Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 9.190
1.
Front Immunol ; 15: 1357706, 2024.
Article En | MEDLINE | ID: mdl-38846946

Introduction: In vivo T cell migration has been of interest to scientists for the past 60 years. T cell kinetics are important in the understanding of the immune response to infectious agents. More recently, adoptive T cell therapies have proven to be a most promising approach to treating a wide range of diseases, including autoimmune and cancer diseases, whereby the characterization of cellular kinetics represents an important step towards the prediction of therapeutic efficacy. Methods: Here, we developed a physiologically-based pharmacokinetic (PBPK) model that describes endogenous T cell homeostasis and the kinetics of exogenously administered T cells in mouse. Parameter calibration was performed using a nonlinear fixed-effects modeling approach based on published data on T cell kinetics and steady-state levels in different tissues of mice. The Partial Rank Correlation Coefficient (PRCC) method was used to perform a global sensitivity assessment. To estimate the impact of kinetic parameters on exogenously administered T cell dynamics, a local sensitivity analysis was conducted. Results: We simulated the model to analyze cellular kinetics following various T cell doses and frequencies of CCR7+ T cells in the population of infused lymphocytes. The model predicted the effects of T cell numbers and of population composition of infused T cells on the resultant concentration of T cells in various organs. For example, a higher percentage of CCR7+ T cells among exogenously administered T lymphocytes led to an augmented accumulation of T cells in the spleen. The model predicted a linear dependence of T cell dynamics on the dose of adoptively transferred T cells. Discussion: The mathematical model of T cell migration presented here can be integrated into a multi-scale model of the immune system and be used in a preclinical setting for predicting the distribution of genetically modified T lymphocytes in various organs, following adoptive T cell therapies.


T-Lymphocytes , Animals , Mice , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Cell Movement , Immunotherapy, Adoptive/methods , Models, Theoretical , Cell- and Tissue-Based Therapy/methods
2.
Nat Commun ; 15(1): 4870, 2024 Jun 07.
Article En | MEDLINE | ID: mdl-38849333

Critical challenges remain in clinical translation of extracellular vesicle (EV)-based therapeutics due to the absence of methods to enrich cells with high EV secretion. Current cell sorting methods are limited to surface markers that are uncorrelated to EV secretion or therapeutic potential. Here, we utilize a nanovial technology for enrichment of millions of single cells based on EV secretion. This approach is applied to select mesenchymal stem cells (MSCs) with high EV secretion as therapeutic cells for improving treatment. The selected MSCs exhibit distinct transcriptional profiles associated with EV biogenesis and vascular regeneration and maintain high levels of EV secretion after sorting and regrowth. In a mouse model of myocardial infarction, treatment with high-secreting MSCs improves heart functions compared to treatment with low-secreting MSCs. These findings highlight the therapeutic importance of EV secretion in regenerative cell therapies and suggest that selecting cells based on EV secretion could enhance therapeutic efficacy.


Extracellular Vesicles , Mesenchymal Stem Cells , Myocardial Infarction , Extracellular Vesicles/metabolism , Extracellular Vesicles/transplantation , Animals , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Mice , Myocardial Infarction/therapy , Myocardial Infarction/metabolism , Humans , Cell- and Tissue-Based Therapy/methods , Mesenchymal Stem Cell Transplantation/methods , Disease Models, Animal , Mice, Inbred C57BL , Cell Separation/methods , Male
3.
Sci Rep ; 14(1): 12747, 2024 06 03.
Article En | MEDLINE | ID: mdl-38830924

