclinical animal models of acute lung injury (ALI). A variety of cell types that have been used in preclinical models of ALI, such as mesenchymal stem cells, endothelial progenitor cells, and induced pluripotent stem cells, were evaluated. At present, two phase I trials have been completed and one phase I/II clinical trial is well underway in order to translate the therapeutic benefit gleaned from preclinical studies in complex animal models of ALI to patients with ARDS, paving the way for what could potentially develop into transformative therapy for critically ill patients. As we await the results of these early cell therapy trials, future success of stem cell therapy for ARDS will depend on selection of the most appropriate cell type, route and timing of cell delivery, enhancing effectiveness of cells (i.e., potency), and potentially combining beneficial cells and genes (cell-based gene therapy) to maximize therapeutic efficacy. The experimental models and scientific methods exploited to date have provided researchers with invaluable knowledge that will be leveraged to engineer cells with enhanced therapeutic capabilities for use in the next generation of clinical trials.
Collapse
Affiliation(s)
- Shirley H J Mei
- 1 Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Claudia C Dos Santos
- 2 The Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, Ontario, Canada.,3 Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Duncan J Stewart
- 1 Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,4 Department of Medicine, University of Ottawa , Ottawa, Ontario, Canada
| |
Collapse
|
18
|
Carneiro PJ, Clevelario AL, Padilha GA, Silva JD, Kitoko JZ, Olsen PC, Capelozzi VL, Rocco PRM, Cruz FF. Bosutinib Therapy Ameliorates Lung Inflammation and Fibrosis in Experimental Silicosis. Front Physiol 2017; 8:159. [PMID: 28360865 PMCID: PMC5350127 DOI: 10.3389/fphys.2017.00159] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 03/01/2017] [Indexed: 12/16/2022] Open
Abstract
Silicosis is an occupational lung disease for which no effective therapy exists. We hypothesized that bosutinib, a tyrosine kinase inhibitor, might ameliorate inflammatory responses, attenuate pulmonary fibrosis, and thus improve lung function in experimental silicosis. For this purpose, we investigated the potential efficacy of bosutinib in the treatment of experimental silicosis induced in C57BL/6 mice by intratracheal administration of silica particles. After 15 days, once disease was established, animals were randomly assigned to receive DMSO or bosutinib (1 mg/kg/dose in 0.1 mL 1% DMSO) by oral gavage, twice daily for 14 days. On day 30, lung mechanics and morphometry, total and differential cell count in alveolar septa and granuloma, levels of interleukin (IL)-1β, tumor necrosis factor (TNF)-α, interferon (IFN)-γ, IL-4, transforming growth factor (TGF)-β, and vascular endothelial growth factor in lung homogenate, M1 and M2 macrophages, total leukocytes, and T cells in BALF, lymph nodes, and thymus, and collagen fiber content in alveolar septa and granuloma were analyzed. In a separate in vitro experiment, RAW264.7 macrophages were exposed to silica particles in the presence or absence of bosutinib. After 24 h, gene expressions of arginase-1, IL-10, IL-12, inducible nitric oxide synthase (iNOS), metalloproteinase (MMP)-9, tissue inhibitor of metalloproteinase (TIMP)-1, and caspase-3 were evaluated. In vivo, in silicotic animals, bosutinib, compared to DMSO, decreased: (1) fraction area of collapsed alveoli, (2) size and number of granulomas, and mononuclear cell granuloma infiltration; (3) IL-1β, TNF-α, IFN-γ, and TGF-β levels in lung homogenates, (4) collagen fiber content in lung parenchyma, and (5) viscoelastic pressure and static lung elastance. Bosutinib also reduced M1 cell counts while increasing M2 macrophage population in both lung parenchyma and granulomas. Total leukocyte, regulatory T, CD4+, and CD8+ cell counts in the lung-draining lymph nodes also decreased with bosutinib therapy without affecting thymus cellularity. In vitro, bosutinib led to a decrease in IL-12 and iNOS and increase in IL-10, arginase-1, MMP-9, and TIMP-1. In conclusion, in the current model of silicosis, bosutinib therapy yielded beneficial effects on lung inflammation and remodeling, therefore resulting in lung mechanics improvement. Bosutinib may hold promise for silicosis; however, further studies are required.
Collapse
Affiliation(s)
- Priscila J Carneiro
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro Rio de Janeiro, Brazil
| | - Amanda L Clevelario
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro Rio de Janeiro, Brazil
| | - Gisele A Padilha
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro Rio de Janeiro, Brazil
| | - Johnatas D Silva
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro Rio de Janeiro, Brazil
| | - Jamil Z Kitoko
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de JaneiroRio de Janeiro, Brazil; Laboratory of Clinical Bacteriology and Immunology, Department of Toxicological and Clinical Analysis, School of Pharmacy, Federal University of Rio de JaneiroRio de Janeiro, Brazil
| | - Priscilla C Olsen
- Laboratory of Clinical Bacteriology and Immunology, Department of Toxicological and Clinical Analysis, School of Pharmacy, Federal University of Rio de Janeiro Rio de Janeiro, Brazil
| | - Vera L Capelozzi
- Laboratory of Pulmonary Genomics, Department of Pathology, School of Medicine, University of São Paulo São Paulo, Brazil
| | - Patricia R M Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro Rio de Janeiro, Brazil
| | - Fernanda F Cruz
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro Rio de Janeiro, Brazil
| |
Collapse
|
19
|
Taki T, Masumoto H, Funamoto M, Minakata K, Yamazaki K, Ikeda T, Sakata R. Fetal mesenchymal stem cells ameliorate acute lung injury in a rat cardiopulmonary bypass model. J Thorac Cardiovasc Surg 2016; 153:726-734. [PMID: 27838010 DOI: 10.1016/j.jtcvs.2016.10.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Revised: 09/20/2016] [Accepted: 10/07/2016] [Indexed: 02/07/2023]
Abstract
BACKGROUND Systemic inflammation after prolonged cardiopulmonary bypass (CPB) can cause serious multiorgan system dysfunction. Mesenchymal stem cells (MSCs) are reported to reduce inflammation and attenuate immune response. We have focused on fetal membrane (FM) as a source to provide a large number of MSCs (FM-MSCs). Allogeneic administration of FM-MSCs has been reported to mitigate autoimmune myocarditis or glomerulonephritis. The aim of this study was to investigate whether allogeneic FM-MSCs attenuate systemic inflammatory responses and lung injury in a rat CPB model. METHODS Male Lewis rats (major histocompatibility complex haplotype: RT-1l) were divided randomly into 3 groups (n = 7 each): cannulation alone (sham group), CPB alone (control group), and CPB + MSC (MSC group). An experimental rat CPB model was established, and CPB was maintained for 30 minutes. In the MSC group, MSCs (1 × 106 cells) derived from the FM of ACI rats with a different major histocompatibility complex haplotype (RT-1a) were administrated intravenously before CPB initiation. RESULTS Serum concentrations of tumor necrosis factor-α, interleukin-6, and interleukin-1β in the MSC group were significantly lower compared with the control group after CPB. Similarly, mRNA expression of proinflammatory cytokines in the lung was lower in the MSC group. Allogeneic administration of FM-MSCs remarkably decreased the lung injury score, protected alveolar structure, inhibited neutrophil infiltration to the lung interstitium, and stimulated cytoprotective cytokine production in the lung. CONCLUSIONS Allogeneic transplantation of FM-MSCs may be a potent strategy to prevent CPB-induced systemic inflammation and acute lung injury by suppressing the expression of inflammatory cytokines and promoting protective factors in the lung.
