Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 727
1.
Eur Rev Med Pharmacol Sci ; 28(10): 3590-3597, 2024 May.
Article En | MEDLINE | ID: mdl-38856134

BACKGROUND: Recently, the infiltration of a subpopulation of cells represented by mononucleated cells extracted from peripheral blood [Peripheral Blood-Mononuclear Cells (PB-MNCs)] is becoming a useful technique for medical and surgical regenerative procedures. Due to the angiogenetic and regenerative properties of PB-MNCs, the infiltration of these cells is, in our opinion, a new option indicated in the treatment of pathologies characterized by tissue dystrophy, loss of vascularization, and non-healing wounds. CASE PRESENTATION: A 25-year-old active smoker patient was diagnosed with Rhabdomyosarcoma of the anterior tibial muscle of his left leg and treated with neoadjuvant chemo- and radiotherapy (RT). After the tumor excision, the patient developed wound dehiscence with bone exposure and a perilesional radiation-induced chronic dermatitis characterized by skin dyschromia and hair thinning along the treated area. The patient underwent surgical debridement and reconstruction with autologous skin grafts and dermal substitutes, with poor outcomes due to graft failure. The patient was subsequently treated with surgical debridement and coverage with a reverse sural fascia-cutaneous flap. After 13 days, wound dehiscence was observed, and reconstruction of the dehiscent areas was performed with a split-thickness autologous skin graft with no success. After wound debridement, a new split-thickness skin graft was performed, and a concentrate of autologous PB-MNCs was injected in the flap and perilesional skin. After 14 days, graft take was reached, and improvements in perilesional tissue tropism were noted. At 2 months follow-up, the patient appeared completely healed. CONCLUSIONS: In our opinion, the use of PB-MNCs to treat conditions characterized by tissue dystrophy, which require neoangiogenesis and cell regeneration, can be a useful and unconsidered technique that could be utilized to improve tissue tropism. Furthermore, prospective trials are necessary to validate our observations.


Leukocytes, Mononuclear , Humans , Male , Adult , Leukocytes, Mononuclear/transplantation , Plastic Surgery Procedures/methods , Lower Extremity , Rhabdomyosarcoma/therapy , Rhabdomyosarcoma/surgery , Wound Healing
2.
J Orthop Surg Res ; 19(1): 323, 2024 May 30.
Article En | MEDLINE | ID: mdl-38811966

BACKGROUND: To investigate the effect and underlying mechanism of umbilical cord blood-mononuclear cells (UCB-MNCs) in treating knee osteoarthritis (KOA) in rabbits. METHODS: A rabbit KOA model was prepared by anterior cruciate ligament transection (ACLT). Fifty New Zealand white rabbits were randomly divided into the control group, model group, sodium hyaluronate (SH) group, platelet-rich plasma (PRP) group and UCB-MNC group. Knee injections were performed once a week for five consecutive weeks. The gross view of the knee joint, morphology of knee cartilage and structural changes in the knee joint were observed on CT scans, and graded by the Lequesne MG behavioral score and the Mankin score. TNF-α and IL-1ß levels in the synovial fluid of the knee were measured by the enzyme-linked immunosorbent assay (ELISA). Expression levels of MMP-13 and COL-II in the knee cartilage were detected by Western blotting and qRT-PCR. RESULTS: The Lequesne MG behavioral score and the Mankin score were significantly higher in the model group than those in the control group (P < 0.05). Rabbits in the SH, PRP and UCB-MNC groups had sequentially lower scores than those in the model group. Imaging features of KOA were more pronounced in the model group than in the remaining groups. CB-MNC significantly relieved KOA, compared to SH and PRP. Significantly higher levels of TNF-α and IL-1ß in the synovial fluid of the knee, and up-regulated MMP-13 and down-regulated COL-II in the knee cartilage were detected in the model group than in the control group. These changes were significantly reversed by the treatment with SH, PRP and UCB-MNCs, especially UCB-MNCs. CONCLUSION: Injections of UCB-MNCs into knees protect the articular cartilage and hinder the progression of KOA in rabbits by improving the local microenvironment at knee joints.


Osteoarthritis, Knee , Animals , Rabbits , Osteoarthritis, Knee/therapy , Osteoarthritis, Knee/pathology , Fetal Blood , Disease Models, Animal , Male , Leukocytes, Mononuclear/transplantation , Leukocytes, Mononuclear/metabolism , Interleukin-1beta/metabolism , Tumor Necrosis Factor-alpha/metabolism , Synovial Fluid/metabolism , Platelet-Rich Plasma , Cord Blood Stem Cell Transplantation/methods , Random Allocation
3.
Cell Transplant ; 33: 9636897241251619, 2024.
Article En | MEDLINE | ID: mdl-38761062

