1
|
Iban-Arias R, Yang EJ, Griggs E, Soares Dias Portela A, Osman A, Trageser KJ, Shahed M, Maria Pasinetti G. Ad-derived bone marrow transplant induces proinflammatory immune peripheral mechanisms accompanied by decreased neuroplasticity and reduced gut microbiome diversity affecting AD-like phenotype in the absence of Aβ neuropathology. Brain Behav Immun 2024; 118:252-272. [PMID: 38461954 DOI: 10.1016/j.bbi.2024.03.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 02/29/2024] [Accepted: 03/07/2024] [Indexed: 03/12/2024] Open
Abstract
Immune system dysfunction is increasingly recognized as a significant feature that contributes to Alzheimer's disease (AD) pathogenesis, reflected by alterations in central and peripheral responses leading to detrimental mechanisms that can contribute to the worsening of the disease. The damaging alterations in the peripheral immune system may disrupt the peripheral-central immune crosstalk, implicating the gut microbiota in this complex interaction. The central hypothesis posits that the immune signature inherently harbored in bone marrow (BM) cells can be transferred through allogeneic transplantation, influencing the recipient's immune system and modulating peripheral, gut, and brain immune responses. Employing a genetically modified mouse model to develop AD-type pathology we found that recipient wild-type (WT) mice engrafted with AD-derived BM, recapitulated the peripheral immune inflammatory donor phenotype, associated with a significant acceleration of cognitive deterioration in the absence of any overt change in AD-type amyloid neuropathology. Moreover, transcriptomic and phylogenetic 16S microbiome analysis evidence on these animals revealed a significantly impaired expression of genes associated with synaptic plasticity and neurotransmission in the brain and reduced bacteria diversity, respectively, compared to mice engrafted with WT BM. This investigation sheds light on the pivotal role of the peripheral immune system in the brain-gut-periphery axis and its profound potential to shape the trajectory of AD. In summary, this study advances our understanding of the complex interplay among the peripheral immune system, brain functionality, and the gut microbiome, which collectively influence AD onset and progression.
Collapse
Affiliation(s)
- Ruth Iban-Arias
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Eun-Jeong Yang
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Elizabeth Griggs
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | | | - Aya Osman
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Kyle J Trageser
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Mahadi Shahed
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Giulio Maria Pasinetti
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Center for Molecular Integrative Neuroresilience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Geriatrics Research, Education and Clinical Center, James J. Peters Department of Veterans Affairs Medical Center, Bronx, NY 10468, USA.
| |
Collapse
|
2
|
Sokolova AV, Domnina AP, Mikhailov VM. Accumulation of Dystrophin-Positive Muscle Fibers and Improvement of Neuromuscular Junctions in mdx Mouse Muscles after Bone Marrow Transplantation under Different Conditions. Int J Mol Sci 2023; 24:ijms24108892. [PMID: 37240237 DOI: 10.3390/ijms24108892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 05/13/2023] [Accepted: 05/14/2023] [Indexed: 05/28/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is a severe muscular disorder caused by mutations in the dystrophin gene. It leads to respiratory and cardiac failure and premature death at a young age. Although recent studies have greatly deepened the understanding of the primary and secondary pathogenetic mechanisms of DMD, an effective treatment remains elusive. In recent decades, stem cells have emerged as a novel therapeutic product for a variety of diseases. In this study, we investigated nonmyeloablative bone marrow cell (BMC) transplantation as a method of cell therapy for DMD in an mdx mouse model. By using BMC transplantation from GFP-positive mice, we confirmed that BMCs participate in the muscle restoration of mdx mice. We analyzed both syngeneic and allogeneic BMC transplantation under different conditions. Our data indicated that 3 Gy X-ray irradiation with subsequent BMC transplantation improved dystrophin synthesis and the structure of striated muscle fibers (SMFs) in mdx mice as well as decreasing the death rate of SMFs. In addition, we observed the normalization of neuromuscular junctions (NMJs) in mdx mice after nonmyeloablative BMC transplantation. In conclusion, we demonstrated that nonmyeloablative BMC transplantation could be considered a method for DMD treatment.
Collapse
Affiliation(s)
| | - Alisa P Domnina
- Institute of Cytology, Russian Academy of Sciences, 194064 Saint-Petersburg, Russia
| | | |
Collapse
|
3
|
Picoli CC, Martins PR, Wong XLC, Righi T, Guimarães PPG, Pinto MCX, Amorim JH, Azevedo VAC, Pereira SR, Kanashiro A, Cruz FC, Resende RR, Mintz A, Frenette PS, Birbrair A. Whole bone subcutaneous transplantation as a strategy to study precisely the bone marrow niche. Stem Cell Rev Rep 2022; 19:906-927. [PMID: 36585572 DOI: 10.1007/s12015-022-10496-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/15/2022] [Indexed: 01/01/2023]
Abstract
Hematopoietic stem cells are maintained in a specialized microenvironment, known as the 'niche', within the bone marrow. Understanding the contribution of cellular and molecular components within the bone marrow niche for the maintenance of hematopoietic stem cells is crucial for the success of therapeutic applications. So far, the roles of crucial mechanisms within the bone marrow niche have been explored in transgenic animals in which genetic modifications are ubiquitously introduced in the whole body. The lack of precise tools to explore genetic alterations exclusively within the bone marrow prevents our determination of whether the observed outcomes result from confounding effects from other organs. Here, we developed a new method - 'whole bone subcutaneous transplantation'- to study the bone marrow niche in transgenic animals precisely. Using immunolabeling of CD45.1 (donor) vs. CD45.2 (recipient) hematopoeitic stem cells, we demonstrated that hematopoeitic stem cells from the host animals colonize the subcutaneously transplanted femurs after transplantation, while the hematopoietic stem cells from the donor disappear. Strikinlgy, the bone marrow niche of these subcutaneously transplanted femurs remain from the donor mice, enabling us to study specifically cells of the bone marrow niche using this model. We also showed that genetic ablation of peri-arteriolar cells specifically in donor femurs reduced the numbers of hematopoietic stem cells in these bones. This supports the use of this strategy as a model, in combination with genetic tools, to evaluate how bone marrow niche specific modifications may impact non-modified hematopoietic stem cells. Thus, this approach can be utilized for genetic manipulation in vivo of specific cell types only within the bone marrow. The combination of whole bone subcutaneous transplantation with rodent transgenic models will facilitate a more precise, complex and comprehensive understanding of existing problems in the study of the hematopoietic stem cell bone marrow niche.
Collapse
Affiliation(s)
- Caroline C Picoli
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | | | - Xiao Lin Casey Wong
- Department of Dermatology, University of Wisconsin-Madison, Medical Sciences Center, Rm 4385, 1300 University Avenue, Madison, WI, 53706, USA
| | - Thamires Righi
- Department of Dermatology, University of Wisconsin-Madison, Medical Sciences Center, Rm 4385, 1300 University Avenue, Madison, WI, 53706, USA.,Department of Pharmacology, Federal University of São Paulo, São Paulo, SP, Brazil
| | - Pedro P G Guimarães
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Mauro C X Pinto
- Institute of Biological Sciences, Federal University of Goiás, Goiânia, GO, Brazil
| | - Jaime H Amorim
- Center of Biological Sciences and Health, Federal University of West Bahia, Barreiras, BA, Brazil
| | - Vasco A C Azevedo
- Department of Genetics, Ecology and Evolution, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | | | - Alexandre Kanashiro
- Department of Dermatology, University of Wisconsin-Madison, Medical Sciences Center, Rm 4385, 1300 University Avenue, Madison, WI, 53706, USA
| | - Fabio Cardoso Cruz
- Department of Pharmacology, Federal University of São Paulo, São Paulo, SP, Brazil
| | - Rodrigo R Resende
- Department of Biochemistry and Immunology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Akiva Mintz
- Department of Radiology, Columbia University Medical Center, New York, NY, USA
| | - Paul S Frenette
- Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, New York, NY, USA.,Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, USA.,Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Alexander Birbrair
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil. .,Department of Dermatology, University of Wisconsin-Madison, Medical Sciences Center, Rm 4385, 1300 University Avenue, Madison, WI, 53706, USA. .,Department of Radiology, Columbia University Medical Center, New York, NY, USA.
| |
Collapse
|
4
|
Garrigós MM, de Oliveira FA, Nucci MP, Nucci LP, Alves ADH, Dias OFM, Gamarra LF. How mesenchymal stem cell cotransplantation with hematopoietic stem cells can improve engraftment in animal models. World J Stem Cells 2022; 14:658-679. [PMID: 36157912 PMCID: PMC9453272 DOI: 10.4252/wjsc.v14.i8.658] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/27/2022] [Accepted: 07/26/2022] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Bone marrow transplantation (BMT) can be applied to both hematopoietic and nonhematopoietic diseases; nonetheless, it still comes with a number of challenges and limitations that contribute to treatment failure. Bearing this in mind, a possible way to increase the success rate of BMT would be cotransplantation of mesenchymal stem cells (MSCs) and hematopoietic stem cells (HSCs) to improve the bone marrow niche and secrete molecules that enhance the hematopoietic engraftment.