Collapse
Affiliation(s)
- Tomofumi Taki
- Department of Cardiovascular Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hidetoshi Masumoto
- Department of Cardiovascular Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan.
| | - Masaki Funamoto
- Department of Cardiovascular Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Kenji Minakata
- Department of Cardiovascular Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Kazuhiro Yamazaki
- Department of Cardiovascular Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Tadashi Ikeda
- Department of Cardiovascular Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Ryuzo Sakata
- Department of Cardiovascular Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
20
|
Cruz FF, Weiss DJ, Rocco PRM. Prospects and progress in cell therapy for acute respiratory distress syndrome. Expert Opin Biol Ther 2016; 16:1353-1360. [DOI: 10.1080/14712598.2016.1218845] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
21
|
Cruz FF, Borg ZD, Goodwin M, Coffey AL, Wagner DE, Rocco PRM, Weiss DJ. CD11b+ and Sca-1+ Cells Exert the Main Beneficial Effects of Systemically Administered Bone Marrow-Derived Mononuclear Cells in a Murine Model of Mixed Th2/Th17 Allergic Airway Inflammation. Stem Cells Transl Med 2016; 5:488-99. [PMID: 26933041 PMCID: PMC4798733 DOI: 10.5966/sctm.2015-0141] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 11/02/2015] [Indexed: 02/06/2023] Open
Abstract
A murine model of severe clinical asthma was used to study which bone marrow-derived mononuclear cells (BMDMCs) are responsible for ameliorating airway hyperresponsiveness and lung inflammation. BMDMCs depleted of either CD11b-positive cells (monocytes, macrophages, dendritic cells) or Sca-1-positive cells (bone marrow-derived mesenchymal stromal cells) were unable to ameliorate these conditions in this model. Depletion of the other cell types did not diminish the ameliorating effects of BMDMC administration. Systemic administration of bone marrow-derived mononuclear cells (BMDMCs) or bone marrow-derived mesenchymal stromal cells (MSCs) reduces inflammation and airway hyperresponsiveness (AHR) in a murine model of Th2-mediated eosinophilic allergic airway inflammation. However, since BMDMCs are a heterogeneous population that includes MSCs, it is unclear whether the MSCs alone are responsible for the BMDMC effects. To determine which BMDMC population(s) is responsible for ameliorating AHR and lung inflammation in a model of mixed Th2-eosinophilic and Th17-neutrophilic allergic airway inflammation, reminiscent of severe clinical asthma, BMDMCs obtained from normal C57Bl/6 mice were serially depleted of CD45, CD34, CD11b, CD3, CD19, CD31, or Sca-1 positive cells. The different resulting cell populations were then assessed for ability to reduce lung inflammation and AHR in mixed Th2/Th17 allergic airway inflammation induced by mucosal sensitization to and challenge with Aspergillus hyphal extract (AHE) in syngeneic C56Bl/6 mice. BMDMCs depleted of either CD11b-positive (CD11b+) or Sca-1-positive (Sca-1+) cells were unable to ameliorate AHR or lung inflammation in this model. Depletion of the other cell types did not diminish the ameliorating effects of BMDMC administration. In conclusion, in the current model of allergic inflammation, CD11b+ cells (monocytes, macrophages, dendritic cells) and Sca-1+ cells (MSCs) are responsible for the beneficial effects of BMDMCs. Significance This study shows that bone marrow-derived mononuclear cells (BMDMCs) are as effective as bone marrow-derived mesenchymal stromal cells (MSCs) in ameliorating experimental asthma. It also demonstrates that not only MSCs present in the pool of BMDMCs are responsible for BMDMCs’ beneficial effects but also monocytes, which are the most important cell population to trigger these effects. All of this is in the setting of a clinically relevant model of severe allergic airways inflammation and thus provides further support for potential clinical use of cell therapy using MSCs, BMDMCs, and also adult cells such as monocytes in patients with severe asthma.
Collapse
Affiliation(s)
- Fernanda F Cruz
- Division of Pulmonary Disease and Critical Care Medicine, Department of Medicine, University of Vermont, Burlington, Vermont, USA Laboratory of Pulmonary Investigation, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Zachary D Borg
- Division of Pulmonary Disease and Critical Care Medicine, Department of Medicine, University of Vermont, Burlington, Vermont, USA
| | - Meagan Goodwin
- Division of Pulmonary Disease and Critical Care Medicine, Department of Medicine, University of Vermont, Burlington, Vermont, USA
| | - Amy L Coffey
- Division of Pulmonary Disease and Critical Care Medicine, Department of Medicine, University of Vermont, Burlington, Vermont, USA
| | - Darcy E Wagner
- Division of Pulmonary Disease and Critical Care Medicine, Department of Medicine, University of Vermont, Burlington, Vermont, USA
| | - Patricia R M Rocco
- Laboratory of Pulmonary Investigation, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Daniel J Weiss
- Division of Pulmonary Disease and Critical Care Medicine, Department of Medicine, University of Vermont, Burlington, Vermont, USA
| |
Collapse
|
22
|
Expanded endothelial progenitor cells mitigate lung injury in septic mice. Stem Cell Res Ther 2015; 6:230. [PMID: 26611795 PMCID: PMC4660838 DOI: 10.1186/s13287-015-0226-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Revised: 10/25/2015] [Accepted: 11/06/2015] [Indexed: 02/04/2023] Open
Abstract
Endothelial progenitor cells (EPCs) improve survival and reduce organ failure in cecal ligation and puncture-induced sepsis; however, expanded EPCs may represent an even better approach for vascular repair. To date, no study has compared the effects of non-expanded EPCs (EPC-NEXP) with those of expanded EPCs (EPC-EXP) and mesenchymal stromal cells of human (MSC-HUMAN) and mouse (MSC-MICE) origin in experimental sepsis. One day after cecal ligation and puncture sepsis induction, BALB/c mice were randomized to receive saline, EPC-EXP, EPC-NEXP, MSC-HUMAN or MSC-MICE (1 × 105) intravenously. EPC-EXP, EPC-NEXP, MSC-HUMAN, and MSC-MICE displayed differences in phenotypic characterization. On days 1 and 3, cecal ligation and puncture mice showed decreased survival rate, and increased elastance, diffuse alveolar damage, and levels of interleukin (IL)-1β, IL-6, IL-10, tumor necrosis factor-α, vascular endothelial growth factor, and platelet-derived growth factor in lung tissue. EPC-EXP and MSC-HUMAN had reduced elastance, diffuse alveolar damage, and platelet-derived growth factor compared to no-cell treatment. Tumor necrosis factor-α levels decreased in the EPC-EXP, MSC-HUMAN, and MSC-MICE groups. IL-1β levels decreased in the EPC-EXP group, while IL-10 decreased in the MSC-MICE. IL-6 levels decreased both in the EPC-EXP and MSC-MICE groups. Vascular endothelial growth factor levels were reduced regardless of therapy. In conclusion, EPC-EXP and MSC-HUMAN yielded better lung function and reduced histologic damage in septic mice.
Collapse
|
23
|
Gavin KM, Gutman JA, Kohrt WM, Wei Q, Shea KL, Miller HL, Sullivan TM, Erickson PF, Helm KM, Acosta AS, Childs CR, Musselwhite E, Varella-Garcia M, Kelly K, Majka SM, Klemm DJ. De novo generation of adipocytes from circulating progenitor cells in mouse and human adipose tissue. FASEB J 2015; 30:1096-108. [PMID: 26581599 DOI: 10.1096/fj.15-278994] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 11/02/2015] [Indexed: 12/21/2022]
Abstract
White adipocytes in adults are typically derived from tissue resident mesenchymal progenitors. The recent identification of de novo production of adipocytes from bone marrow progenitor-derived cells in mice challenges this paradigm and indicates an alternative lineage specification that adipocytes exist. We hypothesized that alternative lineage specification of white adipocytes is also present in human adipose tissue. Bone marrow from transgenic mice in which luciferase expression is governed by the adipocyte-restricted adiponectin gene promoter was adoptively transferred to wild-type recipient mice. Light emission was quantitated in recipients by in vivo imaging and direct enzyme assay. Adipocytes were also obtained from human recipients of hematopoietic stem cell transplantation. DNA was isolated, and microsatellite polymorphisms were exploited to quantify donor/recipient chimerism. Luciferase emission was detected from major fat depots of transplanted mice. No light emission was observed from intestines, liver, or lungs. Up to 35% of adipocytes in humans were generated from donor marrow cells in the absence of cell fusion. Nontransplanted mice and stromal-vascular fraction samples were used as negative and positive controls for the mouse and human experiments, respectively. This study provides evidence for a nontissue resident origin of an adipocyte subpopulation in both mice and humans.