Pressure injuries, or pressure ulcers, are a common problem that may lead to infections and major complications, besides being a social and economic burden due to the costs of treatment and hospitalization. While surgery is sometimes necessary, this also has complications such as recurrence or wound dehiscence. Among the newer methods of pressure injury treatment, advanced therapies are an interesting option. This study examines the healing properties of bone marrow mononuclear cells (BM-MNCs) embedded in a plasma-based scaffold in a mouse model. Pressure ulcers were created on the backs of mice (2 per mouse) using magnets and assigned to a group of ulcers that were left untreated (Control, n = 15), treated with plasma scaffold (Plasma, n = 15), or treated with plasma scaffold containing BM-MNC (Plasma + BM-MNC, n = 15). Each group was examined at three time points (3, 7, and 14 days) after the onset of treatment. At each time point, animals were subjected to biometric assessment, bioluminescence imaging, and tomography. Once treatment had finished, skin biopsies were processed for histological and wound healing reverse transcription polymerase chain reaction (RT-PCR) array studies. While wound closure percentages were higher in the Plasma and Plasma + BM-MNC groups, differences were not significant, and thus descriptive data are provided. In all individuals, the presence of donor cells was revealed by immunohistochemistry on posttreatment onset Days 3, 7, and 14. In the Plasma + BM-MNC group, less inflammation was observed by positron emission tomography-computed tomography (PET/CT) imaging of the mice at 7 days, and a complete morphometabolic response was produced at 14 days, in accordance with histological results. A much more pronounced inflammatory process was observed in controls than in the other two groups, and this persisted until Day 14 after treatment onset. RT-PCR array gene expression patterns were also found to vary significantly, with the greatest difference noted between both treatments at 14 days when 11 genes were differentially expressed.


Bone Marrow Cells , Disease Models, Animal , Pressure Ulcer , Wound Healing , Animals , Pressure Ulcer/therapy , Pressure Ulcer/pathology , Mice , Bone Marrow Cells/cytology , Male , Tissue Scaffolds/chemistry , Mice, Inbred C57BL , Bone Marrow Transplantation/methods , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/transplantation
4.
DNA Cell Biol ; 43(5): 207-218, 2024 May.
Article En | MEDLINE | ID: mdl-38635961

Sjogren's syndrome (SS) is a complex systemic autoimmune disease. This study aims to elucidate a humanized NOD-PrkdcscidIl2rgem1/Smoc (NSG) murine model to better clarify the pathogenesis of SS. NSG female mice were adoptively transferred with 10 million peripheral blood mononuclear cells (PBMCs) through the tail vein from healthy controls (HCs), primary Sjogren's syndrome (pSS), and systemic lupus erythematosus (SLE) patients on D0. The mice were subcutaneously injected with C57/B6j submandibular gland (SG) protein or phosphate-buffered saline on D3, D17 and D31, respectively. NSG mice were successfully transplanted with human PBMCs. Compared with NSG-HC group, NSG-pSS and NSG-SLE mice exhibited a large number of lymphocytes infiltration in the SG, decreased salivary flow rate, lung involvement, decreased expression of genes related to salivary secretion, and the production of autoantibodies. Type I interferon-related genes were increased in the SG of NSG-pSS and NSG-SLE mice. The ratio of BAX/BCL2, BAX, cleaved caspase3, and TUNEL staining were increased in the SG of NSG-pSS and NSG-SLE mice. The expressions of p-MLKL and p-RIPK3 were increased in the SG of NSG-pSS and NSG-SLE mice. Increased expression of type I interferon-related genes, PANoptosis (apoptosis and necroptosis) were identified in the SG of this typical humanized NSG murine model of SS.


Disease Models, Animal , Mice, Inbred NOD , Sjogren's Syndrome , Sjogren's Syndrome/pathology , Sjogren's Syndrome/immunology , Animals , Humans , Female , Mice , Apoptosis , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/transplantation , Submandibular Gland/pathology , Submandibular Gland/metabolism , Lupus Erythematosus, Systemic/pathology , Lupus Erythematosus, Systemic/immunology , Mice, Inbred C57BL , Autoantibodies/immunology , Interferon Type I/metabolism
5.
Brain ; 147(5): 1914-1925, 2024 May 03.
Article En | MEDLINE | ID: mdl-38181433

Autologous bone marrow mononuclear cells (BMMNCs) infused after severe traumatic brain injury have shown promise for treating the injury. We evaluated their impact in children, particularly their hypothesized ability to preserve the blood-brain barrier and diminish neuroinflammation, leading to structural CNS preservation with improved outcomes. We performed a randomized, double-blind, placebo-sham-controlled Bayesian dose-escalation clinical trial at two children's hospitals in Houston, TX and Phoenix, AZ, USA (NCT01851083). Patients 5-17 years of age with severe traumatic brain injury (Glasgow Coma Scale score ≤ 8) were randomized to BMMNC or placebo (3:2). Bone marrow harvest, cell isolation and infusion were completed by 48 h post-injury. A Bayesian continuous reassessment method was used with cohorts of size 3 in the BMMNC group to choose the safest between two doses. Primary end points were quantitative brain volumes using MRI and microstructural integrity of the corpus callosum (diffusivity and oedema measurements) at 6 months and 12 months. Long-term functional outcomes and ventilator days, intracranial pressure monitoring days, intensive care unit days and therapeutic intensity measures were compared between groups. Forty-seven patients were randomized, with 37 completing 1-year follow-up (23 BMMNC, 14 placebo). BMMNC treatment was associated with an almost 3-day (23%) reduction in ventilator days, 1-day (16%) reduction in intracranial pressure monitoring days and 3-day (14%) reduction in intensive care unit (ICU) days. White matter volume at 1 year in the BMMNC group was significantly preserved compared to placebo [decrease of 19 891 versus 40 491, respectively; mean difference of -20 600, 95% confidence interval (CI): -35 868 to -5332; P = 0.01], and the number of corpus callosum streamlines was reduced more in placebo than BMMNC, supporting evidence of preserved corpus callosum connectivity in the treated groups (-431 streamlines placebo versus -37 streamlines BMMNC; mean difference of -394, 95% CI: -803 to 15; P = 0.055), but this did not reach statistical significance due to high variability. We conclude that autologous BMMNC infusion in children within 48 h after severe traumatic brain injury is safe and feasible. Our data show that BMMNC infusion led to: (i) shorter intensive care duration and decreased ICU intensity; (ii) white matter structural preservation; and (iii) enhanced corpus callosum connectivity and improved microstructural metrics.


