Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Cryobiology ; 113: 104592, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37827209

RESUMO

Clinical development of cellular therapies, including mesenchymal stem/stromal cell (MSC) treatments, has been hindered by ineffective cryopreservation methods that result in substantial loss of post-thaw cell viability and function. Proposed solutions to generate high potency MSC for clinical testing include priming cells with potent cytokines such as interferon gamma (IFNγ) prior to cryopreservation, which has been shown to enhance post-thaw function, or briefly culturing to allow recovery from cryopreservation injury prior to administering to patients. However, both solutions have disadvantages: cryorecovery increases the complexity of manufacturing and distribution logistics, while the pleiotropic effects of IFNγ may have uncharacterized and unintended consequences on MSC function. To determine specific cellular functions impacted by cryoinjury, we first evaluated cell cycle status. It was discovered that S phase MSC are exquisitely sensitive to cryoinjury, demonstrating heightened levels of delayed apoptosis post-thaw and reduced immunomodulatory function. Blocking cell cycle progression at G0/G1 by growth factor deprivation (commonly known as serum starvation) greatly reduced post-thaw dysfunction of MSC by preventing apoptosis induced by double-stranded breaks in labile replicating DNA that form during the cryopreservation and thawing processes. Viability, clonal growth and T cell suppression function were preserved at pre-cryopreservation levels and were no different than cells prior to freezing or frozen after priming with IFNγ. Thus, we have developed a robust and effective strategy to enhance post-thaw recovery of therapeutic MSC.


Assuntos
Criopreservação , Linfócitos T , Humanos , Congelamento , Criopreservação/métodos , Proliferação de Células , Ciclo Celular , Sobrevivência Celular
2.
Cryobiology ; 112: 104558, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37451668

RESUMO

The ability to cryopreserve bone marrow within the vertebral body (VB) would offer significant clinical and research benefits. However, cryopreservation of large structures, such as VBs, is challenging due to mass transport limitations that prevent the effective delivery of cryoprotectants into the tissue. To overcome this challenge, we examined the potential of vacuum infiltration, along with carbonation, to increase the penetration of cryoprotectants. In particular, we hypothesized that initial exposure to high-pressure carbon dioxide gas would introduce bubbles into the tissue and that subsequent vacuum cycling would cause expansion and contraction of the bubbles, thus enhancing the transport of cryoprotectant into the tissue. Experiments were carried out using colored dye and agarose gel as a model revealing that carbonation and vacuum cycling result in a 14% increase in dye penetration compared to the atmospheric controls. Experiments were also carried out by exposing VBs isolated from human vertebrae to 40% (v/v) DMSO solution. CT imaging showed the presence of gas bubbles within the tissue pores for carbonated VBs as well as control VBs. Vacuum cycling reduced the bubble volume by more than 50%, most likely resulting in replacement of this volume with DMSO solution. However, we were unable to detect a statistically significant increase in DMSO concentration within the VBs using CT imaging. This research suggests that there may be a modest benefit to carbonation and vacuum cycling for introduction of cryoprotectants into larger structures, like VBs.


Assuntos
Criopreservação , Dimetil Sulfóxido , Humanos , Criopreservação/métodos , Vácuo , Crioprotetores/farmacologia
3.
Transplant Cell Ther ; 29(2): 95.e1-95.e10, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36402456

RESUMO

Despite the readily available graft sources for allogeneic hematopoietic cell transplantation (alloHCT), a significant unmet need remains in the timely provision of suitable unrelated donor grafts. This shortage is related to the rarity of certain HLA alleles in the donor pool, nonclearance of donors owing to infectious disease or general health status, and prolonged graft procurement and processing times. An alternative hematopoietic progenitor cell (HPC) graft source obtained from the vertebral bodies (VBs) of deceased organ donors could alleviate many of the obstacles associated with using grafts from healthy living donors or umbilical cord blood (UCB). Deceased organ donor-derived bone marrow (BM) can be preemptively screened, cryogenically banked for on-demand use, and made available in adequate cell doses for HCT. We have developed a good manufacturing practice (GMP)-compliant process to recover and cryogenically bank VB-derived HPCs from deceased organ donor (OD) BM. Here we present results from an analysis of HPCs from BM obtained from 250 deceased donors to identify any substantial difference in composition or quality compared with HPCs from BM aspirated from the iliac crests of healthy living donors. BM from deceased donor VBs was processed in a central GMP facility and packaged for cryopreservation in 5% DMSO/2.5% human serum albumin. BM aspirated from living donor iliac crests was obtained and used for comparison. A portion of each specimen was analyzed before and after cryopreservation by flow cytometry and colony-forming unit potential. Bone marrow chimerism potential was assessed in irradiated immunocompromised NSG mice. Analysis of variance with Bonferroni correction for multiple comparisons was used to determine how cryopreservation affects BM cells and to evaluate indicators of successful engraftment of BM cells into irradiated murine models. The t test (with 95% confidence intervals [CIs]) was used to compare cells from deceased donors and living donors. A final dataset of complete clinical and matched laboratory data from 226 cryopreserved samples was used in linear regressions to predict outcomes of BM HPC processing. When compared before and after cryopreservation, OD-derived BM HPCs were found to be stable, with CD34+ cells maintaining high viability and function after thawing. The yield from a single donor is sufficient for transplantation of an average of 1.6 patients (range, 1.2 to 7.5). CD34+ cells from OD-derived HPCs from BM productively engrafted sublethally irradiated immunocompromised mouse BM (>44% and >67% chimerism at 8 and 16 weeks, respectively). Flow cytometry and secondary transplantation confirmed that OD HPCs from BM is composed of long-term engrafting CD34+CD38-CD45RA-CD90+CD49f+ HSCs. Linear regression identified no meaningful predictive associations between selected donor-related characteristics and OD BM HPC quality or yield. Collectively, these data demonstrate that cryopreserved BM HPCs from deceased organ donors is potent and functionally equivalent to living donor BM HPCs and is a viable on-demand graft source for clinical HCT. Prospective clinical trials will soon commence in collaboration with the Center for International Blood and Marrow Research to assess the feasibility, safety, and efficacy of Ossium HPCs from BM (ClinicalTrials.gov identifier NCT05068401).