Collapse
Affiliation(s)
- Kathleen M Gavin
- *Division of Geriatric Medicine, Division of Medical Oncology, and Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, School of Medicine, Flow Cytometry Shared Resource, Molecular Pathology/Cytogenetics Shared Resource, University of Colorado Cancer Center, Charles C. Gates Center for Regenerative Medicine and Stem Cell Biology, and Colorado Obesity Research Initiative, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA; Molecular Diagnostic Laboratory, Children's Hospital Colorado, Aurora, Colorado, USA; and Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, and **Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Jonathan A Gutman
- *Division of Geriatric Medicine, Division of Medical Oncology, and Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, School of Medicine, Flow Cytometry Shared Resource, Molecular Pathology/Cytogenetics Shared Resource, University of Colorado Cancer Center, Charles C. Gates Center for Regenerative Medicine and Stem Cell Biology, and Colorado Obesity Research Initiative, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA; Molecular Diagnostic Laboratory, Children's Hospital Colorado, Aurora, Colorado, USA; and Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, and **Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Wendy M Kohrt
- *Division of Geriatric Medicine, Division of Medical Oncology, and Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, School of Medicine, Flow Cytometry Shared Resource, Molecular Pathology/Cytogenetics Shared Resource, University of Colorado Cancer Center, Charles C. Gates Center for Regenerative Medicine and Stem Cell Biology, and Colorado Obesity Research Initiative, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA; Molecular Diagnostic Laboratory, Children's Hospital Colorado, Aurora, Colorado, USA; and Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, and **Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Qi Wei
- *Division of Geriatric Medicine, Division of Medical Oncology, and Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, School of Medicine, Flow Cytometry Shared Resource, Molecular Pathology/Cytogenetics Shared Resource, University of Colorado Cancer Center, Charles C. Gates Center for Regenerative Medicine and Stem Cell Biology, and Colorado Obesity Research Initiative, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA; Molecular Diagnostic Laboratory, Children's Hospital Colorado, Aurora, Colorado, USA; and Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, and **Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Karen L Shea
- *Division of Geriatric Medicine, Division of Medical Oncology, and Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, School of Medicine, Flow Cytometry Shared Resource, Molecular Pathology/Cytogenetics Shared Resource, University of Colorado Cancer Center, Charles C. Gates Center for Regenerative Medicine and Stem Cell Biology, and Colorado Obesity Research Initiative, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA; Molecular Diagnostic Laboratory, Children's Hospital Colorado, Aurora, Colorado, USA; and Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, and **Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Heidi L Miller
- *Division of Geriatric Medicine, Division of Medical Oncology, and Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, School of Medicine, Flow Cytometry Shared Resource, Molecular Pathology/Cytogenetics Shared Resource, University of Colorado Cancer Center, Charles C. Gates Center for Regenerative Medicine and Stem Cell Biology, and Colorado Obesity Research Initiative, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA; Molecular Diagnostic Laboratory, Children's Hospital Colorado, Aurora, Colorado, USA; and Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, and **Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Timothy M Sullivan
- *Division of Geriatric Medicine, Division of Medical Oncology, and Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, School of Medicine, Flow Cytometry Shared Resource, Molecular Pathology/Cytogenetics Shared Resource, University of Colorado Cancer Center, Charles C. Gates Center for Regenerative Medicine and Stem Cell Biology, and Colorado Obesity Research Initiative, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA; Molecular Diagnostic Laboratory, Children's Hospital Colorado, Aurora, Colorado, USA; and Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, and **Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Paul F Erickson
- *Division of Geriatric Medicine, Division of Medical Oncology, and Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, School of Medicine, Flow Cytometry Shared Resource, Molecular Pathology/Cytogenetics Shared Resource, University of Colorado Cancer Center, Charles C. Gates Center for Regenerative Medicine and Stem Cell Biology, and Colorado Obesity Research Initiative, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA; Molecular Diagnostic Laboratory, Children's Hospital Colorado, Aurora, Colorado, USA; and Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, and **Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Karen M Helm
- *Division of Geriatric Medicine, Division of Medical Oncology, and Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, School of Medicine, Flow Cytometry Shared Resource, Molecular Pathology/Cytogenetics Shared Resource, University of Colorado Cancer Center, Charles C. Gates Center for Regenerative Medicine and Stem Cell Biology, and Colorado Obesity Research Initiative, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA; Molecular Diagnostic Laboratory, Children's Hospital Colorado, Aurora, Colorado, USA; and Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, and **Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Alistaire S Acosta
- *Division of Geriatric Medicine, Division of Medical Oncology, and Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, School of Medicine, Flow Cytometry Shared Resource, Molecular Pathology/Cytogenetics Shared Resource, University of Colorado Cancer Center, Charles C. Gates Center for Regenerative Medicine and Stem Cell Biology, and Colorado Obesity Research Initiative, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA; Molecular Diagnostic Laboratory, Children's Hospital Colorado, Aurora, Colorado, USA; and Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, and **Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Christine R Childs
- *Division of Geriatric Medicine, Division of Medical Oncology, and Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, School of Medicine, Flow Cytometry Shared Resource, Molecular Pathology/Cytogenetics Shared Resource, University of Colorado Cancer Center, Charles C. Gates Center for Regenerative Medicine and Stem Cell Biology, and Colorado Obesity Research Initiative, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA; Molecular Diagnostic Laboratory, Children's Hospital Colorado, Aurora, Colorado, USA; and Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, and **Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Evelyn Musselwhite
- *Division of Geriatric Medicine, Division of Medical Oncology, and Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, School of Medicine, Flow Cytometry Shared Resource, Molecular Pathology/Cytogenetics Shared Resource, University of Colorado Cancer Center, Charles C. Gates Center for Regenerative Medicine and Stem Cell Biology, and Colorado Obesity Research Initiative, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA; Molecular Diagnostic Laboratory, Children's Hospital Colorado, Aurora, Colorado, USA; and Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, and **Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Marileila Varella-Garcia
- *Division of Geriatric Medicine, Division of Medical Oncology, and Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, School of Medicine, Flow Cytometry Shared Resource, Molecular Pathology/Cytogenetics Shared Resource, University of Colorado Cancer Center, Charles C. Gates Center for Regenerative Medicine and Stem Cell Biology, and Colorado Obesity Research Initiative, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA; Molecular Diagnostic Laboratory, Children's Hospital Colorado, Aurora, Colorado, USA; and Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, and **Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Kimberly Kelly
- *Division of Geriatric Medicine, Division of Medical Oncology, and Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, School of Medicine, Flow Cytometry Shared Resource, Molecular Pathology/Cytogenetics Shared Resource, University of Colorado Cancer Center, Charles C. Gates Center for Regenerative Medicine and Stem Cell Biology, and Colorado Obesity Research Initiative, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA; Molecular Diagnostic Laboratory, Children's Hospital Colorado, Aurora, Colorado, USA; and Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, and **Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Susan M Majka
- *Division of Geriatric Medicine, Division of Medical Oncology, and Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, School of Medicine, Flow Cytometry Shared Resource, Molecular Pathology/Cytogenetics Shared Resource, University of Colorado Cancer Center, Charles C. Gates Center for Regenerative Medicine and Stem Cell Biology, and Colorado Obesity Research Initiative, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA; Molecular Diagnostic Laboratory, Children's Hospital Colorado, Aurora, Colorado, USA; and Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, and **Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Dwight J Klemm
- *Division of Geriatric Medicine, Division of Medical Oncology, and Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, School of Medicine, Flow Cytometry Shared Resource, Molecular Pathology/Cytogenetics Shared Resource, University of Colorado Cancer Center, Charles C. Gates Center for Regenerative Medicine and Stem Cell Biology, and Colorado Obesity Research Initiative, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA; Molecular Diagnostic Laboratory, Children's Hospital Colorado, Aurora, Colorado, USA; and Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, and **Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| |
Collapse
|
24
|
Abstract
PURPOSE OF REVIEW The prognosis of patients with respiratory failure in the ICU remains poor, while current therapeutic approaches are aimed at minimizing ventilator-induced lung injury. Stem cell-based therapies have the potential to transform respiratory failure treatment by achieving lung repair. The purpose of this article is to critically review the large body of clinical and experimental work performed with respect to the use of stem/progenitor cells in respiratory failure, and to discuss current challenges and future directions. RECENT FINDINGS Since the initial report of cell therapy for lung injury in 2005, numerous preclinical and clinical studies have been performed that support the ability of various stem cell populations to improve physiologic lung function and reduce inflammation in both infective and sterile acute respiratory distress syndrome. Nevertheless, many important issues (e.g., mechanism of action, long-term engraftment, optimal cell type, dose, route of administration) remain to be resolved. SUMMARY Cell-based therapeutics hold promise, particularly for acute respiratory distress syndrome, and early preclinical testing has been encouraging. To advance clinical testing of cell therapies in respiratory failure, and to help ensure that this approach will facilitate bench-to-bedside and bedside-to-bench discoveries, parallel paths of basic and clinical research are needed, including measures of cell therapy effectiveness in vivo and in vitro.