Bone Marrow Transplantation , Brain Injuries, Traumatic , Transplantation, Autologous , Humans , Child , Brain Injuries, Traumatic/therapy , Male , Female , Adolescent , Double-Blind Method , Child, Preschool , Bone Marrow Transplantation/methods , Transplantation, Autologous/methods , Magnetic Resonance Imaging , Treatment Outcome , Leukocytes, Mononuclear/transplantation , Bayes Theorem
6.
Curr Res Transl Med ; 72(2): 103437, 2024 Jun.
Article En | MEDLINE | ID: mdl-38244275

BACKGROUND: Type 2 diabetes (T2D) is a progressive disease. Many drugs currently being used for the management of T2D have minimal effect on pancreatic beta cells regeneration. Cell-based therapies might provide potential benefits in this aspect. METHODS: A pilot study in five T2D patients with 12 months follow-up was performed to evaluate the effect of autologous bone marrow mononuclear stem cells (BM-MNCs) infusion into pancreatic arteries on the insulin requirement, beta-cell function, insulin resistance, and systemic inflammatory marker (CRP). RESULTS: The primary endpoint, a 50 % reduction of total insulin doses from baseline, was not achieved in this study. However, a trend of increasing fasting C-peptide (p = 0.07) and C-peptide 60' (p = 0.07) and 90' (p = 0.07) after a mixed-meal tolerance test was observed 12 months post-infusion compared to baseline levels. A similar result was observed for the homeostatic model assessment of beta cell function (HOMA1-B), an index for beta cell function. No improvement was observed for insulin resistance measured by homeostasis model assessment of insulin resistance (HOMA1-IR) and systemic inflammatory parameter. CONCLUSION: Intraarterial pancreatic autologous BM-MNCs infusion might potentially improve beta cell function in T2D patients, although further study is needed to confirm this finding.


Bone Marrow Transplantation , Diabetes Mellitus, Type 2 , Insulin Resistance , Insulin-Secreting Cells , Transplantation, Autologous , Humans , Insulin-Secreting Cells/physiology , Insulin-Secreting Cells/drug effects , Diabetes Mellitus, Type 2/therapy , Diabetes Mellitus, Type 2/blood , Male , Middle Aged , Female , Bone Marrow Transplantation/methods , Pilot Projects , Biomarkers , Insulin/administration & dosage , Infusions, Intra-Arterial , Pancreas , Adult , Inflammation , C-Peptide/blood , C-Peptide/analysis , Aged , Leukocytes, Mononuclear/transplantation , Leukocytes, Mononuclear/metabolism
7.
Neurosci Res ; 203: 42-50, 2024 Jun.
Article En | MEDLINE | ID: mdl-38154662

Numerous evidences showed that human umbilical cord blood (UCB) mononuclear cells were a promising approach for the therapy of ischemic stroke(IS). The effect of stage-specific embryonic antigen 3 (SSEA3)positive subpopulation in UCB was not investigated in IS. In this study, we isolated SSEA3 positive cells from healthy UCB mononuclear cells, which comprised about 7.01% of the total UCB-mononuclear cells. Flow cytometry analysis revealed that SSEA3(+)UCB cells were almost positive for CD44 and CD45, and negative for CD73, CD90 and CD105. The expression of Oct3/4 in SSEA3(+)UCB cells were higher than that in UCB. SSEA3(+)UCB cells sorted by magnetic cell sorting were intravenously injected into the middle cerebral arterial occlusion(MCAO) rat model. Neurological score showed that SSEA3(+)UCB transplantation group exhibited significant improvements in the functional outcome of MCAO rats than UCB transplantation group. Nissl staining and microtubule association protein-2(MAP2) immunofluorescence staining showed that the SSEA3(+)UCB transplantation group decreased neuronal loss. SSEA3(+)UCB transplantation group reduced neuronal apoptosis, inhibited caspase3 expression, and decreased tumor necrosis factor α(TNF-α). These results indicate that SSEA3 positive cells are a novel subpopulation of UCB cells, which exhibit great potential for the treatment of ischemic stroke.


