Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 93
Filter
1.
J Inflamm (Lond) ; 21(1): 20, 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38867277

ABSTRACT

Autoimmune diseases are caused by an imbalance in the immune system, producing autoantibodies that cause inflammation leading to tissue damage and organ dysfunction. Systemic Lupus Erythematosus (SLE) is one of the most common autoimmune diseases and a major contributor to patient morbidity and mortality. Although many drugs manage the disease, curative therapy remains elusive, and current treatment regimens have substantial side effects. Recently, the therapeutic potential of exosomes has been extensively studied, and novel evidence has been demonstrated. A direct relationship between exosome contents and their ability to regulate the immune system, inflammation, and angiogenesis. The unique properties of extracellular vesicles, such as biomolecule transportation, biodegradability, and stability, make exosomes a promising treatment candidate for autoimmune diseases, particularly SLE. This review summarizes the structural features of exosomes, the isolation/purification/quantification method, their origin, effect, immune regulation, a critical consideration for selecting an appropriate source, and their therapeutic mechanisms in SLE.

2.
Stem Cells Transl Med ; 13(6): 559-571, 2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38530131

ABSTRACT

Mesenchymal stem cells (MSCs) are a promising therapy to potentially treat diabetes given their potent anti-inflammatory and immune-modulatory properties. While these regenerative cells have shown considerable promise in cell culture, their clinical translation has been challenging. In part, this can be attributed to these cells not reaching the pancreas to exert their regenerative effects following conventional intravenous (IV) injection, with the majority of cells being trapped in the lungs in the pulmonary first-pass effect. In the present study, we will therefore examine whether direct delivery of MSCs to the pancreas via an intra-arterial (IA) injection can improve their therapeutic efficacy. Using a mouse model, in which repetitive low doses of STZ induced a gentle, but progressive, hyperglycemia, we tested bone marrow-derived MSCs (BM-MSCs) which we have shown are enriched with pro-angiogenic and immunomodulatory factors. In cell culture studies, BM-MSCs were shown to preserve islet viability and function following exposure to proinflammatory cytokines (IFN-γ, IL-1ß, and TNF-α) through an increase in pAkt. When tested in our animal model, mice receiving IV BM-MSCs were not able to mitigate the effects of STZ, however those which received the same dose and batch of cells via IA injection were able to maintain basal and dynamic glycemic control, to similar levels as seen in healthy control animals, over 10 days. This study shows the importance of considering precision delivery approaches to ensure cell-based therapies reach their intended targets to enable them to exert their therapeutic effects.


Subject(s)
Diabetes Mellitus, Experimental , Injections, Intra-Arterial , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Animals , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cell Transplantation/methods , Humans , Mice , Diabetes Mellitus, Experimental/therapy , Pancreas , Bone Marrow Cells/cytology , Male , Mice, Inbred C57BL , Cytokines/metabolism
3.
J Clin Med ; 12(13)2023 Jun 23.
Article in English | MEDLINE | ID: mdl-37445274

ABSTRACT

Graft versus host disease (GVHD) is one of the most serious complications following stem cell transplant in children and is a major cause of morbidity and mortality. Corticosteroids remain the mainstay of treatment, and although a majority of children respond to systemic steroids, those refractory to or dependent upon corticosteroids suffer from complications secondary to long-term steroid administration. This problem has prompted consideration of steroid-sparing treatment strategies, although the time to clinical remission can be variable. Intraarterial corticosteroid delivery has been used in adults as a rescue therapy in steroid-resistant patients, but its use in children has been limited. We investigated the feasibility of intraarterial steroid administration into the bowel and/or liver in a cohort of six pediatric patients with acute GVHD. All patients successfully underwent treatment with no serious adverse effects. Five of five (100%) patients with gastrointestinal bleeding due to GVHD had rapid symptom improvement by 48 h, which was durable up to three weeks. Three of four (75%) patients with hepatic GVHD had improved cholestasis following intraarterial steroid administration. Our experience with this small cohort preliminarily demonstrated the feasibility and safety of intraarterial steroid administration in children with acute GVHD. This approach warrants consideration as a rescue therapy in steroid-refractory cases and as a "bridge" therapy for children with severe acute GVHD who are transitioning to steroid-sparing regimens.