AIM To analyze HSC and MSC characteristics and their interactions through cotransplantation in murine models.
METHODS We searched for original articles indexed in PubMed and Scopus during the last decade that used HSC and MSC cotransplantation and in vivo BMT in animal models while evaluating cell engraftment. We excluded in vitro studies or studies that involved graft versus host disease or other hematological diseases and publications in languages other than English. In PubMed, we initially identified 555 articles and after selection, only 12 were chosen. In Scopus, 2010 were identified, and six were left after the screening and eligibility process.
RESULTS Of the 2565 articles found in the databases, only 18 original studies met the eligibility criteria. HSC distribution by source showed similar ratios, with human umbilical cord blood or animal bone marrow being administered mainly with a dose of 1 × 107 cells by intravenous or intrabone routes. However, MSCs had a high prevalence of human donors with a variety of sources (umbilical cord blood, bone marrow, tonsil, adipose tissue or fetal lung), using a lower dose, mainly 106 cells and ranging 104 to 1.5 × 107 cells, utilizing the same routes. MSCs were characterized prior to administration in almost every experiment. The recipient used was mostly immunodeficient mice submitted to low-dose irradiation or chemotherapy. The main technique of engraftment for HSC and MSC cotransplantation evaluation was chimerism, followed by hematopoietic reconstitution and survival analysis. Besides the engraftment, homing and cellularity were also evaluated in some studies.
CONCLUSION The preclinical findings validate the potential of MSCs to enable HSC engraftment in vivo in both xenogeneic and allogeneic hematopoietic cell transplantation animal models, in the absence of toxicity.
Collapse
Affiliation(s)
- Murilo Montenegro Garrigós
- Hospital Israelita Albert Einstein, São Paulo 05652-900, São Paulo, Brazil
- Instituto de Química, Universidade de São Paulo, São Paulo 05508-900, São Paulo, Brazil
| | | | - Mariana Penteado Nucci
- Hospital Israelita Albert Einstein, São Paulo 05652-900, São Paulo, Brazil
- LIM44-Hospital das Clínicas, Faculdade Medicina da Universidade de São Paulo, São Paulo 05403-000, Brazil
| | - Leopoldo Penteado Nucci
- Centro Universitário do Planalto Central, Área Especial para Industria nº 02 Setor Leste - Gama-DF, Brasília 72445-020, Distrito Federal, Brazil
| | | | | | | |
Collapse
|
5
|
Báječný M, Chen CL, Faltusová K, Heizer T, Szikszai K, Páral P, Šefc L, Nečas E. Hematopoiesis Remains Permissive to Bone Marrow Transplantation After Expansion of Progenitors and Resumption of Blood Cell Production. Front Cell Dev Biol 2021; 9:660617. [PMID: 34414177 PMCID: PMC8369928 DOI: 10.3389/fcell.2021.660617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 07/05/2021] [Indexed: 11/30/2022] Open
Abstract
The immense regenerative power of hematopoietic tissue stems from the activation of the immature stem cells and the progenitor cells. After partial damage, hematopoiesis is reconstituted through a period of intense regeneration when blood cell production originates from erythro-myeloid progenitors in the virtual absence of stem cells. Since the damaged hematopoiesis can also be reconstituted from transplanted hematopoietic cells, we asked whether this also leads to the transient state when activated progenitors initially execute blood cell production. We first showed that the early reconstitution of hematopoiesis from transplanted cells gives rise to extended populations of developmentally advanced but altered progenitor cells, similar to those previously identified in the bone marrow regenerating from endogenous cells. We then identified the cells that give rise to these progenitors after transplantation as LSK CD48– cells. In the submyeloablative irradiated host mice, the transplanted LSK CD48– cells preferably colonized the spleen. Unlike the endogenous hematopoiesis reconstituting cells, the transplanted whole bone marrow cells and sorted LSK CD48– cells had greater potential to differentiate to B-lymphopoiesis. Separate transplantation of the CD150– and CD150+ subsets of LSK CD48– cells suggested that CD150– cells had a greater preference to B-lymphopoiesis than CD150+ cells. In the intensively regenerating hematopoiesis, the CD71/Sca-1 plot of immature murine hematopoietic cells revealed that the expanded populations of altered myeloid progenitors were highly variable in the different places of hematopoietic tissues. This high variability is likely caused by the heterogeneity of the hematopoiesis supporting stroma. Lastly, we demonstrate that during the period when active hematopoiesis resumes from transplanted cells, the hematopoietic tissues still remain highly permissive for further engraftment of transplanted cells, particularly the stem cells. Thus, these results provide a rationale for the transplantation of the hematopoietic stem cells in successive doses that could be used to boost the transplantation outcome.
Collapse
Affiliation(s)
- Martin Báječný
- 1st Faculty of Medicine, Institute of Pathological Physiology, Charles University, Prague, Czechia.,1st Faculty of Medicine, Center for Advanced Preclinical Imaging (CAPI), Charles University, Prague, Czechia
| | - Chia-Ling Chen
- 1st Faculty of Medicine, Institute of Pathological Physiology, Charles University, Prague, Czechia
| | - Kateřina Faltusová
- 1st Faculty of Medicine, Institute of Pathological Physiology, Charles University, Prague, Czechia
| | - Tomáš Heizer
- 1st Faculty of Medicine, Institute of Pathological Physiology, Charles University, Prague, Czechia.,1st Faculty of Medicine, Center for Advanced Preclinical Imaging (CAPI), Charles University, Prague, Czechia
| | - Katarína Szikszai
- 1st Faculty of Medicine, Institute of Pathological Physiology, Charles University, Prague, Czechia
| | - Petr Páral
- 1st Faculty of Medicine, Institute of Pathological Physiology, Charles University, Prague, Czechia.,1st Faculty of Medicine, Center for Advanced Preclinical Imaging (CAPI), Charles University, Prague, Czechia
| | - Luděk Šefc
- 1st Faculty of Medicine, Center for Advanced Preclinical Imaging (CAPI), Charles University, Prague, Czechia
| | - Emanuel Nečas
- 1st Faculty of Medicine, Institute of Pathological Physiology, Charles University, Prague, Czechia
| |
Collapse
|
6
|
Oliveira FA, Nucci MP, Mamani JB, Alves AH, Rego GNA, Kondo AT, Hamerschlak N, Junqueira MS, de Souza LEB, Gamarra LF. Multimodal Tracking of Hematopoietic Stem Cells from Young and Old Mice Labeled with Magnetic-Fluorescent Nanoparticles and Their Grafting by Bioluminescence in a Bone Marrow Transplant Model. Biomedicines 2021; 9:biomedicines9070752. [PMID: 34209598 PMCID: PMC8301491 DOI: 10.3390/biomedicines9070752] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 06/18/2021] [Accepted: 06/22/2021] [Indexed: 12/12/2022] Open
Abstract
This study proposes an innovative way to evaluate the homing and tracking of hematopoietic stem cells from young and old mice labeled with SPIONNIRF-Rh conjugated with two types of fluorophores (NIRF and Rhodamine), and their grafting by bioluminescence (BLI) in a bone marrow transplant (BMT) model. In an in vitro study, we isolated bone marrow mononuclear cells (BM-MNC) from young and old mice, and analyzed the physical-chemical characteristics of SPIONNIRF-Rh, their internalization, cell viability, and the iron quantification by NIRF, ICP-MS, and MRI. The in vivo study was performed in a BMT model to evaluate the homing, tracking, and grafting of young and old BM-MNC labeled with SPIONNIRF-Rh by NIRF and BLI, as well as the hematological reconstitution for 120 days. 5FU influenced the number of cells isolated mainly in young cells. SPIONNIRF-Rh had adequate characteristics for efficient internalization into BM-MNC. The iron load quantification by NIRF, ICP-MS, and MRI was in the order of 104 SPIONNIRF-Rh/BM-MNC. In the in vivo study, the acute NIRF evaluation showed higher signal intensity in the spinal cord and abdominal region, and the BLI evaluation allowed follow-up (11-120 days), achieving a peak of intensity at 30 days, which remained stable around 108 photons/s until the end. The hematologic evaluation showed similar behavior until 30 days and the histological results confirm that iron is present in almost all tissue evaluated. Our results on BM-MNC homing and tracking in the BMT model did not show a difference in migration or grafting of cells from young or old mice, with the hemogram analysis trending to differentiation towards the myeloid lineage in mice that received cells from old animals. The cell homing by NIRF and long term cell follow-up by BLI highlighted the relevance of the multimodal nanoparticles and combined techniques for evaluation.