Disease Models, Animal , Fetal Blood , Ischemic Stroke , Animals , Humans , Ischemic Stroke/therapy , Ischemic Stroke/metabolism , Fetal Blood/cytology , Leukocytes, Mononuclear/transplantation , Leukocytes, Mononuclear/metabolism , Male , Rats, Sprague-Dawley , Rats , Infarction, Middle Cerebral Artery/therapy , Cord Blood Stem Cell Transplantation/methods , Stage-Specific Embryonic Antigens/metabolism , Brain Ischemia/therapy , Apoptosis/physiology
8.
Gene Ther ; 30(3-4): 309-322, 2023 04.
Article En | MEDLINE | ID: mdl-35931871

A primary goal in transplantation medicine is the induction of a tolerogenic environment for prevention of transplant rejection without the need for long-term pharmacological immunosuppression. Generation of alloantigen-specific regulatory T cells (Tregs) by transduction with chimeric antigen receptors (CARs) is a promising strategy to achieve this goal. This publication reports the preclinical characterization of Tregs (TR101) transduced with a human leukocyte antigen (HLA)-A*02 CAR lentiviral vector (TX200) designated to induce immunosuppression of allograft-specific effector T cells in HLA-A*02-negative recipients of HLA-A*02-positive transplants. In vitro results demonstrated specificity, immunosuppressive function, and safety of TX200-TR101. In NOD scid gamma (NSG) mice, TX200-TR101 prevented graft-versus-host disease (GvHD) in a xenogeneic GvHD model and TX200-TR101 Tregs localized to human HLA-A*02-positive skin transplants in a transplant model. TX200-TR101 persisted over the entire duration of a 3-month study in humanized HLA-A*02 NSG mice and remained stable, without switching to a proinflammatory phenotype. Concomitant tacrolimus did not impair TX200-TR101 Treg survival or their ability to inhibit peripheral blood mononuclear cell (PBMC) engraftment. These data demonstrate that TX200-TR101 is specific, stable, efficacious, and safe in preclinical models, and provide the basis for a first-in-human study.


Graft vs Host Disease , Organ Transplantation , Receptors, Chimeric Antigen , Mice , Animals , Humans , T-Lymphocytes, Regulatory , Leukocytes, Mononuclear/transplantation , HLA-A Antigens
9.
J Virol ; 96(3): e0139421, 2022 02 09.
Article En | MEDLINE | ID: mdl-34818071

Humanized mouse models are based on the engraftment of human cells in immunodeficient mouse strains, most notably the NSG strain. Most used models have a major limitation in common, the development of graft-versus-host disease (GVHD). GVHD not only introduces variabilities into the research data but also leads to animal welfare concerns. A new mouse strain, B6.129S-Rag2tm1Fwa CD47tm1Fpl Il2rgtm1Wjl/J, which lacks Rag1, IL2rg, and CD47 (triple knockout [TKO]), is resistant to GVHD development. We transplanted TKO mice with human peripheral blood mononuclear cells (PBMCs) to establish a new humanized PBMC (hu-PBMC) mouse model. A cohort of these mice was infected with HIV-1 and monitored for plasma HIV viremia and CD4+ T cell depletion. The onset and progression of GVHD were monitored by clinical signs. This study demonstrates that TKO mice transplanted with human PBMCs support engraftment of human immune cells in primary and secondary lymphoid tissues, rectum, and brain. Moreover, the TKO hu-PBMC model supports HIV-1 infection via the intraperitoneal, rectal, or vaginal route, as confirmed by robust plasma HIV viremia and CD4+ T cell depletion. Lastly, TKO mice showed a delayed onset of GVHD clinical signs (∼24 days) and exhibited significant decreases in plasma levels of tumor necrosis factor beta (TNF-ß). Based on these results, the TKO hu-PBMC mouse model not only supports humanization and HIV-1 infection but also has a delayed onset of GVHD development, making this model a valuable tool in HIV research. IMPORTANCE Currently, there is no cure or vaccine for HIV infection; thus, continued research is needed to end the HIV pandemic. While many animal models are used in HIV research, none is used more than the humanized mouse model. A major limitation with current humanized mouse models is the development of graft-versus-host disease (GVHD). Here, we describe a novel humanized-PBMC mouse model that has a delayed onset GVHD development and supports and models HIV infection comparably to well-established humanized mouse models.


Cell Transplantation , Disease Models, Animal , Graft vs Host Disease/etiology , HIV Infections/immunology , HIV Infections/virology , Leukocytes, Mononuclear/transplantation , Animals , Cell Transplantation/adverse effects , Cell Transplantation/methods , Disease Susceptibility , Female , HIV-1/immunology , Humans , Male , Mice , Mice, Knockout , Research , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Time Factors
10.
Fertil Steril ; 116(6): 1449-1454, 2021 12.
Article En | MEDLINE | ID: mdl-34836580

Despite the challenges in studying recurrent implantation failure, progress is currently being made in therapeutic options to help those who suffer from recurrent implantation failure. Three of the most promising therapeutic options for recurrent implantation failure include immune therapies such as peripheral blood mononuclear cells, platelet rich plasma and subcutaneous granulocyte-colony stimulating factor.