Collapse
|
25
|
Souza MC, Silva JD, Pádua TA, Torres ND, Antunes MA, Xisto DG, Abreu TP, Capelozzi VL, Morales MM, Sá Pinheiro AA, Caruso-Neves C, Henriques MG, Rocco PRM. Mesenchymal stromal cell therapy attenuated lung and kidney injury but not brain damage in experimental cerebral malaria. Stem Cell Res Ther 2015; 6:102. [PMID: 25998168 PMCID: PMC4462088 DOI: 10.1186/s13287-015-0093-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Revised: 04/01/2015] [Accepted: 05/11/2015] [Indexed: 12/13/2022] Open
Abstract
Introduction Malaria is the most relevant parasitic disease worldwide, and still accounts for 1 million deaths each year. Since current antimalarial drugs are unable to prevent death in severe cases, new therapeutic strategies have been developed. Mesenchymal stromal cells (MSC) confer host resistance against malaria; however, thus far, no study has evaluated the therapeutic effects of MSC therapy on brain and distal organ damage in experimental cerebral malaria. Methods Forty C57BL/6 mice were injected intraperitoneally with 5 × 106Plasmodium berghei-infected erythrocytes or saline. After 24 h, mice received saline or bone marrow (BM)-derived MSC (1x105) intravenously and were housed individually in metabolic cages. After 4 days, lung and kidney morphofunction; cerebrum, spleen, and liver histology; and markers associated with inflammation, fibrogenesis, and epithelial and endothelial cell damage in lung tissue were analyzed. Results In P. berghei-infected mice, BM-MSCs: 1) reduced parasitemia and mortality; 2) increased phagocytic neutrophil content in brain, even though BM-MSCs did not affect the inflammatory process; 3) decreased malaria pigment detection in spleen, liver, and kidney; 4) reduced hepatocyte derangement, with an increased number of Kupffer cells; 5) decreased kidney damage, without effecting significant changes in serum creatinine levels or urinary flow; and 6) reduced neutrophil infiltration, interstitial edema, number of myofibroblasts within interstitial tissue, and collagen deposition in lungs, resulting in decreased lung static elastance. These morphological and functional changes were not associated with changes in levels of tumor necrosis factor-α, keratinocyte-derived chemokine (KC, a mouse analog of interleukin-8), or interferon-γ, which remained increased and similar to those of P. berghei animals treated with saline. BM-MSCs increased hepatocyte growth factor but decreased VEGF in the P. berghei group. Conclusions BM-MSC treatment increased survival and reduced parasitemia and malaria pigment accumulation in spleen, liver, kidney, and lung, but not in brain. The two main organs associated with worse prognosis in malaria, lung and kidney, sustained less histological damage after BM-MSC therapy, with a more pronounced improvement in lung function.
Collapse
Affiliation(s)
- Mariana C Souza
- Laboratory of Applied Pharmacology, Farmanguinhos, Oswaldo Cruz Foundation, Av Brasil, 4365, Manguinhos, CEP-21040-900, Rio de Janeiro, RJ, Brazil.
| | - Johnatas D Silva
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Av Carlos Chagas Filho, 373 Bloco G, Cidade Universitária, CEP-21941-902, Rio de Janeiro, RJ, Brazil.
| | - Tatiana A Pádua
- Laboratory of Applied Pharmacology, Farmanguinhos, Oswaldo Cruz Foundation, Av Brasil, 4365, Manguinhos, CEP-21040-900, Rio de Janeiro, RJ, Brazil.
| | - Natália D Torres
- Laboratory of Applied Pharmacology, Farmanguinhos, Oswaldo Cruz Foundation, Av Brasil, 4365, Manguinhos, CEP-21040-900, Rio de Janeiro, RJ, Brazil.
| | - Mariana A Antunes
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Av Carlos Chagas Filho, 373 Bloco G, Cidade Universitária, CEP-21941-902, Rio de Janeiro, RJ, Brazil.
| | - Debora G Xisto
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Av Carlos Chagas Filho, 373 Bloco G, Cidade Universitária, CEP-21941-902, Rio de Janeiro, RJ, Brazil.
| | - Thiago P Abreu
- Laboratory of Biochemistry and Cellular Signaling, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Av Carlos Chagas Filho, 373 Bloco G, Cidade Universitária, CEP-21941-902, Rio de Janeiro, RJ, Brazil.
| | - Vera L Capelozzi
- Department of Pathology, Faculty of Medicine, University of São Paulo, Av. Dr. Arnaldo, 455, Cerqueira César, CEP-01246903, São Paulo, SP, Brazil.
| | - Marcelo M Morales
- Laboratory of Cellular and Molecular Physiology, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Av Carlos Chagas Filho, 373 Bloco G, Cidade Universitária, CEP-21941-902, Rio de Janeiro, RJ, Brazil.
| | - Ana A Sá Pinheiro
- Laboratory of Biochemistry and Cellular Signaling, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Av Carlos Chagas Filho, 373 Bloco G, Cidade Universitária, CEP-21941-902, Rio de Janeiro, RJ, Brazil.
| | - Celso Caruso-Neves
- Laboratory of Biochemistry and Cellular Signaling, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Av Carlos Chagas Filho, 373 Bloco G, Cidade Universitária, CEP-21941-902, Rio de Janeiro, RJ, Brazil.
| | - Maria G Henriques
- Laboratory of Applied Pharmacology, Farmanguinhos, Oswaldo Cruz Foundation, Av Brasil, 4365, Manguinhos, CEP-21040-900, Rio de Janeiro, RJ, Brazil. .,National Institute for Science and Technology on Innovation on Neglected Diseases (INCT/IDN), Center for Technological Development in Health (CDTS), Oswaldo Cruz Foundation (Fiocruz), Av Brasil, 4365, Manguinhos, CEP-21040-900, Rio de Janeiro, RJ, Brazil.
| | - Patricia R M Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Av Carlos Chagas Filho, 373 Bloco G, Cidade Universitária, CEP-21941-902, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
26
|
Liu L, He H, Liu A, Xu J, Han J, Chen Q, Hu S, Xu X, Huang Y, Guo F, Yang Y, Qiu H. Therapeutic Effects of Bone Marrow-Derived Mesenchymal Stem Cells in Models of Pulmonary and Extrapulmonary Acute Lung Injury. Cell Transplant 2015; 24:2629-42. [PMID: 25695285 DOI: 10.3727/096368915x687499] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Bone marrow-derived mesenchymal stem cells (MSCs) offer a promising therapy for acute lung injury (ALI). However, whether the same MSC treatments possess similar potential for different ALI models is not fully clear. The present study evaluated the distribution and therapeutic effects of intravenous MSC administration for the treatment of intratracheal lipopolysaccharide (LPS)-induced intrapulmonary ALI and intravenous LPS/zymosan-induced extrapulmonary ALI, matched with lung injury severity, at 30 min and 1, 3, and 7 days. We found that MSC transplantation attenuated lung injury and inhibited lung inflammation in both ALI models. The benefits of MSCs were more significant in the intrapulmonary ALI mice. In vivo and ex vivo fluorescence imaging showed that MSCs primarily homed into the lung. However, more MSCs were recruited into the lungs of the intrapulmonary ALI mice than those of the extrapulmonary ALI mice over the time course. A few MSCs were also detected in the liver and spleen at days 3 and 7. In addition, the two ALI models showed different extrapulmonary organ dysfunction. A lower percentage of cell apoptosis and SDF-1α levels was found in the liver and spleen of the intrapulmonary ALI mice than in those of the extrapulmonary ALI mice. These results suggested that the two ALI models were accompanied with different degrees of extrapulmonary organ damage, which resulted in differences in the trafficking and accumulation of MSCs to the injured lung and consequently accounted for different therapeutic effects of MSCs for lung repair in the two ALI models. These data suggest that intravenous administration of MSCs has a greater potential for the treatment of intrapulmonary ALI than extrapulmonary ALI matched with lung injury severity; these differences were due to more recruitment of MSCs in the lungs of intrapulmonary ALI mice than those of extrapulmonary ALI mice. This finding may contribute to the clinical use of MSCs for the treatment of ALI.
Collapse
Affiliation(s)
- Ling Liu
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, P. R. China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Hayes M, Masterson C, Devaney J, Barry F, Elliman S, O’Brien T, O’ Toole D, Curley GF, Laffey JG, Lee JW, Rocco PR, Pelosi P. Mesenchymal stem cell therapy for acute respiratory distress syndrome: a light at the end of the tunnel? Anesthesiology 2015; 122:238-40. [PMID: 25478942 PMCID: PMC4301977 DOI: 10.1097/aln.0000000000000546] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Jae-Woo Lee
- Department of Anesthesiology, University of California San Francisco, San Francisco, California
| | - Patricia R.M. Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, RJ, Brazil
| | - Paolo Pelosi
- Department of Surgical Sciences and Integrated Diagnostics, IRCCS San Martino IST, University of Genoa, Genoa, Italy
| |
Collapse
|
28
|
Mäkelä T, Takalo R, Arvola O, Haapanen H, Yannopoulos F, Blanco R, Ahvenjärvi L, Kiviluoma K, Kerkelä E, Nystedt J, Juvonen T, Lehenkari P. Safety and biodistribution study of bone marrow-derived mesenchymal stromal cells and mononuclear cells and the impact of the administration route in an intact porcine model. Cytotherapy 2015; 17:392-402. [PMID: 25601140 DOI: 10.1016/j.jcyt.2014.12.004] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Revised: 12/02/2014] [Accepted: 12/10/2014] [Indexed: 12/13/2022]
Abstract
BACKGROUND AIMS Bone marrow mononuclear cells (BM-MNCs) and bone marrow-derived mesenchymal stem stromal cells (BM-MSCs) could have therapeutic potential for numerous conditions, including ischemia-related injury. Cells transplanted intravascularly may become entrapped in the lungs, which potentially decreases their therapeutic effect and increases the risk for embolism. METHODS Twelve pigs were divided into groups of 3 and received (99m)Tc- hydroxymethyl-propylene-amine-oxime-labeled autologous BM-MNCs or allogeneic BM-MSCs by either intravenous (IV) or intra-arterial (IA) transplantation. A whole body scan and single photon emission computed tomography/computed tomography (SPECT/CT) were performed 8 h later, and tissue biopsies were collected for gamma counting. A helical CT scan was also performed on 4 pigs to detect possible pulmonary embolism, 2 after IV BM-MSC injection and 2 after saline injection. RESULTS The transplantation route had a greater impact on the biodistribution of the BM-MSCs than the BM-MNCs. The BM-MNCs accumulated in the spleen and bones, irrespective of the administration route. The BM-MSCs had relatively higher uptake in the kidneys. The IA transplantation decreased the deposition of BM-MSCs in the lungs and increased uptake in other organs, especially in the liver. Lung atelectases were frequent due to mechanical ventilation and attracted transplanted cells. CT did not reveal any pulmonary embolism. CONCLUSIONS Both administration routes were found to be safe, but iatrogenic atelectasis might be an issue when cells accumulate in the lungs. The IA administration is effective in avoiding pulmonary entrapment of BM-MSCs. The cell type and administration method both have a major impact on the acute homing.