Collapse
Affiliation(s)
- Fernando A. Oliveira
- Hospital Israelita Albert Einstein, São Paulo 05652-000, SP, Brazil; (F.A.O.); (M.P.N.); (J.B.M.); (A.H.A.); (G.N.A.R.); (A.T.K.); (N.H.)
| | - Mariana P. Nucci
- Hospital Israelita Albert Einstein, São Paulo 05652-000, SP, Brazil; (F.A.O.); (M.P.N.); (J.B.M.); (A.H.A.); (G.N.A.R.); (A.T.K.); (N.H.)
- LIM44—Hospital das Clínicas da Faculdade Medicina da Universidade de São Paulo, São Paulo 05403-000, SP, Brazil
| | - Javier B. Mamani
- Hospital Israelita Albert Einstein, São Paulo 05652-000, SP, Brazil; (F.A.O.); (M.P.N.); (J.B.M.); (A.H.A.); (G.N.A.R.); (A.T.K.); (N.H.)
| | - Arielly H. Alves
- Hospital Israelita Albert Einstein, São Paulo 05652-000, SP, Brazil; (F.A.O.); (M.P.N.); (J.B.M.); (A.H.A.); (G.N.A.R.); (A.T.K.); (N.H.)
| | - Gabriel N. A. Rego
- Hospital Israelita Albert Einstein, São Paulo 05652-000, SP, Brazil; (F.A.O.); (M.P.N.); (J.B.M.); (A.H.A.); (G.N.A.R.); (A.T.K.); (N.H.)
| | - Andrea T. Kondo
- Hospital Israelita Albert Einstein, São Paulo 05652-000, SP, Brazil; (F.A.O.); (M.P.N.); (J.B.M.); (A.H.A.); (G.N.A.R.); (A.T.K.); (N.H.)
| | - Nelson Hamerschlak
- Hospital Israelita Albert Einstein, São Paulo 05652-000, SP, Brazil; (F.A.O.); (M.P.N.); (J.B.M.); (A.H.A.); (G.N.A.R.); (A.T.K.); (N.H.)
| | - Mara S. Junqueira
- Center for Translational Research in Oncology, Cancer Institute of the State of Sao Paulo—ICESP, São Paulo 01246-000, SP, Brazil;
| | - Lucas E. B. de Souza
- Hemocentro de Ribeirão Preto, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto 14051-060, SP, Brazil;
| | - Lionel F. Gamarra
- Hospital Israelita Albert Einstein, São Paulo 05652-000, SP, Brazil; (F.A.O.); (M.P.N.); (J.B.M.); (A.H.A.); (G.N.A.R.); (A.T.K.); (N.H.)
- Correspondence: ; Tel.: +55-11-2151-0243
| |
Collapse
|
7
|
Zuro D, Madabushi SS, Brooks J, Chen BT, Goud J, Salhotra A, Song JY, Parra LE, Pierini A, Sanchez JF, Stein A, Malki MA, Kortylewski M, Wong JYC, Alaei P, Froelich J, Storme G, Hui SK. First Multimodal, Three-Dimensional, Image-Guided Total Marrow Irradiation Model for Preclinical Bone Marrow Transplantation Studies. Int J Radiat Oncol Biol Phys 2021; 111:671-683. [PMID: 34119592 DOI: 10.1016/j.ijrobp.2021.06.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 05/28/2021] [Accepted: 06/04/2021] [Indexed: 01/13/2023]
Abstract
PURPOSE Total marrow irradiation (TMI) has significantly advanced radiation conditioning for hematopoietic cell transplantation in hematologic malignancies by reducing conditioning-induced toxicities and improving survival outcomes in relapsed/refractory patients. However, the relapse rate remains high, and the lack of a preclinical TMI model has hindered scientific advancements. To accelerate TMI translation to the clinic, we developed a TMI delivery system in preclinical models. METHODS AND MATERIALS A Precision X-RAD SmART irradiator was used for TMI model development. Images acquired with whole-body contrast-enhanced computed tomography (CT) were used to reconstruct and delineate targets and vital organs for each mouse. Multiple beam and CT-guided Monte Carlo-based plans were performed to optimize doses to the targets and to vary doses to the vital organs. Long-term engraftment and reconstitution potential were evaluated by a congenic bone marrow transplantation (BMT) model and serial secondary BMT, respectively. Donor cell engraftment was measured using noninvasive bioluminescence imaging and flow cytometry. RESULTS Multimodal imaging enabled identification of targets (skeleton and spleen) and vital organs (eg, lungs, gut, liver). In contrast to total body irradiation (TBI), TMI treatment allowed variation of radiation dose exposure to organs relative to the target dose. Dose reduction mirrored that in clinical TMI studies. Similar to TBI, mice treated with different TMI regimens showed full long-term donor engraftment in primary BMT and second serial BMT. The TBI-treated mice showed acute gut damage, which was minimized in mice treated with TMI. CONCLUSIONS A novel multimodal image guided preclinical TMI model is reported here. TMI conditioning maintained long-term engraftment with reconstitution potential and reduced organ damage. Therefore, this TMI model provides a unique opportunity to study the therapeutic benefit of reduced organ damage and BM dose escalation to optimize treatment regimens in BMT and hematologic malignancies.
Collapse
Affiliation(s)
- Darren Zuro
- Department of Radiation Oncology, City of Hope Medical Center, Duarte, California
| | | | - Jamison Brooks
- Department of Radiation Oncology, City of Hope Medical Center, Duarte, California
| | - Bihong T Chen
- Department of Diagnostic Radiology, City of Hope Medical Center, Duarte, California
| | - Janagama Goud
- Department of Radiation Oncology, City of Hope Medical Center, Duarte, California
| | - Amandeep Salhotra
- Department of Hematology and HCT, City of Hope Medical Center, Duarte, California
| | - Joo Y Song
- Department of Pathology, City of Hope Medical Center, Duarte, California
| | | | - Antonio Pierini
- Division of Hematology and Clinical Immunology, Department of Medicine, University of Perugia, Perugia, Italy
| | - James F Sanchez
- Beckman Research Institute of City of Hope, Duarte, California
| | - Anthony Stein
- Department of Hematology and HCT, City of Hope Medical Center, Duarte, California
| | - Monzr Al Malki
- Department of Hematology and HCT, City of Hope Medical Center, Duarte, California
| | - Marcin Kortylewski
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope, Duarte, California
| | - Jeffrey Y C Wong
- Department of Radiation Oncology, City of Hope Medical Center, Duarte, California
| | - Parham Alaei
- Department of Radiation Oncology, University of Minnesota, Minneapolis, Minnesota
| | - Jerry Froelich
- Department of Radiology, University of Minnesota, Minneapolis, Minnesota
| | - Guy Storme
- Department of Radiotherapy UZ Brussels, Brussels, Belgium
| | - Susanta K Hui
- Department of Radiation Oncology, City of Hope Medical Center, Duarte, California; Beckman Research Institute of City of Hope, Duarte, California; Department of Radiation Oncology, University of Minnesota, Minneapolis, Minnesota.