5.
Biomolecules ; 13(4)2023 04 04.
Article in English | MEDLINE | ID: mdl-37189396

ABSTRACT

Pancreatic ß cells are central to glycemic regulation through insulin production. Studies show autophagy as an essential process in ß cell function and fate. Autophagy is a catabolic cellular process that regulates cell homeostasis by recycling surplus or damaged cell components. Impaired autophagy results in ß cell loss of function and apoptosis and, as a result, diabetes initiation and progress. It has been shown that in response to endoplasmic reticulum stress, inflammation, and high metabolic demands, autophagy affects ß cell function, insulin synthesis, and secretion. This review highlights recent evidence regarding how autophagy can affect ß cells' fate in the pathogenesis of diabetes. Furthermore, we discuss the role of important intrinsic and extrinsic autophagy modulators, which can lead to ß cell failure.


Subject(s)
Diabetes Mellitus , Insulin-Secreting Cells , Humans , Insulin-Secreting Cells/metabolism , Diabetes Mellitus/metabolism , Insulin/metabolism , Endoplasmic Reticulum Stress/physiology , Autophagy/physiology
6.
J Clin Med ; 12(6)2023 Mar 20.
Article in English | MEDLINE | ID: mdl-36983386

ABSTRACT

Systemic steroid exposure, while useful for the treatment of acute flares in inflammatory bowel disease (IBD), is associated with an array of side effects that are particularly significant in children. Technical advancements have enabled locoregional intraarterial steroid delivery directly into specific segments of the gastrointestinal tract, thereby maximizing tissue concentration while limiting systemic exposure. We investigated the feasibility of intraarterial steroid administration into the bowel in a cohort of nine pediatric patients who had IBD. This treatment approach provided symptom relief in all patients, with sustained relief (>2 weeks) in seven out of nine; no serious adverse effects occurred in any patient. In addition, we identified patterns of vascular morphologic changes indicative of a vasculopathy within the mesenteric circulation of inflamed segments of the bowel in pediatric patients with Crohn's disease, which correlated with disease activity. An analysis of publicly available transcriptomic studies identified vasculitis-associated molecular pathways activated in the endothelial cells of patients with active Crohn's disease, suggesting a possible shared transcriptional program between vasculitis and IBD. Intraarterial corticosteroid treatment is safe and has the potential to be widely accepted as a locoregional approach for therapy delivery directly into the bowel; however, this approach still warrants further consideration as a short-term "bridge" between therapy transitions for symptomatic IBD patients with refractory disease, as part of a broader steroid-minimizing treatment strategy.

7.
Eur J Nucl Med Mol Imaging ; 50(5): 1337-1350, 2023 04.
Article in English | MEDLINE | ID: mdl-36633614

ABSTRACT

PURPOSE: To provide a holistic and complete comparison of the five most advanced AI models in the augmentation of low-dose 18F-FDG PET data over the entire dose reduction spectrum. METHODS: In this multicenter study, five AI models were investigated for restoring low-count whole-body PET/MRI, covering convolutional benchmarks - U-Net, enhanced deep super-resolution network (EDSR), generative adversarial network (GAN) - and the most cutting-edge image reconstruction transformer models in computer vision to date - Swin transformer image restoration network (SwinIR) and EDSR-ViT (vision transformer). The models were evaluated against six groups of count levels representing the simulated 75%, 50%, 25%, 12.5%, 6.25%, and 1% (extremely ultra-low-count) of the clinical standard 3 MBq/kg 18F-FDG dose. The comparisons were performed upon two independent cohorts - (1) a primary cohort from Stanford University and (2) a cross-continental external validation cohort from Tübingen University - in order to ensure the findings are generalizable. A total of 476 original count and simulated low-count whole-body PET/MRI scans were incorporated into this analysis. RESULTS: For low-count PET restoration on the primary cohort, the mean structural similarity index (SSIM) scores for dose 6.25% were 0.898 (95% CI, 0.887-0.910) for EDSR, 0.893 (0.881-0.905) for EDSR-ViT, 0.873 (0.859-0.887) for GAN, 0.885 (0.873-0.898) for U-Net, and 0.910 (0.900-0.920) for SwinIR. In continuation, SwinIR and U-Net's performances were also discreetly evaluated at each simulated radiotracer dose levels. Using the primary Stanford cohort, the mean diagnostic image quality (DIQ; 5-point Likert scale) scores of SwinIR restoration were 5 (SD, 0) for dose 75%, 4.50 (0.535) for dose 50%, 3.75 (0.463) for dose 25%, 3.25 (0.463) for dose 12.5%, 4 (0.926) for dose 6.25%, and 2.5 (0.534) for dose 1%. CONCLUSION: Compared to low-count PET images, with near-to or nondiagnostic images at higher dose reduction levels (up to 6.25%), both SwinIR and U-Net significantly improve the diagnostic quality of PET images. A radiotracer dose reduction to 1% of the current clinical standard radiotracer dose is out of scope for current AI techniques.