Collapse
Affiliation(s)
- Tuomas Mäkelä
- Department of Surgery, Oulu University Hospital, Oulu, Finland.
| | - Reijo Takalo
- Department of Diagnostic Radiology, Oulu University Hospital, Oulu, Finland
| | - Oiva Arvola
- Department of Surgery, Oulu University Hospital, Oulu, Finland
| | - Henri Haapanen
- Department of Surgery, Oulu University Hospital, Oulu, Finland
| | | | - Roberto Blanco
- Department of Diagnostic Radiology, Oulu University Hospital, Oulu, Finland
| | - Lauri Ahvenjärvi
- Department of Diagnostic Radiology, Oulu University Hospital, Oulu, Finland
| | - Kai Kiviluoma
- Department of Anaesthesiology, Oulu University Hospital, Oulu, Finland
| | - Erja Kerkelä
- Finnish Red Cross Blood Service, Research and Cell Therapy Services, Helsinki, Finland
| | - Johanna Nystedt
- Finnish Red Cross Blood Service, Research and Cell Therapy Services, Helsinki, Finland
| | - Tatu Juvonen
- Department of Surgery, Oulu University Hospital, Oulu, Finland
| | - Petri Lehenkari
- Department of Anatomy and Cell Biology, Medical Research Center, University of Oulu, Oulu, Finland
| |
Collapse
|
29
|
Weiss DJ. Concise review: current status of stem cells and regenerative medicine in lung biology and diseases. Stem Cells 2014; 32:16-25. [PMID: 23959715 DOI: 10.1002/stem.1506] [Citation(s) in RCA: 124] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2013] [Accepted: 07/24/2013] [Indexed: 12/29/2022]
Abstract
Lung diseases remain a significant and devastating cause of morbidity and mortality worldwide. In contrast to many other major diseases, lung diseases notably chronic obstructive pulmonary diseases (COPDs), including both asthma and emphysema, are increasing in prevalence and COPD is expected to become the third leading cause of disease mortality worldwide by 2020. New therapeutic options are desperately needed. A rapidly growing number of investigations of stem cells and cell therapies in lung biology and diseases as well as in ex vivo lung bioengineering have offered exciting new avenues for advancing knowledge of lung biology as well as providing novel potential therapeutic approaches for lung diseases. These initial observations have led to a growing exploration of endothelial progenitor cells and mesenchymal stem (stromal) cells in clinical trials of pulmonary hypertension and COPD with other clinical investigations planned. Ex vivo bioengineering of the trachea, larynx, diaphragm, and the lung itself with both biosynthetic constructs as well as decellularized tissues have been used to explore engineering both airway and vascular systems of the lung. Lung is thus a ripe organ for a variety of cell therapy and regenerative medicine approaches. Current state-of-the-art progress for each of the above areas will be presented as will discussion of current considerations for cell therapy-based clinical trials in lung diseases.
Collapse
Affiliation(s)
- Daniel J Weiss
- Department of Medicine, University of Vermont College of Medicine, Burlington, Vermont, USA
| |
Collapse
|
30
|
Abstract
PURPOSE OF REVIEW Acute respiratory distress syndrome (ARDS) is a multifaceted lung disease with no current effective therapy. Many clinical trials using conventional pharmacologic therapies have failed, suggesting the need to examine alternative approaches. Thus, attention has focused on the therapeutic potential of cell-based therapies for ARDS, with promising results demonstrated in relevant preclinical disease models. We review data concerning the therapeutic promise of cell-based therapies for ARDS. RECENT FINDINGS Recent experimental studies provide further evidence for the potential of cell-based therapies in ARDS. A number of cell types, particularly mesenchymal stem/stromal cells (MSCs), bone marrow-derived mononuclear cells, endothelial progenitor cells, and embryonic stem cells have been demonstrated to reduce mortality and modulate the inflammatory and remodeling processes in relevant preclinical ARDS models. Multiple insights have emerged in regard to the mechanisms by which cell therapies - particularly MSCs - exert their effects, with evidence supporting direct cell-mediated and paracrine-mediated mechanisms of action. Diverse paracrine mechanisms exist, including the release of cytokines, growth factors (such as keratinocyte growth factor), and antimicrobial peptides, and transfer of cellular contents such as peptides, nucleic acids, and mitochondria via either microvesicular or direct cell-cell contact-mediated transfer. SUMMARY Cell-based therapies offer considerable promise for the treatment of ARDS. While MSC-based therapies are being rapidly advanced toward clinical testing, clear therapeutic potential exists for other cell types for ARDS. A greater understanding of current knowledge gaps should further enhance the therapeutic potential of cell-based therapies for ARDS.
Collapse
|
31
|
Pelosi P, Sutherasan Y. Bone marrow-derived mononuclear cell therapy in sepsis-induced acute respiratory distress syndrome: different insults, different effects! Stem Cell Res Ther 2014; 4:143. [PMID: 24279925 PMCID: PMC4055016 DOI: 10.1186/scrt354] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) is one of the devastating sequelae of sepsis, and so far no specific promising pharmacotherapies have been proven to decrease mortality from it. Stem cell therapy is a novel therapy that can promote earlier and more effective remodeling and repair of damaged lung tissue. Bone marrow-derived mononuclear cells are an alternative stem cell therapy that is safely and easily administered on the day of harvesting and yields benefits in acute disease processes like ARDS. In a recent issue of Stem Cell Research and Therapy, Maron-Gutierrez and colleagues demonstrated that the effects of transfused bone marrow-derived mononuclear cells on lung mechanics, inflammation and mortality might be different in different septic ARDS models due to different insults.
Collapse
|
32
|
Clinical and biological heterogeneity in acute respiratory distress syndrome: direct versus indirect lung injury. Clin Chest Med 2014; 35:639-53. [PMID: 25453415 DOI: 10.1016/j.ccm.2014.08.004] [Citation(s) in RCA: 102] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The acute respiratory distress syndrome (ARDS) is a heterogeneous group of illnesses affecting the pulmonary parenchyma with acute onset bilateral inflammatory pulmonary infiltrates with associated hypoxemia. ARDS occurs after 2 major types of pulmonary injury: direct lung injury affecting the lung epithelium or indirect lung injury disrupting the vascular endothelium. Greater understanding of the differences between direct and indirect lung injury may refine the classification of patients with ARDS and lead to development of new therapeutics targeted at specific subpopulations of patients with ARDS.