Assuntos
Medula Óssea , Transplante de Células-Tronco Hematopoéticas , Humanos , Animais , Camundongos , Estudos Prospectivos , Transplante de Células-Tronco Hematopoéticas/métodos , Criopreservação/métodos , Doadores Vivos
4.
Front Immunol ; 12: 622604, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33732244

RESUMO

Induction of immune tolerance for solid organ and vascular composite allografts is the Holy Grail for transplantation medicine. This would obviate the need for life-long immunosuppression which is associated with serious adverse outcomes, such as infections, cancers, and renal failure. Currently the most promising means of tolerance induction is through establishing a mixed chimeric state by transplantation of donor hematopoietic stem cells; however, with the exception of living donor renal transplantation, the mixed chimerism approach has not achieved durable immune tolerance on a large scale in preclinical or clinical trials with other solid organs or vascular composite allotransplants (VCA). Ossium Health has established a bank of cryopreserved bone marrow (BM), termed "hematopoietic progenitor cell (HPC), Marrow," recovered from deceased organ donor vertebral bodies. This new source for hematopoietic cell transplant will be a valuable resource for treating hematological malignancies as well as for inducing transplant tolerance. In addition, we have discovered and developed a large source of mesenchymal stem (stromal) cells (MSC) tightly associated with the vertebral body bone fragment byproduct of the HPC, Marrow recovery process. Thus, these vertebral bone adherent MSC (vBA-MSC) are matched to the banked BM obtained from each donor, as opposed to third-party MSC, which enhances safety and potentially efficacy. Isolation and characterization of vBA-MSC from over 30 donors has demonstrated that the cells are no different than traditional BM-MSC; however, their abundance is >1,000-fold higher than obtainable from living donor BM aspirates. Based on our own unpublished data as well as reports published by others, MSC facilitate chimerism, especially at limiting hematopoietic stem and progenitor cell (HSPC) numbers and increase safety by controlling and/or preventing graft-vs.-host-disease (GvHD). Thus, vBA-MSC have the potential to facilitate mixed chimerism, promote complementary peripheral immunomodulatory functions and increase safety of BM infusions. Both HPC, Marrow and vBA-MSC have potential use in current VCA and solid organ transplant (SOT) tolerance clinical protocols that are amenable to "delayed tolerance." Current trials with HPC, Marrow are planned with subsequent phases to include vBA-MSC for tolerance of both VCA and SOT.


Assuntos
Bancos de Espécimes Biológicos , Células da Medula Óssea/imunologia , Transplante de Medula Óssea , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/imunologia , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/imunologia , Doadores de Tecidos , Tolerância ao Transplante , Animais , Transplante de Medula Óssea/efeitos adversos , Seleção do Doador , Rejeição de Enxerto/imunologia , Rejeição de Enxerto/prevenção & controle , Sobrevivência de Enxerto , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Histocompatibilidade , Humanos , Imunossupressores/uso terapêutico , Transplante de Células-Tronco Mesenquimais/efeitos adversos , Fenótipo , Quimeras de Transplante/imunologia , Resultado do Tratamento
5.
Cytotherapy ; 22(11): 617-628, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32873509

RESUMO

BACKGROUND: Therapeutic allogeneic mesenchymal stromal cells (MSCs) are currently in clinical trials to evaluate their effectiveness in treating many different disease indications. Eventual commercialization for broad distribution will require further improvements in manufacturing processes to economically manufacture MSCs at scales sufficient to satisfy projected demands. A key contributor to the present high cost of goods sold for MSC manufacturing is the need to create master cell banks from multiple donors, which leads to variability in large-scale manufacturing runs. Therefore, the availability of large single donor depots of primary MSCs would greatly benefit the cell therapy market by reducing costs associated with manufacturing. METHODS: We have discovered that an abundant population of cells possessing all the hallmarks of MSCs is tightly associated with the vertebral body (VB) bone matrix and only liberated by proteolytic digestion. Here we demonstrate that these vertebral bone-adherent (vBA) MSCs possess all the International Society of Cell and Gene Therapy-defined characteristics (e.g., plastic adherence, surface marker expression and trilineage differentiation) of MSCs, and we have therefore termed them vBA-MSCs to distinguish this population from loosely associated MSCs recovered through aspiration or rinsing of the bone marrow compartment. RESULTS: Pilot banking and expansion were performed with vBA-MSCs obtained from 3 deceased donors, and it was demonstrated that bank sizes averaging 2.9 × 108 ± 1.35 × 108 vBA-MSCs at passage 1 were obtainable from only 5 g of digested VB bone fragments. Each bank of cells demonstrated robust proliferation through a total of 9 passages, without significant reduction in population doubling times. The theoretical total cell yield from the entire amount of bone fragments (approximately 300 g) from each donor with limited expansion through 4 passages is 100 trillion (1 × 1014) vBA-MSCs, equating to over 105 doses at 10 × 106 cells/kg for an average 70-kg recipient. DISCUSSION: Thus, we have established a novel and plentiful source of MSCs that will benefit the cell therapy market by overcoming manufacturing and regulatory inefficiencies due to donor-to-donor variability.