| |
Collapse
|
8
|
Oliveira FA, Nucci MP, Filgueiras IS, Ferreira JM, Nucci LP, Mamani JB, Alvieri F, Souza LEB, Rego GNA, Kondo AT, Hamerschlak N, Gamarra LF. Noninvasive Tracking of Hematopoietic Stem Cells in a Bone Marrow Transplant Model. Cells 2020; 9:cells9040939. [PMID: 32290257 PMCID: PMC7226958 DOI: 10.3390/cells9040939] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 03/31/2020] [Accepted: 04/03/2020] [Indexed: 12/11/2022] Open
Abstract
The hematopoietic stem cell engraftment depends on adequate cell numbers, their homing, and the subsequent short and long-term engraftment of these cells in the niche. We performed a systematic review of the methods employed to track hematopoietic reconstitution using molecular imaging. We searched articles indexed, published prior to January 2020, in PubMed, Cochrane, and Scopus with the following keyword sequences: (Hematopoietic Stem Cell OR Hematopoietic Progenitor Cell) AND (Tracking OR Homing) AND (Transplantation). Of 2191 articles identified, only 21 articles were included in this review, after screening and eligibility assessment. The cell source was in the majority of bone marrow from mice (43%), followed by the umbilical cord from humans (33%). The labeling agent had the follow distribution between the selected studies: 14% nanoparticle, 29% radioisotope, 19% fluorophore, 19% luciferase, and 19% animal transgenic. The type of graft used in the studies was 57% allogeneic, 38% xenogeneic, and 5% autologous, being the HSC receptor: 57% mice, 9% rat, 19% fish, 5% for dog, porcine and salamander. The imaging technique used in the HSC tracking had the following distribution between studies: Positron emission tomography/single-photon emission computed tomography 29%, bioluminescence 33%, fluorescence 19%, magnetic resonance imaging 14%, and near-infrared fluorescence imaging 5%. The efficiency of the graft was evaluated in 61% of the selected studies, and before one month of implantation, the cell renewal was very low (less than 20%), but after three months, the efficiency was more than 50%, mainly in the allogeneic graft. In conclusion, our review showed an increase in using noninvasive imaging techniques in HSC tracking using the bone marrow transplant model. However, successful transplantation depends on the formation of engraftment, and the functionality of cells after the graft, aspects that are poorly explored and that have high relevance for clinical analysis.
Collapse
Affiliation(s)
- Fernando A. Oliveira
- Hospital Israelita Albert Einstein, São Paulo 05652-900, Brazil; (F.A.O.); (I.S.F.); (J.M.F.); (J.B.M.); (F.A.); (G.N.A.R.); (A.T.K.); (N.H.)
| | - Mariana P. Nucci
- LIM44—Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo 01246-903, Brazil;
| | - Igor S. Filgueiras
- Hospital Israelita Albert Einstein, São Paulo 05652-900, Brazil; (F.A.O.); (I.S.F.); (J.M.F.); (J.B.M.); (F.A.); (G.N.A.R.); (A.T.K.); (N.H.)
| | - João M. Ferreira
- Hospital Israelita Albert Einstein, São Paulo 05652-900, Brazil; (F.A.O.); (I.S.F.); (J.M.F.); (J.B.M.); (F.A.); (G.N.A.R.); (A.T.K.); (N.H.)
| | - Leopoldo P. Nucci
- Centro Universitário do Planalto Central, Brasília DF 72445-020, Brazil;
| | - Javier B. Mamani
- Hospital Israelita Albert Einstein, São Paulo 05652-900, Brazil; (F.A.O.); (I.S.F.); (J.M.F.); (J.B.M.); (F.A.); (G.N.A.R.); (A.T.K.); (N.H.)
| | - Fernando Alvieri
- Hospital Israelita Albert Einstein, São Paulo 05652-900, Brazil; (F.A.O.); (I.S.F.); (J.M.F.); (J.B.M.); (F.A.); (G.N.A.R.); (A.T.K.); (N.H.)
| | - Lucas E. B. Souza
- Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto SP 14049-900, Brazil;
| | - Gabriel N. A. Rego
- Hospital Israelita Albert Einstein, São Paulo 05652-900, Brazil; (F.A.O.); (I.S.F.); (J.M.F.); (J.B.M.); (F.A.); (G.N.A.R.); (A.T.K.); (N.H.)
| | - Andrea T. Kondo
- Hospital Israelita Albert Einstein, São Paulo 05652-900, Brazil; (F.A.O.); (I.S.F.); (J.M.F.); (J.B.M.); (F.A.); (G.N.A.R.); (A.T.K.); (N.H.)
| | - Nelson Hamerschlak
- Hospital Israelita Albert Einstein, São Paulo 05652-900, Brazil; (F.A.O.); (I.S.F.); (J.M.F.); (J.B.M.); (F.A.); (G.N.A.R.); (A.T.K.); (N.H.)
| | - Lionel F. Gamarra
- Hospital Israelita Albert Einstein, São Paulo 05652-900, Brazil; (F.A.O.); (I.S.F.); (J.M.F.); (J.B.M.); (F.A.); (G.N.A.R.); (A.T.K.); (N.H.)
- Correspondence: ; Tel.: +55-11-2151-0243
| |
Collapse
|
9
|
Selective local irradiation improves islet engraftment and survival in intra-bone marrow islet transplantation. Cytotherapy 2019; 21:1025-1032. [PMID: 31444049 DOI: 10.1016/j.jcyt.2019.07.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 07/16/2019] [Accepted: 07/22/2019] [Indexed: 01/28/2023]
Abstract
BACKGROUND Bone marrow (BM) is as an alternative site for islet transplantation, but it is not an immunoprotected microenvironment and allogeneic islets are rejected. However, the BM, for its structure and anatomic position, offers the possibility to modulate microenvironment by local interventions. We here investigate whether local irradiation is able to improve islet engraftment and prevent rejection in BM in the absence of immunosuppression. METHODS A model of BM local irradiation was set up. Islets were transplanted in syngeneic and fully major histocompatibility complex-mismatched recipients in control and locally irradiated BM; gain of normoglycemia and time to rejection were evaluated. RESULTS BM local irradiation proved to be a selective and safe procedure. Syngeneic islet transplantation into locally irradiated BM had better outcome compared with not irradiated recipients in terms of capacity to gain normoglycemia (100% versus 56% in irradiated versus not irradiated mice). In the allogenic setting, glycemia was significantly lower in the first days after transplantation in the group of irradiated mice and local irradiation also delayed time to graft rejection (from 4 ± 1 days for not irradiated to 11 ± 1 days for locally irradiated mice). DISCUSSION These data indicate that local immunosuppression by irradiation before islet transplantation in BM favors islet engraftment and delays time to rejection.
Collapse
|
10
|
Cho HJ, Lee JW, Cho HJ, Lee CS, Kim HS. Identification of Adult Mesodermal Progenitor Cells and Hierarchy in Atherosclerotic Vascular Calcification. Stem Cells 2018; 36:1075-1096. [PMID: 29484798 DOI: 10.1002/stem.2814] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 01/26/2018] [Accepted: 02/05/2018] [Indexed: 01/01/2023]
Abstract
The nature of calcifying progenitor cells remains elusive. In this study, we investigated the developmental hierarchy and dynamics of progenitor cells. In vitro and in vivo reconstitution assays demonstrated that Sca-1+/PDGFRα- cells in the bone marrow (BM) are the ancestors of Sca-1+/PDGFRα+ cells. Cells of CD29 + Sca-1+/PDGFRα- lineage in the BM showed both hematopoietic potential with osteoclastic differentiation ability as well as mesenchymal stem cell-like properties with osteoblastic differentiation potential. Clonally-isolated BM-derived artery-infiltrated Sca-1+/PDGFRα- cells maintained osteoblastic/osteoclastic bipotency but lost hematopoietic activity. In hypercholesterolemic apolipoprotein-E-deficient (Apoe-/-) mice, the mobilization from BM to peripheral circulation, followed by migration into atherosclerotic plaques of Sca-1+/PDGFRα- cells, but not Sca-1+/PDGFRα+ cells, were significantly decreased, and Interleukin-1β (IL-1β) and Interleukin-5 (IL-5) mediated this response. Here, we demonstrated that Sca-1+/PDGFRα- cells are mesodermal progenitor cells in adults, and the dynamics of progenitor cells were regulated by atherosclerosis-related humoral factors. These results may contribute to better understanding of vascular homeostasis and assist in the development of novel therapies for atherosclerosis. Stem Cells 2018;36:1075-1096.