Subject(s)
Artificial Intelligence , Fluorodeoxyglucose F18 , Humans , Drug Tapering , Positron-Emission Tomography/methods , Image Processing, Computer-Assisted/methods , Magnetic Resonance Imaging/methods
8.
Neural Regen Res ; 18(1): 18-22, 2023 Jan.
Article in English | MEDLINE | ID: mdl-35799503

ABSTRACT

Alzheimer's disease is a progressive and fatal neurodegenerative disorder that starts many years before the onset of cognitive symptoms. Identifying novel biomarkers for Alzheimer's disease has the potential for patient risk stratification, early diagnosis, and disease monitoring in response to therapy. A novel class of biomarkers is extracellular vesicles given their sensitivity and specificity to specific diseases. In addition, extracellular vesicles can be used as novel biological therapeutics given their ability to efficiently and functionally deliver therapeutic cargo. This is critical given the huge unmet need for novel treatment strategies for Alzheimer's disease. This review summarizes and discusses the most recent findings in this field.

9.
Stem Cell Reports ; 18(1): 190-204, 2023 01 10.
Article in English | MEDLINE | ID: mdl-36493779

ABSTRACT

Mesenchymal stem cells (MSCs) are gaining increasing prominence as an effective regenerative cellular therapy. However, ensuring consistent and reliable effects across clinical populations has proved to be challenging. In part, this can be attributed to heterogeneity in the intrinsic molecular and regenerative signature of MSCs, which is dependent on their source of origin. The present work uses integrated omics-based profiling, at different functional levels, to compare the anti-inflammatory, immunomodulatory, and angiogenic properties between MSCs from neonatal (umbilical cord MSC [UC-MSC]) and adult (adipose tissue MSC [AD-MSC], and bone marrow MSC [BM-MSC]) sources. Using multi-parametric analyses, we identified that UC-MSCs promote a more robust host innate immune response; in contrast, adult-MSCs appear to facilitate remodeling of the extracellular matrix (ECM) with stronger activation of angiogenic cascades. These data should help facilitate the standardization of source-specific MSCs, such that their regenerative signatures can be confidently used to target specific disease processes.


Subject(s)
Adult Stem Cells , Mesenchymal Stem Cells , Infant, Newborn , Humans , Proteome , Transcriptome , Gene Expression Profiling , Bone Marrow Cells
11.
Front Mol Neurosci ; 15: 1011225, 2022.
Article in English | MEDLINE | ID: mdl-36277497

ABSTRACT

Alzheimer's disease (AD) is a major cause of age-related dementia and is characterized by progressive brain damage that gradually destroys memory and the ability to learn, which ultimately leads to the decline of a patient's ability to perform daily activities. Although some of the pharmacological treatments of AD are available for symptomatic relief, they are not able to limit the progression of AD and have several side effects. Mesenchymal stem/stromal cells (MSCs) could be a potential therapeutic option for treating AD due to their immunomodulatory, anti-inflammatory, regenerative, antioxidant, anti-apoptotic, and neuroprotective effects. MSCs not only secret neuroprotective and anti-inflammatory factors to promote the survival of neurons, but they also transfer functional mitochondria and miRNAs to boost their bioenergetic profile as well as improve microglial clearance of accumulated protein aggregates. This review focuses on different clinical and preclinical studies using MSC as a therapy for treating AD, their outcomes, limitations and the strategies to potentiate their clinical translation.

12.
Radiographics ; 42(6): 1580-1597, 2022 10.
Article in English | MEDLINE | ID: mdl-36190845

ABSTRACT

The field of pediatric interventional radiology encompasses the treatment of a broad range of patients. Whether treating a premature infant who weighs less than 1 kg or treating an adult-sized teenager who weighs more than 100 kg, the innovative skills of the interventional radiologist are required to adapt equipment designed for adult patients, to meet the needs of children. Moreover, children cannot be treated simply as small adults owing to a number of factors, including differences in physiology, disease processes, and treatment techniques between pediatric and adult patients. In this article, the unique medical needs of children are highlighted, noting specific areas the interventional radiologist should be aware of when treating patients of all ages. Specific focus is placed on the unique considerations related to children in terms of their periprocedural needs and the procedural modifications required for routine pediatric procedures, with specific diseases of the liver, chest, and musculoskeletal system highlighted. The broader topic of vascular anomalies, although an important part of pediatric interventional radiology, was intentionally excluded to highlight some of the lesser-known procedures performed. ©RSNA, 2022.