Assuntos
Células da Medula Óssea/citologia , Técnicas de Cultura de Células/métodos , Células-Tronco Mesenquimais/citologia , Corpo Vertebral/citologia , Adolescente , Adulto , Antígenos de Superfície/metabolismo , Adesão Celular , Diferenciação Celular , Linhagem da Célula , Proliferação de Células , Células Cultivadas , Ensaio de Unidades Formadoras de Colônias , Feminino , Humanos , Ativação Linfocitária/imunologia , Masculino , Fenótipo , Linfócitos T/imunologia , Adulto Jovem
6.
J Transl Med ; 18(1): 300, 2020 08 05.
Artigo em Inglês | MEDLINE | ID: mdl-32758261

RESUMO

BACKGROUND: Deceased organ donors represent an untapped source of therapeutic bone marrow (BM) that can be recovered in 3-5 times the volume of that obtained from living donors, tested for quality, cryopreserved, and banked indefinitely for future on-demand use. A challenge for a future BM banking system will be to manage the prolonged ischemia times that are inevitable when bones procured at geographically-dispersed locations are shipped to distant facilities for processing. Our objectives were to: (a) quantify, under realistic field conditions, the relationship between ischemia time and the quality of hematopoietic stem and progenitor cells (HSPCs) derived from deceased-donor BM; (b) identify ischemia-time boundaries beyond which HSPC quality is adversely affected; (c) investigate whole-body cooling as a strategy for preserving cell quality; and (d) investigate processing experience as a variable affecting quality. METHODS: Seventy-five bones from 62 donors were analyzed for CD34+ viability following their exposure to various periods of warm-ischemia time (WIT), cold-ischemia time (CIT), and body-cooling time (BCT). Regression models were developed to quantify the independent associations of WIT, CIT, and BCT, with the viability and function of recovered HSPCs. RESULTS: Results demonstrate that under "real-world" scenarios: (a) combinations of warm- and cold-ischemia times favorable to the recovery of high-quality HSPCs are achievable (e.g., CD34+ cell viabilities in the range of 80-90% were commonly observed); (b) body cooling prior to bone recovery is detrimental to cell viability (e.g., CD34+ viability < 73% with, vs. > 89% without body cooling); (c) vertebral bodies (VBs) are a superior source of HSPCs compared to ilia (IL) (e.g., %CD34+ viability > 80% when VBs were the source, vs. < 74% when IL were the source); and (d) processing experience is a critical variable affecting quality. CONCLUSIONS: Our models can be used by an emerging BM banking system to formulate ischemia-time tolerance limits and data-driven HSPC quality-acceptance standards.


Assuntos
Medula Óssea , Doadores de Tecidos , Antígenos CD34 , Células da Medula Óssea , Transplante de Medula Óssea , Células-Tronco Hematopoéticas , Humanos , Isquemia
7.
Artigo em Inglês | MEDLINE | ID: mdl-30198519

RESUMO

AIM: The therapeutic potential of adipose-derived stem cell conditioned medium (ASC-CM) was studied in the rabbit model of critical limb ischemia (CLI). METHODS: Rabbits received treatment with ASC-CM or placebo. Gastrocnemius muscle tissue was collected 35 days after ischemia induction. Ischemic changes were evaluated in hematoxylin-eosin stained tissues for early (necrotic lesions/granulation tissue) and late (fibrous scars) phases of tissue repair. The expression of proangiogenic miR-126 was also evaluated using in situ hybridization. The levels of cytokines, insulin, and C-peptide were measured in blood. RESULTS: Early repair phases were observed more often in placebo-treated samples (45.5%) than in ASC-CM-treated ones (22.2%). However, the difference was not statistically significant. We demonstrated a statistically significant positive correlation between the early healing phases in tissue samples and C-peptide levels in peripheral blood. The expression of proangiogenic miR-126 was also shown in a number of structures in all phases of ischemic tissue healing. CONCLUSION: Based on our results, we believe that treatment with ASC-CM has the potential to accelerate the healing process in ischemic tissues in the rabbit model of CLI. The whole healing process was accompanied by miR-126 tissue expression. C-peptide could be used to monitor the course of the tissue healing process.


Assuntos
Peptídeo C/sangue , Citocinas/sangue , Diabetes Mellitus Experimental/sangue , Insulina/sangue , Isquemia/sangue , Células-Tronco Mesenquimais , Músculo Esquelético/patologia , Cicatrização/fisiologia , Adulto , Animais , Cicatriz/patologia , Meios de Cultivo Condicionados/farmacologia , Pé Diabético , Modelos Animais de Doenças , Fibrose , Tecido de Granulação/patologia , Membro Posterior , Humanos , Hibridização In Situ , Masculino , MicroRNAs/metabolismo , Músculo Esquelético/irrigação sanguínea , Músculo Esquelético/efeitos dos fármacos , Necrose , Neovascularização Fisiológica/efeitos dos fármacos , Coelhos , Cicatrização/efeitos dos fármacos
8.
Med Sci Monit ; 24: 511-522, 2018 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-29371587