Collapse
Affiliation(s)
- Hyun-Jai Cho
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Jin-Woo Lee
- Strategic Center of Cell & Bio Therapy, Seoul National University Hospital, Seoul, Korea.,National Research Laboratory for Stem Cell Niche, Seoul National University College of Medicine, Seoul, Korea
| | - Hyun-Ju Cho
- Strategic Center of Cell & Bio Therapy, Seoul National University Hospital, Seoul, Korea.,National Research Laboratory for Stem Cell Niche, Seoul National University College of Medicine, Seoul, Korea
| | - Choon-Soo Lee
- Strategic Center of Cell & Bio Therapy, Seoul National University Hospital, Seoul, Korea.,National Research Laboratory for Stem Cell Niche, Seoul National University College of Medicine, Seoul, Korea.,World Class University Program, Department of Molecular Medicine and Biopharmaceutical Sciences, Seoul National University, Seoul, Korea
| | - Hyo-Soo Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea.,Strategic Center of Cell & Bio Therapy, Seoul National University Hospital, Seoul, Korea.,National Research Laboratory for Stem Cell Niche, Seoul National University College of Medicine, Seoul, Korea.,World Class University Program, Department of Molecular Medicine and Biopharmaceutical Sciences, Seoul National University, Seoul, Korea
| |
Collapse
|
11
|
Arbab AS, Rashid MH, Angara K, Borin TF, Lin PC, Jain M, Achyut BR. Major Challenges and Potential Microenvironment-Targeted Therapies in Glioblastoma. Int J Mol Sci 2017; 18:ijms18122732. [PMID: 29258180 PMCID: PMC5751333 DOI: 10.3390/ijms18122732] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 12/13/2017] [Accepted: 12/15/2017] [Indexed: 02/07/2023] Open
Abstract
Glioblastoma (GBM) is considered one of the most malignant, genetically heterogeneous, and therapy-resistant solid tumor. Therapeutic options are limited in GBM and involve surgical resection followed by chemotherapy and/or radiotherapy. Adjuvant therapies, including antiangiogenic treatments (AATs) targeting the VEGF–VEGFR pathway, have witnessed enhanced infiltration of bone marrow-derived myeloid cells, causing therapy resistance and tumor relapse in clinics and in preclinical models of GBM. This review article is focused on gathering previous clinical and preclinical reports featuring major challenges and lessons in GBM. Potential combination therapies targeting the tumor microenvironment (TME) to overcome the myeloid cell-mediated resistance problem in GBM are discussed. Future directions are focused on the use of TME-directed therapies in combination with standard therapy in clinical trials, and the exploration of novel therapies and GBM models for preclinical studies. We believe this review will guide the future of GBM research and therapy.
Collapse
Affiliation(s)
- Ali S Arbab
- Tumor Angiogenesis laboratory, Georgia Cancer Center, Department of Biochemistry and Molecular Biology, Augusta University, Augusta, GA 30912, USA.
| | - Mohammad H Rashid
- Tumor Angiogenesis laboratory, Georgia Cancer Center, Department of Biochemistry and Molecular Biology, Augusta University, Augusta, GA 30912, USA.
| | - Kartik Angara
- Tumor Angiogenesis laboratory, Georgia Cancer Center, Department of Biochemistry and Molecular Biology, Augusta University, Augusta, GA 30912, USA.
| | - Thaiz F Borin
- Tumor Angiogenesis laboratory, Georgia Cancer Center, Department of Biochemistry and Molecular Biology, Augusta University, Augusta, GA 30912, USA.
| | - Ping-Chang Lin
- Tumor Angiogenesis laboratory, Georgia Cancer Center, Department of Biochemistry and Molecular Biology, Augusta University, Augusta, GA 30912, USA.
| | - Meenu Jain
- Tumor Angiogenesis laboratory, Georgia Cancer Center, Department of Biochemistry and Molecular Biology, Augusta University, Augusta, GA 30912, USA.
| | - Bhagelu R Achyut
- Tumor Angiogenesis laboratory, Georgia Cancer Center, Department of Biochemistry and Molecular Biology, Augusta University, Augusta, GA 30912, USA.
| |
Collapse
|
12
|
Klomps LV, Zomorodi N, Kim HM. Role of transplanted bone marrow cells in development of rotator cuff muscle fatty degeneration in mice. J Shoulder Elbow Surg 2017; 26:2177-2186. [PMID: 28869071 DOI: 10.1016/j.jse.2017.06.032] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2017] [Revised: 06/12/2017] [Accepted: 06/19/2017] [Indexed: 02/01/2023]
Abstract
BACKGROUND Rotator cuff muscle fatty degeneration after a chronic tendon tear is an irreversible pathologic change associated with poor clinical outcomes of tendon repair, and its exact pathogenesis remains unknown. We sought to investigate the role of transplanted bone marrow cells in the development of fatty degeneration, specifically in adipocyte accumulation, using a mouse model. METHODS Fourteen mice were divided into 2 bone marrow chimeric animal groups: bone marrow transplantation (BMT) group and reverse BMT group. For the BMT group, C57BL/6J wild-type mice underwent whole body irradiation followed by BMT into the retro-orbital sinus from green fluorescent protein (GFP)-transgenic donor mice. For the reverse BMT group, GFP-transgenic mice received BMT from C57BL/6J wild-type donor mice after irradiation. The supraspinatus tendon, infraspinatus tendon, and suprascapular nerve were surgically transected 3 weeks after transplantation. The rotator cuff muscles were harvested 13 weeks after transplantation for histologic analysis and GFP immunohistochemistry. RESULTS On histologic examination, both groups showed substantial fatty degeneration, fibrosis, and atrophy of the cuff muscles. The BMT group showed no noticeable GFP immunostaining, whereas the reverse BMT group showed significantly stronger GFP staining in most adipocytes (P < .001). However, both groups also showed that a small number of adipocytes originated from transplanted bone marrow cells. A small number of myocytes showed a large cytoplasmic lipid vacuole resembling adipocytes. CONCLUSIONS This study's findings suggest that most adipocytes in fatty degeneration of the rotator cuff muscles originate from sources other than bone marrow-derived stem cells, and there may be more than 1 source for the adipocytes.