Subject(s)
Radiology, Interventional , Adolescent , Child , Humans , Infant , Radiology, Interventional/methods
13.
Front Cell Dev Biol ; 10: 1006295, 2022.
Article in English | MEDLINE | ID: mdl-36313578

ABSTRACT

In recent years, mesenchymal stromal cells (MSCs) have generated a lot of attention due to their paracrine and immuno-modulatory properties. mesenchymal stromal cells derived from the umbilical cord (UC) are becoming increasingly recognized as having increased therapeutic potential when compared to mesenchymal stromal cells from other sources. The purpose of this review is to provide an overview of the various compartments of umbilical cord tissue from which mesenchymal stromal cells can be isolated, the differences and similarities with respect to their regenerative and immuno-modulatory properties, as well as the single cell transcriptomic profiles of in vitro expanded and freshly isolated umbilical cord-mesenchymal stromal cells. In addition, we discuss the therapeutic potential and biodistribution of umbilical cord-mesenchymal stromal cells following systemic administration while providing an overview of pre-clinical and clinical trials involving umbilical cord-mesenchymal stromal cells and their associated secretome and extracellular vesicles (EVs). The clinical applications of umbilical cord-mesenchymal stromal cells are also discussed, especially in relation to obstacles and potential solutions for their effective translation from bench to bedside.

14.
Transfus Apher Sci ; 61(6): 103454, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35618640

ABSTRACT

Coronavirus disease 2019 (COVID-19) is still an emergency in many countries. Herein, we report treatment with human placental-derived mesenchymal stromal cells transfusion (hPD-MSCT) in a critically ill infant diagnosed with COVID-19. A 28-day-old male infant with a history of pneumonia was referred to our center with decreased SpO2 (92%) and fever (38.5 °C). Real-time reverse transcription polymerase chain reaction (RT-PCR) and chest computed tomography (CT) confirmed COVID-19 infection. Considering the deteriorating clinical status of the patient despite the routine treatments (SpO2 82%), human placental derived mesenchymal stromal cells (hPD-MSCs) was transfused to him on day 9 and 11 (7 × 106 cells/session). The patient's general condition started to change 3 days after hPD-MSCT and poor feeding and low SpO2 improved day by day. On day 20, the patient was discharged (SpO2 97%) and our one-year follow-up showed a successful response to the treatment with no reported complications. hPD-MSCT may be considered as a possible treatment option in infants/children diagnosed with COVID-19 who fail to respond to conventional therapies. However, required dose, safety, and mechanistic studies are still warranted to further investigate this treatment.


Subject(s)
COVID-19 , Mesenchymal Stem Cells , Humans , Child , Male , Female , Pregnancy , COVID-19/therapy , SARS-CoV-2 , Critical Illness/therapy , Placenta
17.
Rev Endocr Metab Disord ; 23(3): 357-385, 2022 06.
Article in English | MEDLINE | ID: mdl-34647239

ABSTRACT

Diabetes is a chronic disorder characterized by dysregulated glycemic conditions. Diabetic complications include microvascular and macrovascular abnormalities and account for high morbidity and mortality rates in patients. Current clinical approaches for diabetic complications are limited to symptomatic treatments and tight control of blood sugar levels. Extracellular vesicles (EVs) released by somatic and stem cells have recently emerged as a new class of potent cell-free therapeutic delivery packets with a great potential to treat diabetic complications. EVs contain a mixture of bioactive molecules and can affect underlying pathological processes in favor of tissue healing. In addition, EVs have low immunogenicity and high storage capacity while maintaining nearly the same regenerative and immunomodulatory effects compared to current cell-based therapies. Therefore, EVs have received increasing attention for diabetes-related complications in recent years. In this review, we provide an outlook on diabetic complications and summarizes new knowledge and advances in EV applications. Moreover, we highlight recommendations for future EV-related research.


Subject(s)
Diabetes Complications , Diabetes Mellitus , Extracellular Vesicles , Blood Glucose , Diabetes Complications/therapy , Diabetes Mellitus/therapy , Humans , Wound Healing
18.
Growth Factors ; 39(1-6): 59-70, 2021.
Article in English | MEDLINE | ID: mdl-34886733

ABSTRACT

Autologous conditioned serum (ACS) is a blood-derived product that is prepared by the incubation of whole blood with medical-grade glass beads, resulting in serum enrichment in interleukin-1 receptor antagonist (IL-1Ra), anti-inflammatory cytokines (IL-4, IL-10, and IL-13), and high concentrations of growth factors. ACS has shown qualitatively and quantitatively better therapeutic effects than most established pharmacological treatments and surgery for joint diseases given its ability to both target the inflammatory cascade to decrease cartilage destruction as well as improve endogenous repair mechanisms. ACS application is simple and safe with limited adverse effects. This article reviews the role of ACS in degenerative joint disease, in addition to other inflammatory and autoimmune diseases, given its regenerative and immune-modulating properties.