RESUMO

BACKGROUND Paracrine factors secreted by adipose-derived stem cells can be captured, fractionated, and concentrated to produce therapeutic factor concentrate (TFC). The present study examined whether TFC effects could be enhanced by combining TFC with a biological matrix to provide sustained release of factors in the target region. MATERIAL AND METHODS Unilateral hind limb ischemia was induced in rabbits. Ischemic limbs were injected with either placebo control, TFC, micronized small intestinal submucosa tissue (SIS), or TFC absorbed to SIS. Blood flow in both limbs was assessed with laser Doppler perfusion imaging. Tissues harvested at Day 48 were assessed immunohistochemically for vessel density; in situ hybridization and quantitative real-time PCR were employed to determine miR-126 expression. RESULTS LDP ratios were significantly elevated, compared to placebo control, on day 28 in all treatment groups (p=0.0816, p=0.0543, p=0.0639, for groups 2-4, respectively) and on day 36 in the TFC group (p=0.0866). This effect correlated with capillary density in the SIS and TFC+SIS groups (p=0.0093 and p=0.0054, respectively, compared to placebo). A correlation was observed between miR-126 levels and LDP levels at 48 days in SIS and TFC+SIS groups. CONCLUSIONS A single bolus administration of TFC and SIS had early, transient effects on reperfusion and promotion of ischemia repair. The effects were not additive. We also discovered that TFC modulated miR-126 levels that were expressed in cell types other than endothelial cells. These data suggested that TFC, alone or in combination with SIS, may be a potent therapy for patients with CLI that are at risk of amputation.


Assuntos
Tecido Adiposo/citologia , Micropartículas Derivadas de Células/metabolismo , Matriz Extracelular/metabolismo , Extremidades/irrigação sanguínea , Isquemia/terapia , MicroRNAs/metabolismo , Transplante de Células-Tronco , Células-Tronco/citologia , Animais , Modelos Animais de Doenças , Extremidades/patologia , Feminino , Regulação da Expressão Gênica , Humanos , Mucosa Intestinal/fisiologia , Intestino Delgado/fisiologia , Isquemia/genética , Isquemia/patologia , Fluxometria por Laser-Doppler , MicroRNAs/genética , Pessoa de Meia-Idade , Perfusão , Coelhos , Traumatismo por Reperfusão/patologia , Traumatismo por Reperfusão/terapia , Pele/patologia
9.
Am Heart J ; 179: 142-50, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27595689

RESUMO

BACKGROUND: Although several preclinical studies have shown that bone marrow cell (BMC) transplantation promotes cardiac recovery after myocardial infarction, clinical trials with unfractionated bone marrow have shown variable improvements in cardiac function. METHODS: To determine whether in a population of post-myocardial infarction patients, functional recovery after BM transplant is associated with specific BMC subpopulation, we examined the association between BMCs with left ventricular (LV) function in the LateTIME-CCTRN trial. RESULTS: In this population, we found that older individuals had higher numbers of BM CD133(+) and CD3(+) cells. Bone marrow from individuals with high body mass index had lower CD45(dim)/CD11b(dim) levels, whereas those with hypertension and higher C-reactive protein levels had higher numbers of CD133(+) cells. Smoking was associated with higher levels of CD133(+)/CD34(+)/VEGFR2(+) cells and lower levels of CD3(+) cells. Adjusted multivariate analysis indicated that CD11b(dim) cells were negatively associated with changes in LV ejection fraction and wall motion in both the infarct and border zones. Change in LV ejection fraction was positively associated with CD133(+), CD34(+), and CD45(+)/CXCR4(dim) cells as well as faster BMC growth rates in endothelial colony forming assays. CONCLUSIONS: In the LateTIME population, BM composition varied with patient characteristics and treatment. Irrespective of cell therapy, recovery of LV function was greater in patients with greater BM abundance of CD133(+) and CD34(+) cells and worse in those with higher levels of CD11b(dim) cells. Bone marrow phenotype might predict clinical response before BMC therapy and administration of selected BM constituents could potentially improve outcomes of other future clinical trials.


Assuntos
Transplante de Medula Óssea , Infarto do Miocárdio/terapia , Recuperação de Função Fisiológica , Disfunção Ventricular Esquerda/terapia , Antígeno AC133/metabolismo , Adulto , Idoso , Antígenos CD34/metabolismo , Índice de Massa Corporal , Células da Medula Óssea/metabolismo , Proteína C-Reativa/metabolismo , Antígeno CD11b/metabolismo , Estudos de Coortes , Feminino , Humanos , Hipertensão/metabolismo , Antígenos Comuns de Leucócito/metabolismo , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Infarto do Miocárdio/diagnóstico por imagem , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/fisiopatologia , Obesidade/metabolismo , Estudos Prospectivos , Receptores CXCR4/metabolismo , Fumar/metabolismo , Disfunção Ventricular Esquerda/diagnóstico por imagem , Disfunção Ventricular Esquerda/metabolismo , Disfunção Ventricular Esquerda/fisiopatologia , Função Ventricular Esquerda
10.
Oncotarget ; 7(24): 36829-36841, 2016 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-27167204

RESUMO

Adipose tissue stroma contains a population of mesenchymal stem cells (MSC) promote new blood vessel formation and stabilization. These adipose-derived stem cells (ASC) promote de novo formation of vascular structures in vitro. We investigated the angiogenic factors secreted by ASC and discovered that glial-derived neurotrophic factor (GDNF) is a key mediator for endothelial cell network formation. It was found that both GDNF alone or present in ASC-conditioned medium (ASC-CM) stimulated capillary network formation by using human umbilical vein endothelial cells (HUVECs) and such an effect was totally independent of vascular endothelial growth factor (VEGF) activity. Additionally, we showed stimulation of capillary network formation by GDNF, but not VEGF, could be blocked by the Ret (rearranged during transfection) receptor antagonist RPI-1, a GDNF signaling inhibitor. Furthermore, GDNF were found to be overexpressed in cancer cells that were resistant to the anti-angiogenic treatment using the VEGF antibody. Cancer cells in the liver hepatocellular carcinoma (HCC), a non-nervous related cancer, highly overexpressed GDNF as compared to normal liver cells. Our data strongly suggest that, in addition to VEGF, GDNF secreted by ASC and HCC cells, may be another important factor promoting pathological neovascularization. Thus, GDNF may be a potential therapeutic target for HCC and obesity treatments.