Collapse
Affiliation(s)
- Lawrence V Klomps
- Department of Orthopaedics and Rehabilitation, Penn State College of Medicine Milton S. Hershey Medical Center, Hershey, PA, USA
| | - Naseem Zomorodi
- Department of Orthopaedics and Rehabilitation, Penn State College of Medicine Milton S. Hershey Medical Center, Hershey, PA, USA
| | - H Mike Kim
- Department of Orthopaedics and Rehabilitation, Penn State College of Medicine Milton S. Hershey Medical Center, Hershey, PA, USA.
| |
Collapse
|
13
|
Locally Transplanted CD34+ Bone Marrow–Derived Cells Contribute to Vascular Healing After Vascular Injury. Transplant Proc 2017; 49:1467-1476. [DOI: 10.1016/j.transproceed.2017.01.081] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 01/24/2017] [Indexed: 01/13/2023]
|
14
|
Afshar SF, Zawaski JA, Inoue T, Rendon DA, Zieske AW, Punia JN, Sabek OM, Gaber MW. Investigating the Abscopal Effects of Radioablation on Shielded Bone Marrow in Rodent Models Using Multimodality Imaging. Radiat Res 2017; 188:56-65. [PMID: 28475423 DOI: 10.1667/rr14692.1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
The abscopal effect is the response to radiation at sites that are distant from the irradiated site of an organism, and it is thought to play a role in bone marrow (BM) recovery by initiating responses in the unirradiated bone marrow. Understanding the mechanism of this effect has applications in treating BM failure (BMF) and BM transplantation (BMT), and improving survival of nuclear disaster victims. Here, we investigated the use of multimodality imaging as a translational tool to longitudinally assess bone marrow recovery. We used positron emission tomography/computed tomography (PET/CT), magnetic resonance imaging (MRI) and optical imaging to quantify bone marrow activity, vascular response and marrow repopulation in fully and partially irradiated rodent models. We further measured the effects of radiation on serum cytokine levels, hematopoietic cell counts and histology. PET/CT imaging revealed a radiation-induced increase in proliferation in the shielded bone marrow (SBM) compared to exposed bone marrow (EBM) and sham controls. T2-weighted MRI showed radiation-induced hemorrhaging in the EBM and unirradiated SBM. In the EBM and SBM groups, we found alterations in serum cytokine and hormone levels and in hematopoietic cell population proportions, and histological evidence of osteoblast activation at the bone marrow interface. Importantly, we generated a BMT mouse model using fluorescent-labeled bone marrow donor cells and performed fluorescent imaging to reveal the migration of bone marrow cells from shielded to radioablated sites. Our study validates the use of multimodality imaging to monitor bone marrow recovery and provides evidence for the abscopal response in promoting bone marrow recovery after irradiation.
Collapse
Affiliation(s)
- Solmaz F Afshar
- a Department of Surgery, Houston Methodist Hospital Research Institute, Houston, Texas
| | - Janice A Zawaski
- b Hematology-Oncology Section, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Taeko Inoue
- b Hematology-Oncology Section, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - David A Rendon
- b Hematology-Oncology Section, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Arthur W Zieske
- d Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, Texas
| | - Jyotinder N Punia
- c Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas
| | - Omaima M Sabek
- a Department of Surgery, Houston Methodist Hospital Research Institute, Houston, Texas
| | - M Waleed Gaber
- b Hematology-Oncology Section, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
15
|
Verbiest T, Finnon R, Brown N, Finnon P, Bouffler S, Badie C. NOD Scid Gamma Mice Are Permissive to Allogeneic HSC Transplantation without Prior Conditioning. Int J Mol Sci 2016; 17:E1850. [PMID: 27827995 PMCID: PMC5133850 DOI: 10.3390/ijms17111850] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 10/14/2016] [Accepted: 11/02/2016] [Indexed: 11/16/2022] Open
Abstract
Scid hematopoietic stem cells (HSCs) have an intrinsic defect in their maintenance within the bone marrow (BM) niche which facilitates HSC transplantation without the absolute requirement of prior conditioning. Nevertheless, NOD scid mice have a significantly altered life span due to early development of thymic lymphomas, which compromises the ability to study the long-term fate of exogenous HSCs and their progeny. Here, we present data on the transplantation of HSCs into NOD scid gamma (NSG) mice to achieve long-term engraftment without prior conditioning. We transplanted allogeneic HSCs constitutively expressing the mCherry fluorescent marker into age-matched NSG mice and assessed donor chimerism 6 months post-transplantation. All transplanted NSG mice showed long-term myeloid and lymphoid cell chimerism. Also, in vivo irradiated HSCs showed long-term engraftment, although overall white blood cell (WBC) donor chimerism was lower compared with non-irradiated HSCs. Using this novel NSG transplantation model, we will be able to study the effects of low dose in vivo X-ray exposure on the long-term fate of HSCs, without the requirement of prior radio-ablation of the recipient, and thus leaving the recipient's BM microenvironment uncompromised. In conclusion, we demonstrated for the first time that allogeneic HSCs from a different inbred strain can compete for niches in the BM compartment of NSG mice.
Collapse
Affiliation(s)
- Tom Verbiest
- Cancer Mechanisms and Biomarkers Group, Radiation Effects Department, Centre for Radiation, Chemical and Environmental Hazards, Public Health England, Didcot OX11 ORQ, UK.
- CRUK & MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford OX3 7DQ, UK.
| | - Rosemary Finnon
- Cancer Mechanisms and Biomarkers Group, Radiation Effects Department, Centre for Radiation, Chemical and Environmental Hazards, Public Health England, Didcot OX11 ORQ, UK.
| | - Natalie Brown
- Cancer Mechanisms and Biomarkers Group, Radiation Effects Department, Centre for Radiation, Chemical and Environmental Hazards, Public Health England, Didcot OX11 ORQ, UK.
| | - Paul Finnon
- Cancer Mechanisms and Biomarkers Group, Radiation Effects Department, Centre for Radiation, Chemical and Environmental Hazards, Public Health England, Didcot OX11 ORQ, UK.
| | - Simon Bouffler
- Cancer Mechanisms and Biomarkers Group, Radiation Effects Department, Centre for Radiation, Chemical and Environmental Hazards, Public Health England, Didcot OX11 ORQ, UK.
| | - Christophe Badie
- Cancer Mechanisms and Biomarkers Group, Radiation Effects Department, Centre for Radiation, Chemical and Environmental Hazards, Public Health England, Didcot OX11 ORQ, UK.
| |
Collapse
|
16
|
Shaaban S, Alsulami M, Arbab SA, Ara R, Shankar A, Iskander A, Angara K, Jain M, Bagher-Ebadian H, Achyut BR, Arbab AS. Targeting Bone Marrow to Potentiate the Anti-Tumor Effect of Tyrosine Kinase Inhibitor in Preclinical Rat Model of Human Glioblastoma. ACTA ACUST UNITED AC 2016; 12:69-81. [PMID: 27429653 DOI: 10.3923/ijcr.2016.69.81] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Antiangiogenic agents caused paradoxical increase in pro-growth and pro-angiogenic factors and caused tumor growth in glioblastoma (GBM). It is hypothesized that paradoxical increase in pro-angiogenic factors would mobilize Bone Marrow Derived Cells (BMDCs) to the treated tumor and cause refractory tumor growth. The purposes of the studies were to determine whether whole body irradiation (WBIR) or a CXCR4 antagonist (AMD3100) will potentiate the effect of vatalanib (a VEGFR2 tyrosine kinase inhibitor) and prevent the refractory growth of GBM. Human GBM were grown orthotopically in three groups of rats (control, pretreated with WBIR and AMD3100) and randomly selected for vehicle or vatalanib treatments for 2 weeks. Then all animals underwent Magnetic Resonance Imaging (MRI) followed by euthanasia and histochemical analysis. Tumor volume and different vascular parameters (plasma volume (vp), forward transfer constant (Ktrans), back flow constant (kep), extravascular extracellular space volume (ve) were determined from MRI. In control group, vatalanib treatment increased the tumor growth significantly compared to that of vehicle treatment but by preventing the mobilization of BMDCs and interaction of CXCR4-SDF-1 using WBIR and ADM3100, respectively, paradoxical growth of tumor was controlled. Pretreatment with WBIR or AMD3100 also decreased tumor cell migration, despite the fact that ADM3100 increased the accumulation of M1 and M2 macrophages in the tumors. Vatalanib also increased Ktrans and ve in control animals but both of the vascular parameters were decreased when the animals were pretreated with WBIR and AMD3100. In conclusion, depleting bone marrow cells or CXCR4 interaction can potentiate the effect of vatalanib.