Embryo Implantation/physiology , Embryo Transfer/methods , Granulocyte Colony-Stimulating Factor/administration & dosage , Immunotherapy/methods , Platelet-Rich Plasma/physiology , Treatment Failure , Embryo Transfer/trends , Female , Fertilization in Vitro/methods , Fertilization in Vitro/trends , Humans , Immunotherapy/trends , Leukocytes, Mononuclear/physiology , Leukocytes, Mononuclear/transplantation , Pregnancy , Recurrence , Treatment Outcome
11.
Exp Neurol ; 346: 113864, 2021 12.
Article En | MEDLINE | ID: mdl-34520725

BACKGROUND AND PURPOSE: Post-stroke cognitive impairment (PSCI) has become a major public health issue, as a leading cause of dementia. The inflammation that develops soon after cerebral artery occlusion and may persist for weeks or months after stroke is a key component of PSCI. Our aim was to take advantage of the immunomodulatory properties of peripheral blood mononuclear cells (PB-MNC) stimulated with ephrin-B2/fc (PB-MNC+) for preventing PSCI. METHODS: Cortical infarct was induced by thermocoagulation of the middle cerebral artery in male diabetic mice (streptozotocin IP). PB-MNC were isolated from diabetic human donors, washed with recombinant ephrin-B2/Fc and injected into the mice intravenously on the following day. Infarct volume, sensorimotor deficit, cell death and immune cell densities were assessed on day 3. Six weeks later, cognitive assessment was performed using the Barnes maze. RESULTS: PB-MNC+ transplanted in post-stroke diabetic mice reduced the neurological deficit, infarct volume and apoptosis at D3, without modification of microglial cells, astrocytes and T-lymphocytes densities in the brain. Barnes maze assessment of memory showed that the learning, retention and reversal phases were not significantly modified by cell therapy. CONCLUSIONS: Intravenous PB-MNC+ administration the day after stroke induction in diabetic mice improved sensorimotor deficit and reduced infarct volume at the short term, but was unable to prevent long-term memory loss. To what extent diabetes impacts on cell therapy efficacy will have to be specifically investigated in the future. Including vascular risk factors systematically in preclinical studies of cell therapy will provide a comprehensive understanding of the mechanisms potentially limiting cell efficacy and also to identify good and bad responders, particularly in the long term.


Cell- and Tissue-Based Therapy/methods , Diabetes Mellitus, Experimental/therapy , Ephrin-B2/administration & dosage , Leukocytes, Mononuclear/transplantation , Memory Disorders/therapy , Stroke/therapy , Administration, Intravenous , Animals , Diabetes Mellitus, Experimental/physiopathology , Diabetes Mellitus, Experimental/psychology , Male , Memory Disorders/physiopathology , Memory Disorders/psychology , Mice , Mice, Inbred C57BL , Stroke/physiopathology , Stroke/psychology
12.
Front Immunol ; 12: 677970, 2021.
Article En | MEDLINE | ID: mdl-34248959

Objective: The contribution of sustained autologous autoantibody production by B cells to the pathogenesis of systemic sclerosis (SSc) and granulomatosis with polyangiitis (GPA) is not fully understood. To investigate this, a humanized mouse model was generated by transferring patient-derived peripheral blood mononuclear cells (PBMC) into immunocompromised mice. Methods: PBMC derived from patients with SSc and GPA as well as healthy controls (HD) were isolated, characterized by flow cytometry, and infused into Rag2-/-/IL2rg-/- mice. In addition, PBMC from SSc patients treated with rituximab were transferred into mice. Twelve weeks later, human autoantibodies were determined in blood of the recipient mice and affected tissues were analyzed for pathological changes by histology and immunohistochemistry. Results: Mice engrafted with PBMC derived from SSc patients developed autoantibodies such as antinuclear antibodies (ANA) mimicking the pattern of the respective donors. Moreover, cellular infiltrates dominated by B cells were observed in lung, kidney and muscles of the recipient mice. By contrast, PBMC derived from HD or GPA patients survived in recipient mice after transfer, but neither human autoantibodies nor inflammatory infiltrates in tissues were detected. Furthermore, these pathological changes were absent in mice transferred with PBMC from rituximab-treated SSc patients. Conclusion: This humanized mouse model is indicative for cross-reactivity of human lymphocytes to murine autoantigens and argues for a pivotal role of B cells as well as of sustained autoimmunity in the pathogenesis of SSc. It provides a powerful tool to study interstitial lung disease and so far, under-recognized disease manifestations such as myositis and interstitial nephritis.


Antibodies, Antinuclear/immunology , DNA-Binding Proteins/metabolism , Granulomatosis with Polyangiitis/blood , Interleukin Receptor Common gamma Subunit/metabolism , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/transplantation , Scleroderma, Systemic/blood , Adult , Aged , Animals , Antibodies, Antinuclear/blood , B-Lymphocytes/immunology , Cross Reactions , DNA-Binding Proteins/genetics , Female , Granulomatosis with Polyangiitis/immunology , Humans , Immunocompromised Host , Immunoglobulin G/blood , Immunoglobulin G/immunology , Immunologic Factors/therapeutic use , Inflammation/immunology , Interleukin Receptor Common gamma Subunit/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Models, Animal , Rituximab/therapeutic use , Scleroderma, Systemic/drug therapy , Scleroderma, Systemic/immunology , Treatment Outcome , Young Adult
13.
Front Immunol ; 12: 687715, 2021.
Article En | MEDLINE | ID: mdl-34177940