Assuntos
Tecido Adiposo/citologia , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Neovascularização Patológica/metabolismo , Adipócitos/metabolismo , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Células-Tronco Mesenquimais/metabolismo , Fator A de Crescimento do Endotélio Vascular
12.
Cell Transplant ; 25(9): 1675-1687, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26590374

RESUMO

In the current study, we sought to identify bone marrow-derived mononuclear cell (BM-MNC) subpopulations associated with a combined improvement in left ventricular ejection fraction (LVEF), left ventricular end-systolic volume (LVESV), and maximal oxygen consumption (VO2 max) in patients with chronic ischemic cardiomyopathy 6 months after receiving transendocardial injections of autologous BM-MNCs or placebo. For this prospectively planned analysis, we conducted an embedded cohort study comprising 78 patients from the FOCUS-Cardiovascular Cell Therapy Research Network (CCTRN) trial. Baseline BM-MNC immunophenotypes and progenitor cell activity were determined by flow cytometry and colony-forming assays, respectively. Previously stable patients who demonstrated improvement in LVEF, LVESV, and VO2 max during the 6-month course of the FOCUS-CCTRN study (group 1, n = 17) were compared to those who showed no change or worsened in one to three of these endpoints (group 2, n = 61) and to a subset of patients from group 2 who declined in all three functional endpoints (group 2A, n = 11). Group 1 had higher frequencies of B-cell and CXCR4+ BM-MNC subpopulations at study baseline than group 2 or 2A. Furthermore, patients in group 1 had fewer endothelial colony-forming cells and monocytes/macrophages in their bone marrow than those in group 2A. To our knowledge, this is the first study to show that in patients with ischemic cardiomyopathy, certain bone marrow-derived cell subsets are associated with improvement in LVEF, LVESV, and VO2 max at 6 months. These results suggest that the presence of both progenitor and immune cell populations in the bone marrow may influence the natural history of chronic ischemic cardiomyopathy-even in stable patients. Thus, it may be important to consider the bone marrow composition and associated regenerative capacity of patients when assigning them to treatment groups and evaluating the results of cell therapy trials.


Assuntos
Células-Tronco/citologia , Disfunção Ventricular Esquerda/terapia , Transplante de Medula Óssea , Terapia Baseada em Transplante de Células e Tecidos , Ensaios Clínicos como Assunto , Feminino , Insuficiência Cardíaca/fisiopatologia , Insuficiência Cardíaca/terapia , Humanos , Imunofenotipagem , Masculino , Pessoa de Meia-Idade , Isquemia Miocárdica/fisiopatologia , Isquemia Miocárdica/terapia , Estudos Prospectivos , Volume Sistólico/fisiologia , Disfunção Ventricular Esquerda/fisiopatologia , Função Ventricular Esquerda/fisiologia
13.
Cell Transplant ; 25(9): 1623-1633, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26525042

RESUMO

Transplantation of adipose-derived stem cells (ADSCs) is an emerging therapeutic option for addressing intractable diseases such as critical limb ischemia (CLI). Evidence suggests that therapeutic effects of ADSCs are primarily mediated through paracrine mechanisms rather than transdifferentiation. These secreted factors can be captured in conditioned medium (CM) and concentrated to prepare a therapeutic factor concentrate (TFC) composed of a cocktail of beneficial growth factors and cytokines that individually and in combination demonstrate disease-modifying effects. The ability of a TFC to promote reperfusion in a rabbit model of CLI was evaluated. A total of 27 adult female rabbits underwent surgery to induce ischemia in the left hindlimb. An additional five rabbits served as sham controls. One week after surgery, the ischemic limbs received intramuscular injections of either (1) placebo (control medium), (2) a low dose of TFC, or (3) a high dose of TFC. Limb perfusion was serially assessed with a Doppler probe. Blood samples were analyzed for growth factors and cytokines. Tissue was harvested postmortem on day 35 and assessed for capillary density by immunohistochemistry. At 1 month after treatment, tissue perfusion in ischemic limbs treated with a high dose of TFC was almost double (p < 0.05) that of the placebo group [58.8 ± 23 relative perfusion units (RPU) vs. 30.7 ± 13.6 RPU; mean ± SD]. This effect was correlated with greater capillary density in the affected tissues and with transiently higher serum levels of the angiogenic and prosurvival factors vascular endothelial growth factor (VEGF) and hepatocyte growth factor (HGF). The conclusions from this study are that a single bolus administration of TFC demonstrated robust effects for promoting tissue reperfusion in a rabbit model of CLI and that a possible mechanism of revascularization was promotion of angiogenesis by TFC. Results of this study demonstrate that TFC represents a potent therapeutic cocktail for patients with CLI, many of whom are at risk for amputation of the affected limb.