Collapse
Affiliation(s)
- S Shaaban
- Laboratory of Tumor Angiogenesis, Department of Biochemistry and Molecular Biology, Cancer Center, Georgia Regents University, Augusta, GA, 30912, USA
| | - M Alsulami
- Laboratory of Tumor Angiogenesis, Department of Biochemistry and Molecular Biology, Cancer Center, Georgia Regents University, Augusta, GA, 30912, USA
| | - S A Arbab
- Laboratory of Tumor Angiogenesis, Department of Biochemistry and Molecular Biology, Cancer Center, Georgia Regents University, Augusta, GA, 30912, USA
| | - R Ara
- Laboratory of Tumor Angiogenesis, Department of Biochemistry and Molecular Biology, Cancer Center, Georgia Regents University, Augusta, GA, 30912, USA
| | - A Shankar
- Laboratory of Tumor Angiogenesis, Department of Biochemistry and Molecular Biology, Cancer Center, Georgia Regents University, Augusta, GA, 30912, USA
| | - A Iskander
- Laboratory of Tumor Angiogenesis, Department of Biochemistry and Molecular Biology, Cancer Center, Georgia Regents University, Augusta, GA, 30912, USA
| | - K Angara
- Laboratory of Tumor Angiogenesis, Department of Biochemistry and Molecular Biology, Cancer Center, Georgia Regents University, Augusta, GA, 30912, USA
| | - M Jain
- Laboratory of Tumor Angiogenesis, Department of Biochemistry and Molecular Biology, Cancer Center, Georgia Regents University, Augusta, GA, 30912, USA
| | - H Bagher-Ebadian
- Department of Radiology, Henry Ford Health System, Detroit, MI, USA
| | - B R Achyut
- Laboratory of Tumor Angiogenesis, Department of Biochemistry and Molecular Biology, Cancer Center, Georgia Regents University, Augusta, GA, 30912, USA
| | - A S Arbab
- Laboratory of Tumor Angiogenesis, Department of Biochemistry and Molecular Biology, Cancer Center, Georgia Regents University, Augusta, GA, 30912, USA
| |
Collapse
|
17
|
Engraftment and bone mass are enhanced by PTHrP 1-34 in ectopically transplanted vertebrae (vossicle model) and can be non-invasively monitored with bioluminescence and fluorescence imaging. Transgenic Res 2015; 24:955-69. [PMID: 26271486 DOI: 10.1007/s11248-015-9901-9] [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: 05/01/2015] [Accepted: 08/04/2015] [Indexed: 10/23/2022]
Abstract
Evidence exists that parathyroid hormone-related protein (PTHrP) 1-34 may be more anabolic in bone than parathyroid hormone 1-34. While optical imaging is growing in popularity, scant information exists on the relationships between traditional bone imaging and histology and bioluminescence (BLI) and fluorescence (FLI) imaging. We aimed to evaluate the effects of PTHrP 1-34 on bone mass and determine if relationships existed between radiographic and histologic findings in bone and BLI and FLI indices. Vertebrae (vossicles) from mice coexpressing luciferase and green fluorescent protein were implanted subcutaneously into allogenic nude mice. Transplant recipients were treated daily with saline or PTHrP 1-34 for 4 weeks. BLI, FLI, radiography, histology, and µCT of the vossicles were performed over time. PTHrP 1-34 increased bioluminescence the most after 2 weeks, fluorescence at all time points, and decreased the time to peak bioluminescence at 4 weeks (P ≤ 0.027), the latter of which suggesting enhanced engraftment. PTHrP 1-34 maximized vertebral body volume at 4 weeks (P < 0.0001). The total amount of bone observed histologically increased in both groups at 2 and 4 weeks (P ≤ 0.002); however, PTHrP 1-34 exceeded time-matched controls (P ≤ 0.044). A positive linear relationship existed between the percentage of trabecular bone and (1) total bioluminescence (r = 0.595; P = 0.019); (2) total fluorescence (r = 0.474; P = 0.074); and (3) max fluorescence (r = 0.587; P = 0.021). In conclusion, PTHrP 1-34 enhances engraftment and bone mass, which can be monitored non-invasively by BLI and FLI.
Collapse
|
18
|
Chandrasekaran D, Nakamoto B, Watts KL, Kiem HP, Papayannopoulou T. Modeling promising nonmyeloablative conditioning regimens in nonhuman primates. Hum Gene Ther 2015; 25:1013-22. [PMID: 24937231 DOI: 10.1089/hum.2014.031] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Minimal conditioning or even no conditioning would be the preferred preparation for most gene therapy applications for nonmalignant diseases. However, reduced intensity conditioning (RIC) regimens in patients with nonhematologic malignancies have not led to long-term engraftment unless a selective advantage was present for the transplanted donor cells. Similar findings have also been observed in a number of large animal studies. Inadequate myelosuppression levels were thought to be responsible for the outcomes. To address this issue several innovative protocols in small animals have been presented with selective hematopoietic myelosuppression and less systemic toxicity. Such protocols promised to curb the transplant-related morbidity and mortality in myeloablative conditioning and provide effective long-term engraftment, especially in patients with gene-corrected autografts. In the present study we have tested some of these promising RIC regimens in nonhuman primates, a clinically relevant large animal model. Our data suggest that transient myelosuppression induced by anti-c-Kit antibody in conjunction with low-dose irradiation may lead to long-term engraftment, albeit at low levels. The animals with busulfan conditioning with or without anti-c-Kit that received gene-modified autologous transplants with green fluorescent protein expression had similar myelosuppression, but failed long-term engraftment and despite immunosuppressive treatment had all the hallmarks seen previously in similar models without immunosuppression. Our preliminary data expand current knowledge of RIC and emphasize the need to explore whether specific and directed myelosuppression alone is adequate in the absence of microenvironmental modulation, or whether innovative combinations are necessary for safe and effective engraftment.
Collapse
Affiliation(s)
- Devikha Chandrasekaran
- 1 Clinical Research Division, Fred Hutchinson Cancer Research Center , Seattle, WA 98109
| | | | | | | | | |
Collapse
|
19
|
Ibrahim M, Widjajanto E, Widodo MA, Sumitro SB. EMSA Eritin Drives Expansion of Regulatory T Cells and Promotes T Cells Differentiation in Irradiated Mice. J Evid Based Complementary Altern Med 2015; 21:171-6. [PMID: 26170134 DOI: 10.1177/2156587215595146] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Accepted: 06/14/2015] [Indexed: 01/30/2023] Open
Abstract
Sublethal irradiation therapy in cancer treatment causes generalized immunosuppression, which results in a range of DNA damage. We examined the significance of a polyherbal medicine called "EMSA Eritin" on immunological responses in sublethally irradiated mice focusing on the involvement of Treg, naïve T cell, and also the development and differentiation of T cells in thymus. Normal BALB/c mice were sublethally irradiated with dose of 600 rad. The irradiated mice were then orally administered by EMSA Eritin once a day at different doses: 1.04, 3.12, 9.37 mg/g body weight. The treatment was performed for 14 days. On day 15, immunological responses were observed by analyzing the status of Treg and differentiation of T cells in thymus. The administration of EMSA Eritin to irradiated mice resulted in a significant increase of pre T cells, Treg cells, and naïve T cells, which in general could maintain and normalize healthy condition in mice.
Collapse
|
20
|
Mouse xenograft modeling of human adult acute lymphoblastic leukemia provides mechanistic insights into adult LIC biology. Blood 2014; 124:96-105. [PMID: 24825861 DOI: 10.1182/blood-2014-01-549352] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The distinct nature of acute lymphoblastic leukemia (ALL) in adults, evidenced by inferior treatment outcome and different genetic landscape, mandates specific studies of disease-initiating mechanisms. In this study, we used NOD/LtSz-scid IL2Rγ null(c) (NSG) mouse xenotransplantation approaches to elucidate leukemia-initiating cell (LIC) biology in primary adult precursor B (pre-B) ALL to optimize disease modeling. In contrast with xenografting studies of pediatric ALL, we found that modification of the NSG host environment using preconditioning total body irradiation (TBI) was indispensable for efficient engraftment of adult non-t(4;11) pre-B ALL, whereas t(4;11) pre-B ALL was successfully reconstituted without this adaptation. Furthermore, TBI-based xenotransplantation of non-t(4;11) pre-B ALL enabled detection of a high frequency of LICs (<1:6900) and permitted frank leukemic engraftment from a remission sample containing drug-resistant minimal residual disease. Investigation of TBI-sensitive stromal-derived factor-1/chemokine receptor type 4 signaling revealed greater functional dependence of non-t(4;11) pre-B ALL on this niche-based interaction, providing a possible basis for the differential engraftment behavior. Thus, our studies establish the optimal conditions for experimental modeling of human adult pre-B ALL and demonstrate the critical protumorogenic role of microenvironment-derived SDF-1 in regulating adult pre-B LIC activity that may present a therapeutic opportunity.