Subject(s)
Interleukin 1 Receptor Antagonist Protein , Serum , Cytokines/metabolism , Intercellular Signaling Peptides and Proteins , Serum/metabolism
19.
ACS Appl Mater Interfaces ; 13(45): 53618-53629, 2021 Nov 17.
Article in English | MEDLINE | ID: mdl-34751556

ABSTRACT

Type-1 diabetes (T1DM) is a chronic metabolic disorder resulting from the autoimmune destruction of ß cells. The current standard of care requires multiple, daily injections of insulin and accurate monitoring of blood glucose levels (BGLs); in some cases, this results in diminished patient compliance and increased risk of hypoglycemia. Herein, we engineered hierarchically structured particles comprising a poly(lactic-co-glycolic) acid (PLGA) prismatic matrix, with a 20 × 20 µm base, encapsulating 200 nm insulin granules. Five configurations of these insulin-microPlates (INS-µPLs) were realized with different heights (5, 10, and 20 µm) and PLGA contents (10, 40, and, 60 mg). After detailed physicochemical and biopharmacological characterizations, the tissue-compliant 10H INS-µPL, realized with 10 mg of PLGA, presented the most effective release profile with ∼50% of the loaded insulin delivered at 4 weeks. In diabetic mice, a single 10H INS-µPL intraperitoneal deposition reduced BGLs to that of healthy mice within 1 h post-implantation (167.4 ± 49.0 vs 140.0 ± 9.2 mg/dL, respectively) and supported normoglycemic conditions for about 2 weeks. Furthermore, following the glucose challenge, diabetic mice implanted with 10H INS-µPL successfully regained glycemic control with a significant reduction in AUC0-120min (799.9 ± 134.83 vs 2234.60 ± 82.72 mg/dL) and increased insulin levels at 7 days post-implantation (1.14 ± 0.11 vs 0.38 ± 0.02 ng/mL), as compared to untreated diabetic mice. Collectively, these results demonstrate that INS-µPLs are a promising platform for the treatment of T1DM to be further optimized with the integration of smart glucose sensors.


Subject(s)
Biocompatible Materials/pharmacology , Blood Glucose/analysis , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 1/drug therapy , Hypoglycemic Agents/pharmacology , Insulin/pharmacology , Polyglycolic Acid/pharmacology , Animals , Biocompatible Materials/chemical synthesis , Biocompatible Materials/chemistry , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 1/chemically induced , Diabetes Mellitus, Type 1/metabolism , Dose-Response Relationship, Drug , Hypoglycemia/chemically induced , Hypoglycemia/drug therapy , Hypoglycemia/metabolism , Hypoglycemic Agents/chemical synthesis , Hypoglycemic Agents/chemistry , Insulin/chemistry , Male , Mice , Mice, Inbred C57BL , Particle Size , Polyglycolic Acid/chemistry , Streptozocin
20.
Sci Adv ; 7(47): eabf9221, 2021 Nov 19.
Article in English | MEDLINE | ID: mdl-34788097

ABSTRACT

In the present work, we developed, characterized, and tested an implantable graphene bioscaffold which elutes dexamethasone (Dex) that can accommodate islets and adipose tissue­derived mesenchymal stem cells (AD-MSCs). In vitro studies demonstrated that islets in graphene­0.5 w/v% Dex bioscaffolds had a substantial higher viability and function compared to islets in graphene-alone bioscaffolds or islets cultured alone (P < 0.05). In vivo studies, in which bioscaffolds were transplanted into the epididymal fat pad of diabetic mice, demonstrated that, when islet:AD-MSC units were seeded into graphene­0.5 w/v% Dex bioscaffolds, this resulted in complete restoration of glycemic control immediately after transplantation with these islets also showing a faster response to glucose challenges (P < 0.05). Hence, this combination approach of using a graphene bioscaffold that can be functionalized for local delivery of Dex into the surrounding microenvironment, together with AD-MSC therapy, can significantly improve the survival and function of transplanted islets.

SELECTION OF CITATIONS
SEARCH DETAIL
...