Assuntos
Tecido Adiposo/metabolismo , Membro Posterior/patologia , Isquemia/tratamento farmacológico , Animais , Células Cultivadas , Meios de Cultivo Condicionados/farmacologia , Citocinas/uso terapêutico , Feminino , Citometria de Fluxo , Fator de Crescimento de Hepatócito/metabolismo , Humanos , Injeções Intramusculares , Peptídeos e Proteínas de Sinalização Intercelular/uso terapêutico , Células-Tronco Mesenquimais/metabolismo , Neovascularização Fisiológica/efeitos dos fármacos , Coelhos , Fator A de Crescimento do Endotélio Vascular/metabolismo
14.
Sci Rep ; 5: 16953, 2015 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-26586020

RESUMO

Adipose stromal cells (ASC) secrete various trophic factors that assist in the protection of neurons in a variety of neuronal death models. In this study, we tested the effects of human ASC conditional medium (ASC-CM) in human amyotrophic lateral sclerosis (ALS) transgenic mouse model expressing mutant superoxide dismutase (SOD1(G93A)). Treating symptomatic SOD1(G93A) mice with ASC-CM significantly increased post-onset survival time and lifespan. Moreover, SOD1(G93A) mice given ASC-CM treatment showed high motor neuron counts, less activation of microglia and astrocytes at an early symptomatic stage in the spinal cords under immunohistochemical analysis. SOD1(G93A) mice treated with ASC-CM for 7 days showed reduced levels of phosphorylated p38 (pp38) in the spinal cord, a mitogen-activated protein kinase that is involved in both inflammation and neuronal death. Additionally, the levels of α-II spectrin in spinal cords were also inhibited in SOD1(G93A) mice treated with ASC-CM for 3 days. Interestingly, nerve growth factor (NGF), a neurotrophic factor found in ASC-CM, played a significant role in the protection of neurodegeneration inSOD1(G93A) mouse. These results indicate that ASC-CM has the potential to develop into a novel and effective therapeutic treatment for ALS.


Assuntos
Tecido Adiposo/citologia , Esclerose Lateral Amiotrófica/tratamento farmacológico , Meios de Cultivo Condicionados/farmacologia , Modelos Animais de Doenças , Células-Tronco/fisiologia , Esclerose Lateral Amiotrófica/genética , Esclerose Lateral Amiotrófica/patologia , Animais , Astrócitos/metabolismo , Astrócitos/patologia , Western Blotting , Antígeno CD11b/metabolismo , Proteína Glial Fibrilar Ácida/metabolismo , Humanos , Imuno-Histoquímica , Camundongos Transgênicos , Microglia/metabolismo , Microglia/patologia , Neurônios Motores/metabolismo , Neurônios Motores/patologia , Fosforilação/efeitos dos fármacos , Mutação Puntual , Espectrina/metabolismo , Medula Espinal/efeitos dos fármacos , Medula Espinal/metabolismo , Medula Espinal/patologia , Superóxido Dismutase/genética , Superóxido Dismutase/metabolismo , Análise de Sobrevida , Fatores de Tempo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
15.
Circ Res ; 116(1): 99-107, 2015 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-25406300

RESUMO

RATIONALE: Despite significant interest in bone marrow mononuclear cell (BMC) therapy for ischemic heart disease, current techniques have resulted in only modest benefits. However, selected patients have shown improvements after autologous BMC therapy, but the contributing factors are unclear. OBJECTIVE: The purpose of this study was to identify BMC characteristics associated with a reduction in infarct size after ST-segment-elevation-myocardial infarction. METHODS AND RESULTS: This prospective study comprised patients consecutively enrolled in the CCTRN TIME (Cardiovascular Cell Therapy Research Network Timing in Myocardial Infarction Evaluation) trial who agreed to have their BMCs stored and analyzed at the CCTRN Biorepository. Change in infarct size between baseline (3 days after percutaneous coronary intervention) and 6-month follow-up was measured by cardiac MRI. Infarct-size measurements and BMC phenotype and function data were obtained for 101 patients (mean age, 56.5 years; mean screening ejection fraction, 37%; mean baseline cardiac MRI ejection fraction, 45%). At 6 months, 75 patients (74.3%) showed a reduction in infarct size (mean change, -21.0±17.6%). Multiple regression analysis indicated that infarct size reduction was greater in patients who had a larger percentage of CD31(+) BMCs (P=0.046) and in those with faster BMC growth rates in colony-forming unit Hill and endothelial-colony forming cell functional assays (P=0.033 and P=0.032, respectively). CONCLUSIONS: This study identified BMC characteristics associated with a better clinical outcome in patients with segment-elevation-myocardial infarction and highlighted the importance of endothelial precursor activity in regenerating infarcted myocardium. Furthermore, it suggests that for these patients with segment-elevation-myocardial infarction, myocardial repair was more dependent on baseline BMC characteristics than on whether the patient underwent intracoronary BMC transplantation. CLINICAL TRIAL REGISTRATION INFORMATION URL: http://www.clinicaltrials.gov. Unique identifier: NCT00684021.


Assuntos
Células da Medula Óssea/fisiologia , Transplante de Medula Óssea/métodos , Infarto do Miocárdio/diagnóstico , Infarto do Miocárdio/terapia , Adulto , Idoso , Estudos de Coortes , Método Duplo-Cego , Feminino , Seguimentos , Humanos , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos
16.
Catheter Cardiovasc Interv ; 86(2): E38-48, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24905889

RESUMO

OBJECTIVES: The potential for beneficial effects of adipose-derived stem cells (ASCs) on myocardial perfusion and left ventricular dysfunction in myocardial ischemia (MI) has not been tested following intravenous delivery. METHODS: Surviving pigs following induction of MI were randomly assigned to 1 of 3 different groups: the placebo group (n = 7), the single bolus group (SB) (n = 7, 15 × 10(7) ASCs), or the divided dose group (DD) (n = 7, 5 × 10(7) ASCs/day for three consecutive days). Myocardial perfusion defect area and coronary flow reserve (CFR) were compared during the 28-day follow-up. Also, serial changes in the absolute number of circulating CD4(+) T and CD8(+) T cells were measured. RESULTS: The increases in ejection fraction were significantly greater in both the SB and the DD groups compared to the placebo group (5.4 ± 0.9%, 3.7 ± 0.7%, and -0.4 ± 0.6%, respectively), and the decrease in the perfusion defect area was significantly greater in the SB group than the placebo group (-36.3 ± 1.8 and -11.5 ± 2.8). CFR increased to a greater degree in the SB and the DD groups than in the placebo group (0.9 ± 0.2, 0.8 ± 0.1, and 0.2 ± 0.2, respectively). The circulating number of CD8(+) T cells was significantly greater in the SB and DD groups than the placebo group at day 7 (3,687 ± 317/µL, 3,454 ± 787/µL, and 1,928 ± 457/µL, respectively). The numbers of small vessels were significantly greater in the SB and the DD groups than the placebo group in the peri-infarct area. CONCLUSIONS: Both intravenous SB and DD delivery of ASCs are effective modalities for the treatment of MI in swine. Intravenous delivery of ASCs, with its immunomodulatory and angiogenic effects, is an attractive noninvasive approach for myocardial rescue.