The outcome of organ transplantation is largely dictated by selection of a well-matched donor, which results in less chance of graft rejection. An allogeneic immune response is the main immunological barrier for successful organ transplantation. Donor and recipient human leukocyte antigen (HLA) mismatching diminishes outcomes after solid organ transplantation. The current evaluation of HLA incompatibility does not provide information on the immunogenicity of individual HLA mismatches and impact of non-HLA-related alloantigens, especially in vivo. Here we demonstrate a new method for analysis of alloimmune responsiveness between donor and recipient in vivo by introducing a humanized mouse model. Using molecular, cellular, and genomic analyses, we demonstrated that a recipient's personalized humanized mouse provided the most sensitive assessment of allogeneic responsiveness to potential donors. In our study, HLA typing provided a better recipient-donor match for one donor among two related donors. In contrast, assessment of an allogeneic response by mixed lymphocyte reaction (MLR) was indistinguishable between these donors. We determined that, in the recipient's humanized mouse model, the donor selected by HLA typing induced the strongest allogeneic response with markedly increased allograft rejection markers, including activated cytotoxic Granzyme B-expressing CD8+ T cells. Moreover, the same donor induced stronger upregulation of genes involved in the allograft rejection pathway as determined by transcriptome analysis of isolated human CD45+cells. Thus, the humanized mouse model determined the lowest degree of recipient-donor alloimmune response, allowing for better selection of donor and minimized immunological risk of allograft rejection in organ transplantation. In addition, this approach could be used to evaluate the level of alloresponse in allogeneic cell-based therapies that include cell products derived from pluripotent embryonic stem cells or adult stem cells, both undifferentiated and differentiated, all of which will produce allogeneic immune responses.


CD8-Positive T-Lymphocytes/immunology , Graft Rejection/prevention & control , HLA Antigens/immunology , Histocompatibility Testing , Histocompatibility , Leukocytes, Mononuclear/transplantation , Organ Transplantation , Spleen/immunology , Transplantation Tolerance , Animals , CD8-Positive T-Lymphocytes/metabolism , Cells, Cultured , Databases, Genetic , Graft Rejection/immunology , Graft Rejection/metabolism , Graft Survival , HLA Antigens/genetics , Humans , Isoantibodies/metabolism , Leukocytes, Mononuclear/immunology , Lymphocyte Culture Test, Mixed , Mice, Inbred NOD , Mice, SCID , Organ Transplantation/adverse effects , Phenotype , Predictive Value of Tests , Spleen/metabolism , Transcriptome , Transplantation, Homologous
14.
Front Immunol ; 12: 628059, 2021.
Article En | MEDLINE | ID: mdl-34122401

Artificial antigen-presenting cells (aAPCs) are synthetic versions of naturally occurring antigen-presenting cells (APCs) that, similar to natural APCs, promote efficient T effector cell responses in vitro. This report describes a method to produce acellular tolerogenic aAPCs made of biodegradable poly lactic-co-glycolic acid (PLGA) nanoparticles (NPs) and encapsulating IL-2 and TGF-ß for a paracrine release to T cells. We document that these aAPCs can induce both human CD4+ and CD8+ T cells to become FoxP3+ T regulatory cells (Tregs). The aAPC NP-expanded human Tregs are functional in vitro and can modulate systemic autoimmunity in vivo in humanized NSG mice. These findings establish a proof-of-concept to use PLGA NPs as aAPCs for the induction of human Tregs in vitro and in vivo, highlighting the immunotherapeutic potential of this targeted approach to repair IL-2 and/or TGF-ß defects documented in certain autoimmune diseases such as systemic lupus erythematosus.


Antigen-Presenting Cells/immunology , CD8-Positive T-Lymphocytes/drug effects , Graft vs Host Disease/prevention & control , Interleukin-2/pharmacology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/transplantation , Nanoparticles , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , T-Lymphocytes, Regulatory/drug effects , Transforming Growth Factor beta/pharmacology , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Disease Models, Animal , Forkhead Transcription Factors/metabolism , Graft vs Host Disease/genetics , Graft vs Host Disease/immunology , Graft vs Host Disease/metabolism , Humans , Interleukin-2/chemistry , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Mice, Inbred NOD , Mice, SCID , Proof of Concept Study , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Transforming Growth Factor beta/chemistry
15.
Signal Transduct Target Ther ; 6(1): 174, 2021 05 07.
Article En | MEDLINE | ID: mdl-33958574

Severely immunosuppressed AIDS patients with recurrent opportunistic infections (OIs) represent an unmet medical need even in the era of antiretroviral therapy (ART). Here we report the development of a human leukocyte antigen (HLA)-mismatched allogeneic adaptive immune therapy (AAIT) for severely immunosuppressed AIDS patients. Twelve severely immunosuppressed AIDS patients with severe OIs were enrolled in this single-arm study. Qualified donors received subcutaneous recombinant granulocyte-colony-stimulating factor twice daily for 4-5 days to stimulate hematopoiesis. Peripheral blood mononuclear cells were collected from these donors via leukapheresis and transfused into the coupled patients. Clinical, immunological, and virological parameters were monitored during a 12-month follow-up period. We found AAIT combined with ART was safe and well-tolerated at the examined doses and transfusion regimen in all 12 patients. Improvements in clinical symptoms were evident throughout the study period. All patients exhibited a steady increase of peripheral CD4+ T cells from a median 10.5 to 207.5 cells/µl. Rapid increase in peripheral CD8+ T-cell count from a median 416.5 to 1206.5 cells/µl was found in the first 90 days since initiation of AAIT. In addition, their inflammatory cytokine levels and HIV RNA viral load decreased. A short-term microchimerism with donor cells was found. There were no adverse events associated with graft-versus-host disease throughout the study period. Overall, AAIT treatment was safe, and might help severely immunosuppressed AIDS patients to achieve a better immune restoration. A further clinical trial with control is necessary to confirm the efficacy of AAIT medication.