Collapse
|
33
|
Silva JD, Paredes BD, Araújo IM, Lopes-Pacheco M, Oliveira MV, Suhett GD, Faccioli LAP, Assis E, Castro-Faria-Neto HC, Goldenberg RCS, Capelozzi VL, Morales MM, Pelosi P, Xisto DG, Rocco PRM. Effects of bone marrow-derived mononuclear cells from healthy or acute respiratory distress syndrome donors on recipient lung-injured mice. Crit Care Med 2014; 42:e510-24. [PMID: 24633189 DOI: 10.1097/ccm.0000000000000296] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
OBJECTIVE The advantage of using autologous bone marrow-derived mononuclear cells to treat acute respiratory distress syndrome patients is to prevent immunological rejection. However, bone marrow-derived mononuclear cells may be altered by different acute respiratory distress syndrome etiologies, resulting in questionable efficacy and thus limited clinical application. We aimed to investigate the effects of bone marrow-derived mononuclear cells obtained from healthy and acute respiratory distress syndrome donors on pulmonary and extrapulmonary acute respiratory distress syndrome. DESIGN Prospective, randomized, controlled experimental study. SETTING University research laboratory. SUBJECTS Two hundred and twenty-five C57BL/6 mice. INTERVENTIONS Acute respiratory distress syndrome was induced by Escherichia coli lipopolysaccharide intratracheally (ARDSp) or intraperitoneally (ARDSexp). Control mice (Healthy) received saline solution intratracheally (Cp) or intraperitoneally (Cexp). After 24 hours, whole bone marrow cells were analyzed in vitro: 1) colony-forming unit-fibroblasts and 2) hematopoietic stem cells, neutrophils, T helper lymphocytes, B lymphocytes, and nonhematopoietic precursors. After cell characterization, all groups received saline or bone marrow-derived mononuclear cells (2 × 10), obtained from Cp, Cexp, ARDSp, and ARDSexp donor mice, IV, on day 1. MEASUREMENTS AND MAIN RESULTS On day 1, in ARDSp, different patterns of colony formation were found, with nonstromal cells (mainly neutrophils) predominating over fibroblastoid colonies. In ARDSexp, irregular colony-forming unit-fibroblasts morphology with dispersed proliferating colonies and a greater number of hematopoietic stem cells were observed. In ARDSp, colony-forming unit-fibroblasts count was higher but not measurable in ARDSexp. In ARDSp, monocytes and T lymphocytes were increased and hematopoietic precursor cells reduced, with no significant changes in ARDSexp. On day 7, bone marrow-derived mononuclear cells improved survival and attenuated changes in lung mechanics, alveolar collapse, inflammation, pulmonary fibrosis, and apoptosis in the lung and distal organs, regardless of donor type. CONCLUSIONS Bone marrow-derived mononuclear cells from ARDSp and ARDSexp donors showed different characteristics but were as effective as cells obtained from healthy donors in reducing inflammation and remodeling, suggesting the utility of autologous transplant of bone marrow-derived mononuclear cells in the clinical setting.
Collapse
Affiliation(s)
- Johnatas D Silva
- 1Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil. 2Laboratory of Cellular and Molecular Cardiology, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil. 3Laboratory of Cellular and Molecular Physiology, Carlos Chagas Filho, Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil. 4Department of Radiology, School of Medicine, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil. 5Laboratory of Immunopharmacology, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil. 6Department of Pathology, School of Medicine, University of São Paulo, São Paulo, Brazil. 7Department of Surgical Sciences and Integrated Diagnostics, IRCCS AOU San Martino-IST, University of Genoa, Genoa, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Stem cells, cell therapies, and bioengineering in lung biology and diseases. Comprehensive review of the recent literature 2010-2012. Ann Am Thorac Soc 2014; 10:S45-97. [PMID: 23869446 DOI: 10.1513/annalsats.201304-090aw] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
A conference, "Stem Cells and Cell Therapies in Lung Biology and Lung Diseases," was held July 25 to 28, 2011 at the University of Vermont to review the current understanding of the role of stem and progenitor cells in lung repair after injury and to review the current status of cell therapy and ex vivo bioengineering approaches for lung diseases. These are rapidly expanding areas of study that provide further insight into and challenge traditional views of mechanisms of lung repair after injury and pathogenesis of several lung diseases. The goals of the conference were to summarize the current state of the field, to discuss and debate current controversies, and to identify future research directions and opportunities for basic and translational research in cell-based therapies for lung diseases. The goal of this article, which accompanies the formal conference report, is to provide a comprehensive review of the published literature in lung regenerative medicine from the last conference report through December 2012.
Collapse
|
35
|
Zhu YG, Hao Q, Monsel A, Feng XM, Lee JW. Adult stem cells for acute lung injury: remaining questions and concerns. Respirology 2014; 18:744-56. [PMID: 23578018 DOI: 10.1111/resp.12093] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Accepted: 04/02/2013] [Indexed: 12/22/2022]
Abstract
Acute lung injury (ALI) or acute respiratory distress syndrome remains a major cause of morbidity and mortality in hospitalized patients. The pathophysiology of ALI involves complex interactions between the inciting event, such as pneumonia, sepsis or aspiration, and the host immune response resulting in lung protein permeability, impaired resolution of pulmonary oedema, an intense inflammatory response in the injured alveolus and hypoxemia. In multiple preclinical studies, adult stem cells have been shown to be therapeutic due to both the ability to mitigate injury and inflammation through paracrine mechanisms and perhaps to regenerate tissue by virtue of their multi-potency. These characteristics have stimulated intensive research efforts to explore the possibility of using stem or progenitor cells for the treatment of lung injury. A variety of stem or progenitor cells have been isolated, characterized and tested experimentally in preclinical animal models of ALI. However, questions remain concerning the optimal dose, route and the adult stem or progenitor cell to use. Here, the current mechanisms underlying the therapeutic effect of stem cells in ALI as well as the questions that will arise as clinical trials for ALI are planned are reviewed.
Collapse
Affiliation(s)
- Ying-Gang Zhu
- Department of Pulmonary Medicine, Huadong Hospital, Fudan University, Shanghai, China
| | | | | | | | | |
Collapse
|
36
|
Effects of Mesenchymal Stem Cell Therapy on the Time Course of Pulmonary Remodeling Depend on the Etiology of Lung Injury in Mice. Crit Care Med 2013; 41:e319-33. [DOI: 10.1097/ccm.0b013e31828a663e] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
37
|
Maron-Gutierrez T, Silva JD, Cruz FF, Alegria S, Xisto DG, Assis EF, Castro-Faria-Neto HC, Dos Santos CC, Morales MM, Rocco PRM. Insult-dependent effect of bone marrow cell therapy on inflammatory response in a murine model of extrapulmonary acute respiratory distress syndrome. Stem Cell Res Ther 2013; 4:123. [PMID: 24406030 PMCID: PMC3856598 DOI: 10.1186/scrt334] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Accepted: 10/03/2013] [Indexed: 02/08/2023] Open
Abstract
Introduction Administration of bone marrow-derived cells produces beneficial effects in experimental extrapulmonary acute respiratory distress syndrome (ARDS). However, there are controversies regarding the effects of timing of cell administration and initial insult severity on inflammatory response. We evaluated the effects of bone marrow-derived mononuclear cells (BMDMC) in two models of extrapulmonary ARDS once lung morphofunctional changes had already been installed. Methods BALB/c mice received lipopolysaccharide (LPS) intraperitoneally (5 mg/kg in 0.5 ml saline) or underwent cecal ligation and puncture (CLP). Control mice received saline intraperitoneally (0.5 ml) or underwent sham surgery. At 24 hours, groups were further randomized to receive saline or BMDMC (2 × 106) intravenously. Lung mechanics, histology, and humoral and cellular parameters of lung inflammation and remodeling were analyzed 1, 3 and 7 days after ARDS induction. Results BMDMC therapy led to improved survival in the CLP group, reduced lung elastance, alveolar collapse, tissue and bronchoalveolar lavage fluid cellularity, collagen fiber content, and interleukin-1β and increased chemokine (keratinocyte-derived chemokine and monocyte chemotactic protein-1) expression in lung tissue regardless of the experimental ARDS model. Intercellular adhesion molecule-1 and vascular cell adhesion molecule-1 expression in lung tissue increased after cell therapy depending on the insult (LPS or CLP). Conclusions BMDMC therapy at day 1 successfully reduced lung inflammation and remodeling, thus contributing to improvement of lung mechanics in both extrapulmonary ARDS models. Nevertheless, the different inflammatory responses induced by LPS and CLP resulted in distinct effects of BMDMC therapy. These data may be useful in the clinical setting, as they suggest that the type of initial insult plays a key role in the outcome of treatment.