Cell directed therapy is an evolving therapeutic approach to treat organ dysfunction arising from hyperinflammation and cytokine storm by processing immune cells in an extracorporeal circuit. To investigate the mechanism of action of the Selective Cytopheretic Device (SCD), in vitro blood circuits were utilized to interrogate several aspects of the immunomodulatory therapy. SCD immunomodulatory activity is due to its effects on circulating neutrophils and monocytes in a low ionized calcium (iCa, Ca2+) blood circuit. Activated neutrophils adhere to the SCD fibers and degranulate with release of the constituents of their exocytotic vesicles. Adhered neutrophils in the low iCa environment display characteristics of apoptotic senescence. These neutrophils are subsequently released and returned back to circulation, demonstrating a clear potential for in vivo feedback. For monocytes, SCD treatment results in the selective adhesion of more pro-inflammatory subsets of the circulating monocyte pool, as demonstrated by both cell surface markers and cytokine secretory rates. Once bound, over time a subset of monocytes are released from the membrane with a less inflammatory functional phenotype. Similar methods to interrogate mechanism in vitro have been used to preliminarily confirm comparable findings in vivo. Therefore, the progressive amelioration of circulating leukocyte activation and immunomodulation of excessive inflammation observed in SCD clinical trials to date is likely due to this continuous autologous leukocyte processing.


Immunomodulation , Inflammation , Monocytes , Humans , Monocytes/immunology , Monocytes/metabolism , Inflammation/metabolism , Inflammation/immunology , Neutrophils/metabolism , Neutrophils/immunology , Cytokines/metabolism , Cell Adhesion , Cell- and Tissue-Based Therapy/methods , Calcium/metabolism
4.
Med Sci (Paris) ; 40(5): 437-444, 2024 May.
Article Fr | MEDLINE | ID: mdl-38819279

Recent advances in synthetic biology have paved the way for new cellular therapies, using cells capable of autonomously treating chronic diseases. These cells integrate a set of genes functioning in a closed-loop synthetic circuit, delivering a therapeutic effector in response to a specific pathological signal. While promising in mice, these therapies face clinical challenges related to safety and feasibility of in vivo implementation. The latest generations of synthetic circuits aim to address these issues through advanced bioengineering strategies outlined in this article.


Title: Les circuits synthétiques de gènes fonctionnant en boucle fermée - Concept et dernières avancées. Abstract: Les progrès récents de la biologie synthétique ont ouvert la voie à de nouvelles thérapies fondées sur des cellules rendues aptes à produire de manière autonome des substrats afin de traiter des maladies chroniques. Ces cellules modifiées intègrent un ensemble de gènes fonctionnant en circuit synthétique à boucle fermée, qui permettent de délivrer un effecteur thérapeutique en réponse à un signal pathologique déterminé. Bien que prometteuses chez la souris, ces thérapies font face à des obstacles cliniques liés à leur sûreté et à leur implémentation in vivo. Les dernières générations de circuits synthétiques cherchent à résoudre ces problèmes grâce à des stratégies de bioingénierie avancées, que nous présentons dans cet article.


Cell- and Tissue-Based Therapy , Gene Regulatory Networks , Genes, Synthetic , Synthetic Biology , Humans , Animals , Synthetic Biology/methods , Synthetic Biology/trends , Cell- and Tissue-Based Therapy/methods , Cell- and Tissue-Based Therapy/trends , Mice , Genetic Therapy/methods , Genetic Therapy/trends
5.
J Biomed Sci ; 31(1): 47, 2024 May 09.
Article En | MEDLINE | ID: mdl-38724973