Acquired Immunodeficiency Syndrome , Adoptive Transfer , HIV-1/immunology , HLA Antigens/immunology , Leukocytes, Mononuclear , Acquired Immunodeficiency Syndrome/immunology , Acquired Immunodeficiency Syndrome/pathology , Acquired Immunodeficiency Syndrome/therapy , Adolescent , Adult , Allografts , Female , Humans , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/transplantation , Male , Middle Aged
16.
J Reprod Immunol ; 145: 103323, 2021 06.
Article En | MEDLINE | ID: mdl-33878637

The purpose of this study was to assess whether intrauterine administration of peripheral blood mononuclear cells (PBMCs) activated by human chorionic gonadotropin (hCG) could improve the pregnancy and live birth rates in women with repeated implantation failure (RIF), and whether the parameters of co-culture of hCG and PBMCs would affect the clinical outcomes. Six databases (PubMed, Ovid, Medline, NCBI, Cqvip and Wanfang) were searched up to October 2020 by two independent reviewers. Seven studies were included according to specific inclusion and exclusion criteria. A meta-analysis showed that the pregnancy and live birth rates were significantly increased in the case group compared with the control group (odds ratio [OR]: 3.43, 95 % confidence interval [CI]: 1.78-6.61; P = 0.0002 and OR: 2.79, 95 % CI: 1.09-7.15; P = 0.03), especially when hCG was cultured with PBMCs for 48 h or PBMCs administration was performed two or three days before embryo transfer (ET). Neither the dosage of the hCG co-cultured with PBMCs nor the mean concentration of the administered PBMCs appeared to influence the therapeutic efficiency. In conclusion, intrauterine administration of PBMCs co-cultured with hCG for 48 h, conducted two or three days before ET, could be an effective therapy for women experiencing RIF. Due to the limitations of sample size and quality of the included studies, further high-quality studies with large sample sizes are warranted to optimize the parameters of hCG and PBMC co-culture to help more RIF patients benefit from this therapy.


Chorionic Gonadotropin/metabolism , Embryo Implantation/immunology , Embryo Transfer/methods , Infertility, Female/therapy , Leukocytes, Mononuclear/transplantation , Primary Cell Culture/methods , Cells, Cultured , Culture Media/metabolism , Female , Humans , Infertility, Female/immunology , Leukocytes, Mononuclear/immunology , Pregnancy , Pregnancy Rate , Treatment Outcome , Uterus/immunology
17.
Clin Podiatr Med Surg ; 38(2): 235-244, 2021 Apr.
Article En | MEDLINE | ID: mdl-33745654

The treatment of Achilles tendinitis from conservative to minimally invasive to surgery gives patients a wide range of treatment options for this common pathology. The use and role of biologics to augment this treatment is emerging. The use of biologics may enhance the healing potential of the Achilles tendon when conservative treatment fails. There are a handful of biologics being investigated to obtain if improved outcomes can be maximized.


Achilles Tendon , Tendinopathy/therapy , Bone Marrow Transplantation , Hepatocytes/transplantation , Humans , Leukocytes, Mononuclear/transplantation , Platelet-Derived Growth Factor/administration & dosage , Platelet-Rich Plasma , Tissue Scaffolds , Transforming Growth Factor beta/administration & dosage , Ultrasonography, Interventional , Vascular Endothelial Growth Factor A/administration & dosage , Wound Healing
18.
Stem Cells Transl Med ; 10(6): 895-909, 2021 06.
Article En | MEDLINE | ID: mdl-33599112

The quality and quantity of endothelial progenitor cells (EPCs) are impaired in patients with diabetes mellitus patients, leading to reduced tissue repair during autologous EPC therapy. This study aimed to address the limitations of the previously described serum-free Quantity and Quality Control Culture System (QQc) using CD34+ cells by investigating the therapeutic potential of a novel mononuclear cell (MNC)-QQ. MNCs were isolated from 50 mL of peripheral blood of patients with diabetes mellitus and healthy volunteers (n = 13 each) and subjected to QQc for 7 days in serum-free expansion media with VEGF, Flt-3 ligand, TPO, IL-6, and SCF. The vascular regeneration capability of MNC-QQ cells pre- or post-QQc was evaluated with an EPC colony-forming assay, FACS, EPC culture, tube formation assay, and quantitative real time PCR. For in vivo assessment, 1 × 104 pre- and post-MNC-QQc cells from diabetic donors were injected into a murine wound-healing model using Balb/c nude mice. The percentage of wound closure and angio-vasculogenesis was then assessed. This study revealed vasculogenic, anti-inflammatory, and wound-healing effects of MNC-QQ therapy in both in vitro and in vivo models. This system addresses the low efficiency and efficacy of the current naïve MNC therapy for wound-healing in diabetic patients. As this technique requires a simple blood draw, isolation, and peripheral blood MNC suspension culture for only a week, it can be used as a simple and effective outpatient-based vascular and regenerative therapy for patients with diabetes mellitus.