Collapse
|
21
|
Marino R, Otsuru S, Hofmann TJ, Olson TS, Rasini V, Veronesi E, Boyd K, Gaber MW, Martinez C, Paolucci P, Dominici M, Horwitz EM. Delayed marrow infusion in mice enhances hematopoietic and osteopoietic engraftment by facilitating transient expansion of the osteoblastic niche. Biol Blood Marrow Transplant 2013; 19:1566-73. [PMID: 23916672 DOI: 10.1016/j.bbmt.2013.07.025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Accepted: 07/26/2013] [Indexed: 01/15/2023]
Abstract
Transplantation of bone marrow cells leads to engraftment of osteopoietic and hematopoietic progenitors. We sought to determine whether the recently described transient expansion of the host osteoblastic niche after marrow radioablation promotes engraftment of both osteopoietic and hematopoietic progenitor cells. Mice infused with marrow cells 24 hours after total body irradiation (TBI) demonstrated significantly greater osteopoietic and hematopoietic progenitor chimerism than did mice infused at 30 minutes or 6 hours. Irradiated mice with a lead shield over 1 hind limb showed greater hematopoietic chimerism in the irradiated limb than in the shielded limb at both the 6- and 24-hour intervals. By contrast, the osteopoietic chimerism was essentially equal in the 2 limbs at each of these intervals, although it significantly increased when cells were infused 24 hours compared with 6 hours after TBI. Similarly, the number of donor phenotypic long-term hematopoietic stem cells was equivalent in the irradiated and shielded limbs after each irradiation-to-infusion interval but was significantly increased at the 24-hour interval. Our findings indicate that a 24-hour delay in marrow cell infusion after TBI facilitates expansion of the endosteal osteoblastic niche, leading to enhanced osteopoietic and hematopoietic engraftment.
Collapse
Affiliation(s)
- Roberta Marino
- Division of Oncology/Blood and Marrow Transplantation, Children's Hospital of Philadelphia and University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Yang Y, Jorstad NL, Shiao C, Cherne MK, Khademi SB, Montine KS, Montine TJ, Keene CD. Perivascular, but not parenchymal, cerebral engraftment of donor cells after non-myeloablative bone marrow transplantation. Exp Mol Pathol 2013; 95:7-17. [PMID: 23567123 DOI: 10.1016/j.yexmp.2013.03.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Accepted: 03/26/2013] [Indexed: 12/26/2022]
Abstract
Myeloablative (MyA) bone marrow transplantation (BMT) results in robust engraftment of BMT-derived cells in the central nervous system (CNS) and is neuroprotective in diverse experimental models of neurodegenerative diseases of the brain and retina. However, MyA irradiation is associated with significant morbidity and mortality and does not represent a viable therapeutic option for the elderly. Non-myeloablative (NMyA) BMT is less toxic, but it is not known if the therapeutic efficacy observed with MyA BMT is preserved. As a first step to address this important gap in knowledge, we evaluated and compared engraftment characteristics of BMT-derived monocytes/microglia using several clinically relevant NMyA pretransplant conditioning regimens in C57BL/6 mice. These included chemotherapy (fludarabine and cyclophosphamide) with or without 2 Gy irradiation, and 5.5 Gy irradiation alone. Each regimen was followed by transplantation of whole bone marrow from green fluorescent protein-expressing wild type (wt) mice. While stable hematopoietic engraftment occurred, to varying degrees, in all NMyA regimens, only 5.5 Gy irradiation resulted in significant engraftment of BMT-derived cells in the brain, where these cells were exclusively localized to perivascular, leptomeningeal, and related anatomic regions. Engraftment in retina under 5.5 Gy NMyA conditions was significantly reduced compared to MyA, but robust engraftment was identified in the optic nerve. Advancing the therapeutic applications of BMT to neurodegenerative diseases will require identification of the barrier mechanisms that MyA, but not NMyA, BMT is able to overcome.
Collapse
Affiliation(s)
- Yue Yang
- Department of Pathology, University of Washington, Seattle, WA, USA
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Cuddihy AR, Suterwala BT, Ge S, Kohn LA, Jang J, Andrade J, Wang X, Crooks GM. Rapid thymic reconstitution following bone marrow transplantation in neonatal mice is VEGF-dependent. Biol Blood Marrow Transplant 2012; 18:683-9. [PMID: 22281302 DOI: 10.1016/j.bbmt.2012.01.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2011] [Accepted: 01/10/2012] [Indexed: 12/27/2022]
Abstract
Age-related differences in thymic function influence the rapidity of T cell reconstitution following hematopoietic stem cell transplantation (HSCT). In adults, thymic reconstitution is delayed until after marrow engraftment is established, and is significantly improved by approaches that increase marrow chimerism, such as pretransplantation irradiation. In contrast, we show that neonatal mice undergo more rapid and efficient thymic reconstitution than adults, even when bone marrow (BM) engraftment is minimal and in the absence of pretransplantation radiation. We have previously shown that the neonatal thymus produces high levels of vascular endothelial growth factor (VEGF) that drives angiogenesis locally. In this report, we show that inhibition of VEGF prior to HSCT prevents rapid thymic reconstitution in neonates, but has no effect on thymic reconstitution in adults. These data suggest that the early radiation-independent thymic reconstitution unique to the neonatal host is mediated through VEGF, and reveals a novel pathway that might be targeted to improve immune reconstitution post-HSCT.
Collapse
Affiliation(s)
- Andrew R Cuddihy
- Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, California, USA
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Jiang H, Tu H, Chen Z, Chen R, Wang Y, Wang M, Jin J, Feng S, Chen W, Bi Y, Wang H, Mao Y, Shou Z, Chen J. Effects of chimerism on the mice heart transplanted survival with the bone marrow infusion. Transpl Immunol 2011; 25:202-6. [PMID: 21911062 DOI: 10.1016/j.trim.2011.08.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2011] [Revised: 08/07/2011] [Accepted: 08/20/2011] [Indexed: 11/19/2022]
Abstract
AIMS To evaluate the effects of chimerism on the mice heart transplanted survival with the bone marrow infusion. METHODS Bone marrow cells (BMCs) were obtained from BALB/c mice. These BMCs were injected into the irradiated (2Gy-Co60) C57BL/6 mice through femoral vein. Then Group A mice were treated with Cyclosporine (1mg/kg) for 21days and Group B were not treated with Cyclosporine. Group C were treated as the control group without BMCs infusion. Group D were treated with Cyclosporine (1mg/kg) for 21days pre-hearttransplantation without BMCs infusion. After 21days, the C57BL/6 mice received heart allografts from BALB/c. To determine the degree of chimerism in BMCs infusion recipients, peripheral blood were isolated on day 7, 14, 21. Allografts were harvested 10days after heart transplantation for the histological analysis. RESULTS (1) Chimerism detected in the peripheral blood of Group A mice on day 7 after BMCs infusion was 6.1±2.5%, on day 14 was 15.4±2.9% and on day 21 was 10.7±2.6%. For the Group B mice on day 7 after BMCs infusion, the chimerism was 2.8±1.1%, on day 14 was 11.2±4.8% and on day 21 was 7.4±3.7%. For the Groups C and D mice, no chimerism was observed. Group A mice had the tendency toward improved level of chimerism than Group B mice. (2) The survival time of Group A (n=6) was 13.0±1.4days which was significantly longer than Group B (n=6) with the survival time was 8.5±1.3days (p<0.001), also longer than the mice in Groups C and D, the survival time of which were 10.0±1.3days (p=0.008) and 9.4±1.1days (p=0.004). There is no significant difference among Groups B, C, and D. (3) The HE staining showed the much more seriously heart rejection in Groups B, C and D than Group A. CONCLUSIONS The chimerism was found in the BMCs infusion groups. Without the CsA treatment combined with chimerism could not protect the transplanted heart. There was no obvious evidence showed that the chimerism alone could improve the survival time of cardiac allografts in mice.
Collapse
Affiliation(s)
- Hong Jiang
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, 310003, PR China
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|