The field of regenerative medicine has witnessed remarkable advancements with the emergence of induced pluripotent stem cells (iPSCs) derived from a variety of sources. Among these, urine-derived induced pluripotent stem cells (u-iPSCs) have garnered substantial attention due to their non-invasive and patient-friendly acquisition method. This review manuscript delves into the potential and application of u-iPSCs in advancing precision medicine, particularly in the realms of drug testing, disease modeling, and cell therapy. U-iPSCs are generated through the reprogramming of somatic cells found in urine samples, offering a unique and renewable source of patient-specific pluripotent cells. Their utility in drug testing has revolutionized the pharmaceutical industry by providing personalized platforms for drug screening, toxicity assessment, and efficacy evaluation. The availability of u-iPSCs with diverse genetic backgrounds facilitates the development of tailored therapeutic approaches, minimizing adverse effects and optimizing treatment outcomes. Furthermore, u-iPSCs have demonstrated remarkable efficacy in disease modeling, allowing researchers to recapitulate patient-specific pathologies in vitro. This not only enhances our understanding of disease mechanisms but also serves as a valuable tool for drug discovery and development. In addition, u-iPSC-based disease models offer a platform for studying rare and genetically complex diseases, often underserved by traditional research methods. The versatility of u-iPSCs extends to cell therapy applications, where they hold immense promise for regenerative medicine. Their potential to differentiate into various cell types, including neurons, cardiomyocytes, and hepatocytes, enables the development of patient-specific cell replacement therapies. This personalized approach can revolutionize the treatment of degenerative diseases, organ failure, and tissue damage by minimizing immune rejection and optimizing therapeutic outcomes. However, several challenges and considerations, such as standardization of reprogramming protocols, genomic stability, and scalability, must be addressed to fully exploit u-iPSCs' potential in precision medicine. In conclusion, this review underscores the transformative impact of u-iPSCs on advancing precision medicine and highlights the future prospects and challenges in harnessing this innovative technology for improved healthcare outcomes.


Cell- and Tissue-Based Therapy , Induced Pluripotent Stem Cells , Precision Medicine , Humans , Precision Medicine/methods , Induced Pluripotent Stem Cells/cytology , Cell- and Tissue-Based Therapy/methods , Drug Evaluation, Preclinical/methods , Urine/cytology , Regenerative Medicine/methods
6.
J Am Acad Orthop Surg ; 32(10): e476-e481, 2024 May 15.
Article En | MEDLINE | ID: mdl-38700858

With an aging population, and an anticipated increase in overall fracture incidence, a sound understanding of bone healing and how technology can optimize this process is crucial. Concentrated bone marrow aspirate (cBMA) is a technology that capitalizes on skeletal stem and progenitor cells (SSPCs) to enhance the regenerative capacity of bone. This overview highlights the science behind cBMA, discusses the role of SSPCs in bone homeostasis and fracture repair, and briefly details the clinical evidence supporting the use of cBMA in fracture healing. Despite promising early clinical results, a lack of standardization in harvest and processing techniques, coupled with patient variability, presents challenges in optimizing the use of cBMA. However, cBMA remains an emerging technology that may certainly play a crucial role in the future of fracture healing augmentation.


Fracture Healing , Humans , Fracture Healing/physiology , Bone Marrow Transplantation/methods , Cell- and Tissue-Based Therapy/methods , Bone Regeneration/physiology , Fractures, Bone/therapy , Bone Marrow Cells/cytology
9.
J Diabetes ; 16(6): e13557, 2024 Jun.
Article En | MEDLINE | ID: mdl-38751366

Diabetes mellitus (DM) is a common chronic disease affecting humans globally. It is characterized by abnormally elevated blood glucose levels due to the failure of insulin production or reduction of insulin sensitivity and functionality. Insulin and glucagon-like peptide (GLP)-1 replenishment or improvement of insulin resistance are the two major strategies to treat diabetes. Recently, optogenetics that uses genetically encoded light-sensitive proteins to precisely control cell functions has been regarded as a novel therapeutic strategy for diabetes. Here, we summarize the latest development of optogenetics and its integration with synthetic biology approaches to produce light-responsive cells for insulin/GLP-1 production, amelioration of insulin resistance and neuromodulation of insulin secretion. In addition, we introduce the development of cell encapsulation and delivery methods and smart bioelectronic devices for the in vivo application of optogenetics-based cell therapy in diabetes. The remaining challenges for optogenetics-based cell therapy in the clinical translational study are also discussed.


Diabetes Mellitus , Optogenetics , Humans , Optogenetics/methods , Diabetes Mellitus/therapy , Animals , Insulin/metabolism , Insulin Resistance , Glucagon-Like Peptide 1 , Cell- and Tissue-Based Therapy/methods , Insulin-Secreting Cells/metabolism
10.
Biomolecules ; 14(5)2024 Apr 30.
Article En | MEDLINE | ID: mdl-38785945

Mesenchymal stem cells (MSCs) are multipotent stromal cells with the ability to self-renew and multi-directional differentiation potential. Exogenously administered MSCs can migrate to damaged tissue sites and participate in the repair of damaged tissues. A large number of pre-clinical studies and clinical trials have demonstrated that MSCs have the potential to treat the abnormalities of congenital nervous system and neurodegenerative diseases. Therefore, MSCs hold great promise in the treatment of neurological diseases. Here, we summarize and highlight current progress in the understanding of the underlying mechanisms and strategies of MSC application in neurological diseases.


Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Nervous System Diseases , Humans , Nervous System Diseases/therapy , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cell Transplantation/methods , Animals , Cell- and Tissue-Based Therapy/methods , Clinical Trials as Topic , Cell Differentiation
11.
Biomolecules ; 14(5)2024 May 13.
Article En | MEDLINE | ID: mdl-38785982

Chimerism-based strategies represent a pioneering concept which has led to groundbreaking advancements in regenerative medicine and transplantation. This new approach offers therapeutic potential for the treatment of various diseases, including inherited disorders. The ongoing studies on chimeric cells prompted the development of Dystrophin-Expressing Chimeric (DEC) cells which were introduced as a potential therapy for Duchenne Muscular Dystrophy (DMD). DMD is a genetic condition that leads to premature death in adolescent boys and remains incurable with current methods. DEC therapy, created via the fusion of human myoblasts derived from normal and DMD-affected donors, has proven to be safe and efficacious when tested in experimental models of DMD after systemic-intraosseous administration. These studies confirmed increased dystrophin expression, which correlated with functional and morphological improvements in DMD-affected muscles, including cardiac, respiratory, and skeletal muscles. Furthermore, the application of DEC therapy in a clinical study confirmed its long-term safety and efficacy in DMD patients. This review summarizes the development of chimeric cell technology tested in preclinical models and clinical studies, highlighting the potential of DEC therapy in muscle regeneration and repair, and introduces chimeric cell-based therapies as a promising, novel approach for muscle regeneration and the treatment of DMD and other neuromuscular disorders.


Cell- and Tissue-Based Therapy , Dystrophin , Muscle, Skeletal , Muscular Dystrophy, Duchenne , Regeneration , Muscular Dystrophy, Duchenne/therapy , Muscular Dystrophy, Duchenne/genetics , Humans , Animals , Cell- and Tissue-Based Therapy/methods , Dystrophin/genetics , Dystrophin/metabolism , Myoblasts/metabolism
12.
Cells ; 13(10)2024 May 17.
Article En | MEDLINE | ID: mdl-38786076

Cardiovascular diseases continue to challenge global health, demanding innovative therapeutic solutions. This review delves into the transformative role of mesenchymal stem cells (MSCs) in advancing cardiovascular therapeutics. Beginning with a historical perspective, we trace the development of stem cell research related to cardiovascular diseases, highlighting foundational therapeutic approaches and the evolution of cell-based treatments. Recognizing the inherent challenges of MSC-based cardiovascular therapeutics, which range from understanding the pro-reparative activity of MSCs to tailoring patient-specific treatments, we emphasize the need to refine the pro-regenerative capacity of these cells. Crucially, our focus then shifts to the strategies of the fourth generation of cell-based therapies: leveraging the secretomic prowess of MSCs, particularly the role of extracellular vesicles; integrating biocompatible scaffolds and artificial sheets to amplify MSCs' potential; adopting three-dimensional ex vivo propagation tailored to specific tissue niches; harnessing the promise of genetic modifications for targeted tissue repair; and institutionalizing good manufacturing practice protocols to ensure therapeutic safety and efficacy. We conclude with reflections on these advancements, envisaging a future landscape redefined by MSCs in cardiovascular regeneration. This review offers both a consolidation of our current understanding and a view toward imminent therapeutic horizons.