Diabetes Mellitus , Leukocytes, Mononuclear , Wound Healing , Animals , Culture Media, Serum-Free , Humans , Leukocytes, Mononuclear/transplantation , Mice , Mice, Inbred BALB C , Mice, Nude , Neovascularization, Physiologic
19.
J Immunol Methods ; 492: 113000, 2021 05.
Article En | MEDLINE | ID: mdl-33609532

BACKGROUND: Immunomodulation by mesenchymal stromal cells (MSCs) is a potentially important therapeutic modality. MSCs suppress peripheral blood mononuclear cell (PBMC) proliferation in vitro, suggesting a mechanism for suppressing inflammatory responses in vivo. This study details the interactions of PBMCs and MSCs. METHODS: Pooled human PBMCs and MSCs were co-cultured at different MSC:PBMC ratios and harvested from 0 to 120 h, with and without phytohaemagglutin A (PHA) stimulation. Proliferation of adherent MSCs and non-adherent PBMCs was assessed by quantitation of ATP levels. PBMC surface marker expression was analyzed by flow cytometry. Indoleamine 2,3-dioxygenase (IDO) activity was determined by kynurenine assay and IDO mRNA by RT-PCR. Cytokine release was measured by ELISA. Immunofluorescent microscopy detected MSC, PBMC, monocyte (CD14+) and apoptotic events. RESULTS: PBMC proliferation in response to PHA gave a robust ATP signal by 72 h, which was suppressed by co-culture with densely plated MSCs. Very low level MSC seeding densities relative to PBMC number reproducibly stimulated PBMC proliferation. The CD4+/CD3+ population significantly decreased over time while the CD8+/CD3+ population significantly increased. No change in CD4+/CD8+ ratio is seen with high density MSC co-culture; very low density MSCs augment the changes seen in PHA stimulated PBMCs alone. IDO activity in MSCs co-cultured with PBMCs correlated with PBMC suppression. MSCs increased the secretion of IL-10 and IL-6 from stimulated co-cultures and decreased TNF-α secretion. In stimulated co-culture, low density MSCs decreased in number; fluorescence immunomicroscopy detected association of PBMC with MSC and phosphatidyl serine externalization in both cell populations. CONCLUSIONS: A bidirectional interaction between MSCs and PBMCs occurs during co-culture. High numbers of MSCs inhibit PHA-stimulated PBMC proliferation and the PBMC response to stimulation; low numbers of MSCs augment these responses. Low density MSCs are susceptible to attrition, apparently by PBMC-induced apoptosis. These results may have direct application when considering therapeutic dosing of patients; low MSC doses may have unintended detrimental consequences.


Cell Communication/immunology , Leukocytes, Mononuclear/immunology , Mesenchymal Stem Cells/immunology , Primary Cell Culture/methods , Cell Proliferation/drug effects , Cell Transplantation , Coculture Techniques/methods , Humans , Leukocytes, Mononuclear/transplantation , Mitogens/pharmacology , Phytohemagglutinins/pharmacology
20.
Nat Commun ; 12(1): 135, 2021 01 08.
Article En | MEDLINE | ID: mdl-33420081

Since most variants that impact polygenic disease phenotypes localize to non-coding genomic regions, understanding the consequences of regulatory element variants will advance understanding of human disease mechanisms. Here, we report that the systemic lupus erythematosus (SLE) risk variant rs2431697 as likely causal for SLE through disruption of a regulatory element, modulating miR-146a expression. Using epigenomic analysis, genome-editing and 3D chromatin structure analysis, we show that rs2431697 tags a cell-type dependent distal enhancer specific for miR-146a that physically interacts with the miR-146a promoter. NF-kB binds the disease protective allele in a sequence-specific manner, increasing expression of this immunoregulatory microRNA. Finally, CRISPR activation-based modulation of this enhancer in the PBMCs of SLE patients attenuates type I interferon pathway activation by increasing miR-146a expression. Our work provides a strategy to define non-coding RNA functional regulatory elements using disease-associated variants and provides mechanistic links between autoimmune disease risk genetic variation and disease etiology.


Epigenesis, Genetic/immunology , Lupus Erythematosus, Systemic/genetics , MicroRNAs/genetics , Regulatory Sequences, Nucleic Acid/genetics , Animals , CRISPR-Cas Systems , Case-Control Studies , Cell Line, Tumor , Female , Genetic Predisposition to Disease , Genotyping Techniques , HEK293 Cells , Healthy Volunteers , Humans , Interferon Type I/metabolism , Leukocytes, Mononuclear/transplantation , Lupus Erythematosus, Systemic/blood , Lupus Erythematosus, Systemic/immunology , Mice , MicroRNAs/metabolism , NF-kappa B/metabolism , Polymorphism, Single Nucleotide , Primary Cell Culture , Promoter Regions, Genetic , RNA-Seq , Signal Transduction/genetics , Signal Transduction/immunology
...