Collapse
|
38
|
Liu YY, Li LF, Yang CT, Lu KH, Huang CC, Kao KC, Chiou SH. Suppressing NF-κB and NKRF Pathways by Induced Pluripotent Stem Cell Therapy in Mice with Ventilator-Induced Lung Injury. PLoS One 2013; 8:e66760. [PMID: 23840526 PMCID: PMC3694116 DOI: 10.1371/journal.pone.0066760] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2013] [Accepted: 05/12/2013] [Indexed: 01/14/2023] Open
Abstract
Background High-tidal-volume mechanical ventilation used in patients with acute lung injury (ALI) can induce the release of inflammatory cytokines, as macrophage inflammatory protein-2 (MIP-2), recruitment of neutrophils, and disruption of alveolar epithelial and endothelial barriers. Induced pluripotent stem cells (iPSCs) have been shown to improve ALI in mice, but the mechanisms regulating the interactions between mechanical ventilation and iPSCs are not fully elucidated. Nuclear factor kappa B (NF-κB) and NF-κB repressing factor (NKRF) have been proposed to modulate the neutrophil activation involved in ALI. Thus, we hypothesized intravenous injection of iPSCs or iPSC-derived conditioned medium (iPSC-CM) would decrease high-tidal-volume ventilation-induced neutrophil infiltration, oxidative stress, and MIP-2 production through NF-κB/NKRF pathways. Methods Male C57BL/6 mice, aged between 6 and 8 weeks, weighing between 20 and 25 g, were exposed to high-tidal-volume (30 ml/kg) mechanical ventilation with room air for 1 to 4 h after 5×107 cells/kg mouse iPSCs or iPSC-CM administration. Nonventilated mice were used as control groups. Results High-tidal-volume mechanical ventilation induced the increases of integration of iPSCs into the injured lungs of mice, microvascular permeability, neutrophil infiltration, malondialdehyde, MIP-2 production, and NF-κB and NKRF activation. Lung injury indices including inflammation, lung edema, ultrastructure pathologic changes and functional gas exchange impairment induced by mechanical ventilation were attenuated with administration of iPSCs or iPSC-CM, which was mimicked by pharmacological inhibition of NF-κB activity with SN50 or NKRF expression with NKRF short interfering RNA. Conclusions Our data suggest that iPSC-based therapy attenuates high-tidal-volume mechanical ventilation-induced lung injury, at least partly, through inhibition of NF-κB/NKRF pathways. Notably, the conditioned medium of iPSCs revealed beneficial effects equal to those of iPSCs.
Collapse
Affiliation(s)
- Yung-Yang Liu
- Chest Department, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Clinical Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Li-Fu Li
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Department of Medicine, Chang Gung University, Taoyuan, Taiwan
- Department of Respiratory Therapy, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- * E-mail: (L-FL); (S-HC)
| | - Cheng-Ta Yang
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Department of Medicine, Chang Gung University, Taoyuan, Taiwan
- Department of Respiratory Therapy, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Kai-Hsi Lu
- Graduate Institute of Basic Medicine, Fu Jen Catholic University, New Taipei City, Taiwan
- Department of Medical Research and Education, Cheng-Hsin General Hospital, Taipei, Taiwan
| | - Chung-Chi Huang
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Department of Medicine, Chang Gung University, Taoyuan, Taiwan
- Department of Respiratory Therapy, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Kuo-Chin Kao
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Department of Medicine, Chang Gung University, Taoyuan, Taiwan
- Department of Respiratory Therapy, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Shih-Hwa Chiou
- Institute of Clinical Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan
- Department of Ophthalmology, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Medical Research and Education, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Pharmacology, School of Medicine, National Yang-Ming University, Taipei, Taiwan
- * E-mail: (L-FL); (S-HC)
| |
Collapse
|
39
|
Effects of intratracheal mesenchymal stromal cell therapy during recovery and resolution after ventilator-induced lung injury. Anesthesiology 2013; 118:924-32. [PMID: 23377221 DOI: 10.1097/aln.0b013e318287ba08] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND Mesenchymal stromal cells (MSCs) have been demonstrated to attenuate acute lung injury when delivered by intravenous or intratracheal routes. The authors aimed to determine the efficacy of and mechanism of action of intratracheal MSC therapy and to compare their efficacy in enhancing lung repair after ventilation-induced lung injury with intravenous MSC therapy. METHODS : After induction of anesthesia, rats were orotracheally intubated and subjected to ventilation-induced lung injury (respiratory rate 18(-1) min, P insp 35 cm H2O,) to produce severe lung injury. After recovery, animals were randomized to receive: (1) no therapy, n = 4; (2) intratracheal vehicle (phosphate-buffered saline, 300 µl, n = 8); (3) intratracheal fibroblasts (4 × 10 cells, n = 8); (4) intratracheal MSCs (4 × 10(6) cells, n = 8); (5) intratracheal conditioned medium (300 µl, n = 8); or (6) intravenous MSCs (4 × 10(6) cells, n = 4). The extent of recovery after acute lung injury and the inflammatory response was assessed after 48 h. RESULTS Intratracheal MSC therapy enhanced repair after ventilation-induced lung injury, improving arterial oxygenation (mean ± SD, 146 ± 3.9 vs. 110.8 ± 21.5 mmHg), restoring lung compliance (1.04 ± 0.11 vs. 0.83 ± 0.06 ml · cm H2O(-1)), reducing total lung water, and decreasing lung inflammation and histologic injury compared with control. Intratracheal MSC therapy attenuated alveolar tumor necrosis factor-α (130 ± 43 vs. 488 ± 211 pg · ml(-1)) and interleukin-6 concentrations (138 ± 18 vs. 260 ± 82 pg · ml(-1)). The efficacy of intratracheal MSCs was comparable with intravenous MSC therapy. Intratracheal MSCs seemed to act via a paracine mechanism, with conditioned MSC medium also enhancing lung repair after injury. CONCLUSIONS Intratracheal MSC therapy enhanced recovery after ventilation-induced lung injury via a paracrine mechanism, and was as effective as intravenous MSC therapy.
Collapse
|
40
|
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
|
41
|
Abreu SC, Antunes MA, de Castro JC, de Oliveira MV, Bandeira E, Ornellas DS, Diaz BL, Morales MM, Xisto DG, Rocco PRM. Bone marrow-derived mononuclear cells vs. mesenchymal stromal cells in experimental allergic asthma. Respir Physiol Neurobiol 2013; 187:190-8. [PMID: 23548824 DOI: 10.1016/j.resp.2013.03.014] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Revised: 03/13/2013] [Accepted: 03/25/2013] [Indexed: 01/10/2023]
Abstract
We compared the effects of bone marrow-derived mononuclear cells (BMMCs) and mesenchymal stromal cells (MSCs) on airway inflammation and remodeling and lung mechanics in experimental allergic asthma. C57BL/6 mice were sensitized and challenged with ovalbumin (OVA group). A control group received saline using the same protocol. Twenty-four hours after the last challenge, groups were further randomized into subgroups to receive saline, BMMCs (2×10(6)) or MSCs (1×10(5)) intratracheally. BMMC and MSC administration decreased cell infiltration, bronchoconstriction index, alveolar collapse, collagen fiber content in the alveolar septa, and interleukin (IL)-4, IL-13, transforming growth factor (TGF)-β and vascular endothelial growth factor (VEGF) levels compared to OVA-SAL. Lung function, alveolar collapse, collagen fiber deposition in alveolar septa, and levels of TGF-β and VEGF improved more after BMMC than MSC therapy. In conclusion, intratracheal BMMC and MSC administration effectively modulated inflammation and fibrogenesis in an experimental model of asthma, but BMMCs was associated with greater benefit in terms of reducing levels of fibrogenesis-related growth factors.
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
|
42
|
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
|
43
|
Tejera P, Meyer NJ, Chen F, Feng R, Zhao Y, O'Mahony DS, Li L, Sheu CC, Zhai R, Wang Z, Su L, Bajwa E, Ahasic AM, Clardy PF, Gong MN, Frank AJ, Lanken PN, Thompson BT, Christie JD, Wurfel MM, O'Keefe GE, Christiani DC. Distinct and replicable genetic risk factors for acute respiratory distress syndrome of pulmonary or extrapulmonary origin. J Med Genet 2012; 49:671-80. [PMID: 23048207 DOI: 10.1136/jmedgenet-2012-100972] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND The role of genetics in the development of acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) from direct or indirect lung injury has not been specifically investigated. The aim of this study was to identify genetic variants contributing to ALI/ARDS from pulmonary or extrapulmonary causes. METHODS We conducted a multistage genetic association study. We first performed a large-scale genotyping (50K ITMAT-Broad_CARe Chip) in 1717 critically ill Caucasian patients with either pulmonary or extrapulmonary injury, to identify single nucleotide polymorphisms (SNPs) associated with the development of ARDS from direct or indirect insults to the lung. Identified SNPs (p≤0.0005) were validated in two separated populations (Stage II), with trauma (Population I; n=765) and pneumonia/pulmonary sepsis (Population II; n=838), as causes for ALI/ARDS. Genetic variants replicating their association with trauma related-ALI in Stage II were validated in a second trauma-associated ALI population (n=224, Stage III). RESULTS In Stage I, non-overlapping SNPs were significantly associated with ARDS from direct/indirect lung injury, respectively. The association between rs1190286 (POPDC3) and reduced risk of ARDS from pulmonary injury was validated in Stage II (p<0.003). SNP rs324420 (FAAH) was consistently associated with increased risk of ARDS from extrapulmonary causes in two independent ALI-trauma populations (p<0.006, Stage II; p<0.05, Stage III). Meta-analysis confirmed these associations. CONCLUSIONS Different genetic variants may influence ARDS susceptibility depending on direct versus indirect insults. Functional SNPs in POPDC3 and FAAH genes may be driving the association with direct and indirect ALI/ARDS, respectively.