Assuntos
Tecido Adiposo/citologia , Vasos Coronários/fisiopatologia , Microvasos/fisiopatologia , Infarto do Miocárdio/cirurgia , Transplante de Células-Tronco , Função Ventricular Esquerda , Adulto , Animais , Contagem de Linfócito CD4 , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Células Cultivadas , Circulação Coronária , Modelos Animais de Doenças , Feminino , Xenoenxertos , Humanos , Microcirculação , Infarto do Miocárdio/diagnóstico , Infarto do Miocárdio/imunologia , Infarto do Miocárdio/fisiopatologia , Imagem de Perfusão do Miocárdio , Neovascularização Fisiológica , Neurogênese , Recuperação de Função Fisiológica , Volume Sistólico , Sus scrofa , Fatores de Tempo , Complexos Ventriculares Prematuros/fisiopatologia , Complexos Ventriculares Prematuros/prevenção & controle , Adulto Jovem
17.
J Craniofac Surg ; 23(3): 805-11, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22565925

RESUMO

BACKGROUND: Recent clinical trials indicate that human adipose-derived stromal cells (hASCs) have beneficial effects on antiaging and wound healing. This study examined the morphologic changes in photodamaged human organotypic skin culture after treatment of autologous hASCs. METHODS: Abdominal skin flaps were obtained from 8 white females who underwent abdominoplasties with liposuction. The adipose layer was removed and used for hASC isolation. Sections of skin were removed and cultured in serum-free medium. To induce photodamage, some of the skin pieces were irradiated with maximum subcytotoxic doses of UVB (1600 J/m(2)) and UVA (250 J/m(2)). The effects of hASC on skin segments were evaluated by coculture as a feeder layer or by injecting intradermally. Portions of the skin samples were removed for analysis on days 3, 5, 7, and 9 of culture and analyzed histologically for morphology, viability, and proliferation status. RESULTS: Epidermal necrosis of irradiated skin was significantly reduced by the presence of hASCs. Increased parakeratosis was observed at early time points, and apoptosis in epidermis was markedly decreased by hASCs. Differences were observed in epidermal differentiation but not basal cell proliferation. Similar results were obtained by both methods of hASC treatment to the skin. CONCLUSIONS: Exposure of UV-irradiated skin to hASC attenuated cell senescence and promoted repair from photodamage in an organotypic skin culture. These results suggest that hASC treatment may have a useful therapeutic effect for salvaging photodamaged skin.


Assuntos
Tecido Adiposo/citologia , Tecido Adiposo/efeitos da radiação , Envelhecimento da Pele/patologia , Pele/efeitos da radiação , Células Estromais/citologia , Células Estromais/efeitos da radiação , Adulto , Análise de Variância , Apoptose , Proliferação de Células , Células Cultivadas , Técnicas de Cocultura , Meios de Cultura Livres de Soro , Feminino , Humanos , Marcação In Situ das Extremidades Cortadas , Raios Ultravioleta
18.
J Biol Chem ; 287(16): 12858-66, 2012 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-22362783

RESUMO

Prion diseases are a group of rare, fatal neurodegenerative disorders associated with a conformational transformation of the cellular prion protein (PrP(C)) into a self-replicating and proteinase K-resistant conformer, termed scrapie PrP (PrP(Sc)). Aggregates of PrP(Sc) deposited around neurons lead to neuropathological alterations. Currently, there is no effective treatment for these fatal illnesses; thus, the development of an effective therapy is a priority. PrP peptide-based ELISA assay methods were developed for detection and immunoaffinity chromatography capture was developed for purification of naturally occurring PrP peptide autoantibodies present in human CSF, individual donor serum, and commercial preparations of pooled intravenous immunoglobulin (IVIg). The ratio of anti-PrP autoantibodies (PrP-AA) to total IgG was ∼1:1200. The binding epitope of purified PrP-AA was mapped to an N-terminal region comprising the PrP amino acid sequence KTNMK. Purified PrP-AA potently blocked fibril formation by a toxic 21-amino acid fragment of the PrP peptide containing the amino acid alanine to valine substitution corresponding to position 117 of the full-length peptide (A117V). Furthermore, PrP-AA attenuated the neurotoxicity of PrP(A117V) and wild-type peptides in rat cerebellar granule neuron (CGN) cultures. In contrast, IgG preparations depleted of PrP-AA had little effect on PrP fibril formation or PrP neurotoxicity. The specificity of PrP-AA was demonstrated by immunoprecipitating PrP protein in brain tissues of transgenic mice expressing the human PrP(A117V) epitope and Sc237 hamster. Based on these intriguing findings, it is suggested that human PrP-AA may be useful for interfering with the pathogenic effects of pathogenic prion proteins and, thereby has the potential to be an effective means for preventing or attenuating human prion disease progression.