Cardiovascular Diseases , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Humans , Mesenchymal Stem Cells/cytology , Cardiovascular Diseases/therapy , Mesenchymal Stem Cell Transplantation/methods , Animals , Extracellular Vesicles/metabolism , Extracellular Vesicles/transplantation , Cell- and Tissue-Based Therapy/methods
13.
Front Immunol ; 15: 1349138, 2024.
Article En | MEDLINE | ID: mdl-38720903

Autoimmune diseases can damage specific or multiple organs and tissues, influence the quality of life, and even cause disability and death. A 'disease in a dish' can be developed based on patients-derived induced pluripotent stem cells (iPSCs) and iPSCs-derived disease-relevant cell types to provide a platform for pathogenesis research, phenotypical assays, cell therapy, and drug discovery. With rapid progress in molecular biology research methods including genome-sequencing technology, epigenetic analysis, '-omics' analysis and organoid technology, large amount of data represents an opportunity to help in gaining an in-depth understanding of pathological mechanisms and developing novel therapeutic strategies for these diseases. This paper aimed to review the iPSCs-based research on phenotype confirmation, mechanism exploration, drug discovery, and cell therapy for autoimmune diseases, especially multiple sclerosis, inflammatory bowel disease, and type 1 diabetes using iPSCs and iPSCs-derived cells.


Autoimmune Diseases , Induced Pluripotent Stem Cells , Humans , Autoimmune Diseases/immunology , Autoimmune Diseases/therapy , Animals , Drug Discovery , Cell- and Tissue-Based Therapy/methods
14.
J Transl Med ; 22(1): 416, 2024 May 02.
Article En | MEDLINE | ID: mdl-38698408

One of the most challenging aspects of developing advanced cell therapy products (CTPs) is defining the mechanism of action (MOA), potency and efficacy of the product. This perspective examines these concepts and presents helpful ways to think about them through the lens of metrology. A logical framework for thinking about MOA, potency and efficacy is presented that is consistent with the existing regulatory guidelines, but also accommodates what has been learned from the 27 US FDA-approved CTPs. Available information regarding MOA, potency and efficacy for the 27 FDA-approved CTPs is reviewed to provide background and perspective. Potency process and efficacy process charts are introduced to clarify and illustrate the relationships between six key concepts: MOA, potency, potency test, efficacy, efficacy endpoint and efficacy endpoint test. Careful consideration of the meaning of these terms makes it easier to discuss the challenges of correlating potency test results with clinical outcomes and to understand how the relationships between the concepts can be misunderstood during development and clinical trials. Examples of how a product can be "potent but not efficacious" or "not potent but efficacious" are presented. Two example applications of the framework compare how MOA is assessed in cell cultures, animal models and human clinical trials and reveals the challenge of establishing MOA in humans. Lastly, important considerations for the development of potency tests for a CTP are discussed. These perspectives can help product developers set appropriate expectations for understanding a product's MOA and potency, avoid unrealistic assumptions and improve communication among team members during the development of CTPs.


Cell- and Tissue-Based Therapy , Humans , Cell- and Tissue-Based Therapy/methods , Animals , Treatment Outcome , United States Food and Drug Administration , United States , Clinical Trials as Topic
15.
J Orthop Traumatol ; 25(1): 28, 2024 May 24.
Article En | MEDLINE | ID: mdl-38789881

Mesenchymal stem cells are core to bone homeostasis and repair. They both provide the progenitor cells from which bone cells are formed and regulate the local cytokine environment to create a pro-osteogenic environment. Dysregulation of these cells is often seen in orthopaedic pathology and can be manipulated by the physician treating the patient. This narrative review aims to describe the common applications of cell therapies to bone healing whilst also suggesting the future direction of these techniques.


Mesenchymal Stem Cell Transplantation , Humans , Mesenchymal Stem Cell Transplantation/methods , Bone Regeneration/physiology , Mesenchymal Stem Cells , Fracture Healing/physiology , Cell- and Tissue-Based Therapy/methods , Osteogenesis/physiology
16.
JAMA ; 331(20): 1703-1704, 2024 05 28.
Article En | MEDLINE | ID: mdl-38696201

This Viewpoint discusses the death of a patient caused by unregulated biological products and efforts to encourage federal government oversight of such products.