Collapse
Affiliation(s)
- Paula Tejera
- Department of Environmental Health, Harvard School of Public Health, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Majka SM, Miller HL, Sullivan T, Erickson PF, Kong R, Weiser-Evans M, Nemenoff R, Moldovan R, Morandi SA, Davis JA, Klemm DJ. Adipose lineage specification of bone marrow-derived myeloid cells. Adipocyte 2012; 1:215-229. [PMID: 23700536 PMCID: PMC3609111 DOI: 10.4161/adip.21496] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
We have reported the production of white adipocytes in adipose tissue from hematopoietic progenitors arising from bone marrow. However, technical challenges have hindered detection of this adipocyte population by certain other laboratories. These disparate results highlight the need for sensitive and definitive techniques to identify bone marrow progenitor (BMP)-derived adipocytes. In these studies we exploited new models and methods to enhance detection of this adipocyte population. Here we showed that confocal microscopy with spectrum acquisition could effectively identify green fluorescent protein (GFP) positive BMP-derived adipocytes by matching their fluorescence spectrum to that of native GFP. Likewise, imaging flow cytometry made it possible to visualize intact unilocular and multilocular GFP-positive BMP-derived adipocytes and distinguished them from non-fluorescent adipocytes and cell debris in the cytometer flow stream. We also devised a strategy to detect marker genes in flow-enriched adipocytes from which stromal cells were excluded. This technique also proved to be an efficient means for detecting genetically labeled adipocytes and should be applicable to models in which marker gene expression is low or absent. Finally, in vivo imaging of mice transplanted with BM from adipocyte-targeted luciferase donors showed a time-dependent increase in luciferase activity, with the bulk of luciferase activity confined to adipocytes rather than stromal cells. These results confirmed and extended our previous reports and provided proof-of-principle for sensitive techniques and models for detection and study of these unique cells.
Collapse
|
45
|
Protective effects of bone marrow mononuclear cell therapy on lung and heart in an elastase-induced emphysema model. Respir Physiol Neurobiol 2012; 182:26-36. [PMID: 22266352 DOI: 10.1016/j.resp.2012.01.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2011] [Revised: 01/07/2012] [Accepted: 01/08/2012] [Indexed: 12/21/2022]
Abstract
We hypothesized that bone marrow-derived mononuclear cell (BMDMC) therapy protects the lung and consequently the heart in experimental elastase-induced emphysema. Twenty-four female C57BL/6 mice were intratracheally instilled with saline (C group) or porcine pancreatic elastase (E group) once a week during 4 weeks. C and E groups were randomized into subgroups receiving saline (SAL) or male BMDMCs (2 × 10(6), CELL) intravenously 3h after the first saline or elastase instillation. Compared to E-SAL group, E-CELL mice showed, at 5 weeks: lower mean linear intercept, neutrophil infiltration, elastolysis, collagen fiber deposition in alveolar septa and pulmonary vessel wall, lung cell apoptosis, right ventricle wall thickness and area, higher endothelial growth factor and insulin-like growth factor mRNA expressions in lung tissue, and reduced platelet-derived growth factor, transforming growth factor-β, and caspase-3 expressions. In conclusion, BMDMC therapy was effective at modulating the inflammatory and remodeling processes in the present model of elastase-induced emphysema.
Collapse
|
46
|
Ornellas DS, Maron-Gutierrez T, Ornellas FM, Cruz FF, Oliveira GP, Lucas IH, Fujisaki L, Oliveira MG, Teodoro WR, Capelozzi VL, Pelosi P, Morales MM, Rocco PRM. Early and late effects of bone marrow-derived mononuclear cell therapy on lung and distal organs in experimental sepsis. Respir Physiol Neurobiol 2011; 178:304-14. [PMID: 21763473 DOI: 10.1016/j.resp.2011.06.029] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2011] [Revised: 06/23/2011] [Accepted: 06/30/2011] [Indexed: 01/01/2023]
Abstract
We tested the hypothesis that bone marrow-derived mononuclear cells (BMDMCs) at an early phase of cecal ligation and puncture (CLP)-induced sepsis may have lasting effects on: (1) lung mechanics and histology, (2) the structural remodelling of lung parenchyma, (3) lung, kidney, and liver cell apoptosis, and (4) pro- and anti-inflammatory cytokines and growth factors. At day 1, BMDMC significantly reduced mortality, as well as caspase-3, interleukin (IL)-6 and IL-1β, vascular endothelial growth factor, platelet-derived growth factor, hepatocyte growth factor, and transforming growth factor-β, but increased IL-10 mRNA expression in lung tissue in septic mice contributing to endothelium and epithelium alveolar repair and improvement of lung mechanics. BMDMC also prevented the increase of apoptotic cells in lung, liver, and kidney. At day 7, these early functional and morphological effects were preserved or further improved. In conclusion, in the present model of sepsis, the beneficial effects of early administration of BMDMCs on lung and distal organs were preserved, possibly by paracrine mechanisms.
Collapse
Affiliation(s)
- Debora S Ornellas
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Mechanisms of cellular therapy in respiratory diseases. Intensive Care Med 2011; 37:1421-31. [PMID: 21656291 DOI: 10.1007/s00134-011-2268-3] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2011] [Accepted: 05/05/2011] [Indexed: 01/08/2023]
Abstract
PURPOSE Stem cells present a variety of clinical implications in the lungs. According to their origin, these cells can be divided into embryonic and adult stem cells; however, due to the important ethical and safety limitations that are involved in the embryonic stem cell use, most studies have chosen to focus on adult stem cell therapy. This article aims to present and clarify the recent advances in the field of stem cell biology, as well as to highlight the effects of mesenchymal stem cell (MSC) therapy in the context of acute lung injury/acute respiratory distress syndrome and chronic disorders such as lung fibrosis and chronic obstructive pulmonary disease. METHODS For this purpose, we performed a critical review of adult stem cell therapies, covering the main clinical and experimental studies published in Pubmed databases in the past 11 years. Different characteristics were extracted from these articles, such as: the experimental model, strain, cellular type and administration route used as well as the positive or negative effects obtained. RESULTS There is evidence for beneficial effects of MSC on lung development, repair, and remodeling. The engraftment in the injured lung does not occur easily, but several studies report that paracrine factors can be effective in reducing inflammation and promoting tissue repair. MSC releases several growth factors and anti-inflammatory cytokines that regulate endothelial and epithelial permeability and reduce the severity of inflammation. CONCLUSION A better understanding of the mechanisms that control cell division and differentiation, as well as of their paracrine effects, is required to enable the optimal use of bone marrow-derived stem cell therapy to treat human respiratory diseases.
Collapse
|
48
|
Abreu SC, Antunes MA, Maron-Gutierrez T, Cruz FF, Carmo LGRR, Ornellas DS, Junior HC, Absaber AM, Parra ER, Capelozzi VL, Morales MM, Rocco PRM. Effects of bone marrow-derived mononuclear cells on airway and lung parenchyma remodeling in a murine model of chronic allergic inflammation. Respir Physiol Neurobiol 2010; 175:153-63. [PMID: 21050897 DOI: 10.1016/j.resp.2010.10.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2010] [Revised: 10/17/2010] [Accepted: 10/22/2010] [Indexed: 10/18/2022]
Abstract
We hypothesized that bone marrow-derived mononuclear cells (BMDMC) would attenuate the remodeling process in a chronic allergic inflammation model. C57BL/6 mice were assigned to two groups. In OVA, mice were sensitized and repeatedly challenged with ovalbumin. Control mice (C) received saline under the same protocol. C and OVA were further randomized to receive BMDMC (2 × 10⁶) or saline intravenously 24 h before the first challenge. BMDMC therapy reduced eosinophil infiltration, smooth muscle-specific actin expression, subepithelial fibrosis, and myocyte hypertrophy and hyperplasia, thus causing a decrease in airway hyperresponsiveness and lung mechanical parameters. BMDMC from green fluorescent protein (GFP)-transgenic mice transplanted into GFP-negative mice yielded lower engraftment in OVA. BMDMC increased insulin-like growth factor expression, but reduced interleukin-5, transforming growth factor-β, platelet-derived growth factor, and vascular endothelial growth factor mRNA expression. In conclusion, in the present chronic allergic inflammation model, BMDMC therapy was an effective pre-treatment protocol that potentiated airway epithelial cell repair and prevented inflammatory and remodeling processes.
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
|