Assuntos
Amiloide/imunologia , Anticorpos Bloqueadores/farmacologia , Autoanticorpos/farmacologia , Proteínas PrPC/imunologia , Proteínas PrPSc/imunologia , Doenças Priônicas , Animais , Anticorpos Bloqueadores/imunologia , Especificidade de Anticorpos , Autoanticorpos/imunologia , Cricetinae , Mapeamento de Epitopos , Epitopos , Heterozigoto , Humanos , Imunoglobulinas Intravenosas/farmacologia , Camundongos , Camundongos Transgênicos , Neuroglia/citologia , Neuroglia/imunologia , Neuroglia/patologia , Neurônios/citologia , Neurônios/imunologia , Neurônios/patologia , Proteínas PrPC/genética , Proteínas PrPC/metabolismo , Proteínas PrPSc/genética , Proteínas PrPSc/metabolismo , Cultura Primária de Células , Doenças Priônicas/imunologia , Doenças Priônicas/prevenção & controle , Doenças Priônicas/terapia , Ratos , Ratos Sprague-Dawley
19.
Am J Respir Crit Care Med ; 183(2): 215-25, 2011 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-20709815

RESUMO

RATIONALE: Adipose-derived stem cells express multiple growth factors that inhibit endothelial cell apoptosis, and demonstrate substantial pulmonary trapping after intravascular delivery. OBJECTIVES: We hypothesized that adipose stem cells would ameliorate chronic lung injury associated with endothelial cell apoptosis, such as that occurring in emphysema. METHODS: Therapeutic effects of systemically delivered human or mouse adult adipose stem cells were evaluated in murine models of emphysema induced by chronic exposure to cigarette smoke or by inhibition of vascular endothelial growth factor receptors. MEASUREMENTS AND MAIN RESULTS: Adipose stem cells were detectable in the parenchyma and large airways of lungs up to 21 days after injection. Adipose stem cell treatment was associated with reduced inflammatory infiltration in response to cigarette smoke exposure, and markedly decreased lung cell death and airspace enlargement in both models of emphysema. Remarkably, therapeutic results of adipose stem cells extended beyond lung protection by rescuing the suppressive effects of cigarette smoke on bone marrow hematopoietic progenitor cell function, and by restoring weight loss sustained by mice during cigarette smoke exposure. Pulmonary vascular protective effects of adipose stem cells were recapitulated by application of cell-free conditioned medium, which improved lung endothelial cell repair and recovery in a wound injury repair model and antagonized effects of cigarette smoke in vitro. CONCLUSIONS: These results suggest a useful therapeutic effect of adipose stem cells on both lung and systemic injury induced by cigarette smoke, and implicate a lung vascular protective function of adipose stem cell derived paracrine factors.


Assuntos
Tecido Adiposo/citologia , Células-Tronco Adultas/transplante , Lesão Pulmonar/terapia , Enfisema Pulmonar/terapia , Fumar/efeitos adversos , Transplante de Células-Tronco/métodos , Tecido Adiposo/transplante , Animais , Apoptose , Western Blotting , Técnicas de Cultura de Células , Modelos Animais de Doenças , Feminino , Citometria de Fluxo , Humanos , Inflamação/fisiopatologia , Inflamação/prevenção & controle , Lesão Pulmonar/etiologia , Lesão Pulmonar/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Alvéolos Pulmonares/fisiopatologia , Enfisema Pulmonar/etiologia , Enfisema Pulmonar/fisiopatologia , Transplante Heterólogo/métodos , Transplante Homólogo/métodos , Redução de Peso
20.
Int J Radiat Oncol Biol Phys ; 78(3): 888-96, 2010 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-20708345

RESUMO

PURPOSE: A delayed full-thickness wound-healing model was developed and used for examining the capacity of adipose-derived stem cells (ASCs), either alone or in platelet-rich fibrin gels, to promote healing. METHODS AND MATERIALS: Four pigs received electron beam radiation to the dorsal skin surface. Five weeks after radiation, subcutaneous fat was harvested from nonirradiated areas and processed to yield ASCs. Two weeks later, 28 to 30 full-thickness 1.5-cm(2) wounds were made in irradiated and nonirradiated skin. Wounds were treated with either saline solution, ASCs in saline solution, platelet-rich plasma (PRP) fibrin gel, ASCs in PRP, or non-autologous green fluorescence protein-labeled ASCs. RESULTS: The single radiation dose produced a significant loss of dermal microvasculature density (75%) by 7 weeks. There was a significant difference in the rate of healing between irradiated and nonirradiated skin treated with saline solution. The ASCs in PRP-treated wounds exhibited a significant 11.2% improvement in wound healing compared with saline solution. Enhancement was dependent on the combination of ASCs and PRP, because neither ASCs nor PRP alone had an effect. CONCLUSIONS: We have created a model that simulates the clinically relevant late radiation effects of delayed wound healing. Using this model, we showed that a combination of ASCs and PRP improves the healing rates of perfusion-depleted tissues, possibly through enhancing local levels of growth factors.


Assuntos
Adipócitos/transplante , Plasma Rico em Plaquetas , Lesões Experimentais por Radiação/terapia , Pele/lesões , Cicatrização/fisiologia , Adipócitos/citologia , Adipócitos/fisiologia , Animais , Contratura/patologia , Contratura/fisiopatologia , Feminino , Fibrina/uso terapêutico , Microvasos/patologia , Microvasos/efeitos da radiação , Modelos Animais , Lesões Experimentais por Radiação/patologia , Radiodermite/patologia , Radiodermite/terapia , Pele/irrigação sanguínea , Pele/efeitos da radiação , Cloreto de Sódio/uso terapêutico , Transplante de Células-Tronco , Suínos , Cicatrização/efeitos dos fármacos , Cicatrização/efeitos da radiação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...