Government Regulation , United States Food and Drug Administration , Humans , United States , Cell- and Tissue-Based Therapy , Tissue Banks/legislation & jurisprudence
17.
J Transl Med ; 22(1): 435, 2024 May 08.
Article En | MEDLINE | ID: mdl-38720379

Diabetes mellitus is a significant global public health challenge, with a rising prevalence and associated morbidity and mortality. Cell therapy has evolved over time and holds great potential in diabetes treatment. In the present review, we discussed the recent progresses in cell-based therapies for diabetes that provides an overview of islet and stem cell transplantation technologies used in clinical settings, highlighting their strengths and limitations. We also discussed immunomodulatory strategies employed in cell therapies. Therefore, this review highlights key progresses that pave the way to design transformative treatments to improve the life quality among diabetic patients.


Cell- and Tissue-Based Therapy , Diabetes Mellitus , Stem Cell Transplantation , Humans , Diabetes Mellitus/therapy , Cell- and Tissue-Based Therapy/methods , Islets of Langerhans Transplantation , Animals
18.
Biofabrication ; 16(3)2024 Jun 03.
Article En | MEDLINE | ID: mdl-38768586

Toward the translation of allogeneic cell therapy products, cell banks are needed not only to manufacture the final human product but also during the preclinical evaluation of an animal-based analogous cellular product (ACP). These cell banks need to be established at both the master cell bank (MCB) level and the working cell bank (WCB) level. Inasmuch as most of the development of cell therapy products is at academic centers, it is imperative that academic researchers understand how to establish MCBs and WCBs within an academic environment. To illustrate this process, using articular cartilage as the model, a cell bank for an ACP was developed (MCBs at passage 2, WCBs at passage 5) to produce self-assembled neocartilage for preclinical evaluation (constructs at passage 7). The cell bank system is estimated to be able to produce between 160 000 and 400 000 constructs for each of the six MCBs. Overall, the ACP cell bank yielded constructs that are analogous to the intended human product, which is critical toward conducting preclinical evaluations of the ACP for inclusion in an Investigational New Drug application to the FDA.


Cell- and Tissue-Based Therapy , Humans , Animals , Cartilage, Articular/cytology , Tissue Engineering , Tissue Banks
20.
Biomed Pharmacother ; 175: 116645, 2024 Jun.
Article En | MEDLINE | ID: mdl-38729050

Peripheral nerve injuries (PNIs) frequently occur due to various factors, including mechanical trauma such as accidents or tool-related incidents, as well as complications arising from diseases like tumor resection. These injuries frequently result in persistent numbness, impaired motor and sensory functions, neuropathic pain, or even paralysis, which can impose a significant financial burden on patients due to outcomes that often fall short of expectations. The most frequently employed clinical treatment for PNIs involves either direct sutures of the severed ends or bridging the proximal and distal stumps using autologous nerve grafts. However, autologous nerve transplantation may result in sensory and motor functional loss at the donor site, as well as neuroma formation and scarring. Transplantation of Schwann cells/Schwann cell-like cells has emerged as a promising cellular therapy to reconstruct the microenvironment and facilitate peripheral nerve regeneration. In this review, we summarize the role of Schwann cells and recent advances in Schwann cell therapy in peripheral nerve regeneration. We summarize current techniques used in cell therapy, including cell injection, 3D-printed scaffolds for cell delivery, cell encapsulation techniques, as well as the cell types employed in experiments, experimental models, and research findings. At the end of the paper, we summarize the challenges and advantages of various cells (including ESCs, iPSCs, and BMSCs) in clinical cell therapy. Our goal is to provide the theoretical and experimental basis for future treatments targeting peripheral nerves, highlighting the potential of cell therapy and tissue engineering as invaluable resources for promoting nerve regeneration.


Nerve Regeneration , Peripheral Nerve Injuries , Schwann Cells , Schwann Cells/physiology , Humans , Animals , Nerve Regeneration/physiology , Peripheral Nerve Injuries/therapy , Cell- and Tissue-Based Therapy/methods , Peripheral Nerves/physiology
...