Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 87
Filter
Add more filters

Publication year range
1.
Comput Math Methods Med ; 2022: 8361665, 2022.
Article in English | MEDLINE | ID: mdl-35047059

ABSTRACT

OBJECTIVE: To explore the effect of autoimmune cell therapy on immune cells in patients with chronic obstructive pulmonary disease (COPD) and to provide a reference for clinical treatment of COPD. METHODS: Sixty patients with stable COPD were randomly divided into control group and treatment group (n = 30). The control group was given conventional treatment, and the treatment group was given one autoimmune cell therapy on the basis of conventional treatment. The serum levels of CD3+ T cells, CD4+ T cells, CD8+ cells, B cells, and NK cells in the peripheral blood were detected by flow cytometry. Possible adverse reactions were detected at any time during treatment. RESULTS: There were no significant differences in the contents of CD3+ T cells, CD4+ T cells, CD8+ cells, B cells, and NK cells in the serum of the control group (P > 0.05). Compared with before treatment, the contents of CD3+ T cells, CD4+ T cells, CD8+ cells, B cells, and NK cells in the serum of the treatment group were significantly increased (P < 0.05). The ratio of CD4 + /CD8+ T cells in both control and treatment groups did not change significantly during treatment (P > 0.05). There were no significant differences in serum CD3+ T cells, CD4+ T cells, CD8+ cells, B cells, and NK cells in the treatment group at 30 days and 90 days after treatment (P > 0.05), but they were significantly higher than those in the control group (P < 0.05). CONCLUSION: Autoimmune cell therapy can significantly increase the level of immune cells in the body and can be maintained for a long period of time, which has certain clinical benefits for recurrent respiratory tract infections and acute exacerbation in patients with COPD.


Subject(s)
Cell- and Tissue-Based Therapy/methods , Pulmonary Disease, Chronic Obstructive/immunology , Pulmonary Disease, Chronic Obstructive/therapy , Aged , Aged, 80 and over , Autoimmunity , B-Lymphocytes/immunology , B-Lymphocytes/transplantation , Blood Transfusion, Autologous/methods , Blood Transfusion, Autologous/statistics & numerical data , Cell- and Tissue-Based Therapy/statistics & numerical data , Computational Biology , Female , Humans , Killer Cells, Natural/immunology , Killer Cells, Natural/transplantation , Leukocyte Transfusion/methods , Leukocyte Transfusion/statistics & numerical data , Male , Middle Aged , Pulmonary Disease, Chronic Obstructive/blood , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/transplantation
2.
Biomed Res Int ; 2021: 6548554, 2021.
Article in English | MEDLINE | ID: mdl-34840978

ABSTRACT

Tissue engineering is biomedical engineering that uses suitable biochemical and physicochemical factors to assemble functional constructs that restore or improve damaged tissues. Recently, cell therapies as a subset of tissue engineering have been very promising in the treatment of ocular diseases. One of the most important biophysical factors to make this happen is noninvasive electrical stimulation (ES) to target ocular cells that may preserve vision in multiple retinal and optic nerve diseases. The science of cellular and biophysical interactions is very exciting in regenerative medicine now. Although the exact effect of ES on cells is unknown, multiple mechanisms are considered to underlie the effects of ES, including increased production of neurotrophic agents, improved cell migration, and inhibition of proinflammatory cytokines and cellular apoptosis. In this review, we highlighted the effects of ES on ocular cells, especially on the corneal, retinal, and optic nerve cells. Initially, we summarized the current literature on the in vitro and in vivo effects of ES on ocular cells and then we provided the clinical studies describing the effect of ES on ocular complications. For each area, we used some of the most impactful articles to show the important concepts and results that advanced the state of these interactions. We conclude with reflections on emerging new areas and perspectives for future development in this field.


Subject(s)
Electric Stimulation Therapy/methods , Eye Diseases/therapy , Eye/cytology , Tissue Engineering/methods , Animals , Cell- and Tissue-Based Therapy/methods , Eye Diseases/physiopathology , Humans , In Vitro Techniques , Regenerative Medicine/methods , Stem Cells/cytology
3.
J Extracell Vesicles ; 10(10): e12134, 2021 08.
Article in English | MEDLINE | ID: mdl-34429860

ABSTRACT

Mesenchymal stromal cell-derived extracellular vesicles (MSC-EVs) turn out to be a promising source of cell-free therapy. Here, we investigated the biodistribution and effect of nebulized human adipose-derived MSC-EVs (haMSC-EVs) in the preclinical lung injury model and explored the safety of nebulized haMSC-EVs in healthy volunteers. DiR-labelled haMSC-EVs were used to explore the distribution of nebulized haMSC-EVs in the murine model. Pseudomonas aeruginosa-induced murine lung injury model was established, and survival rate, as well as WBC counts, histology, IL-6, TNF-α and IL-10 levels in bronchoalveolar lavage fluid (BALF) were measured to explore the optimal therapeutic dose of haMSC-EVs through the nebulized route. Twenty-four healthy volunteers were involved and received the haMSC-EVs once, ranging from 2 × 108 particles to 16 × 108 particles (MEXVT study, NCT04313647). Nebulizing haMSC-EVs improved survival rate to 80% at 96 h in P. aeruginosa-induced murine lung injury model by decreasing lung inflammation and histological severity. All volunteers tolerated the haMSC-EVs nebulization well, and no serious adverse events were observed from starting nebulization to the 7th day after nebulization. These findings suggest that nebulized haMSC-EVs could be a promising therapeutic strategy, offering preliminary evidence to promote the future clinical applications of nebulized haMSC-EVs in lung injury diseases.


Subject(s)
Cell- and Tissue-Based Therapy/methods , Cytokines/metabolism , Drug Evaluation, Preclinical , Extracellular Vesicles/physiology , Lung Injury/therapy , Mesenchymal Stem Cells/physiology , Adolescent , Adult , Animals , Bronchoalveolar Lavage Fluid/chemistry , Disease Models, Animal , Female , Humans , Lung Injury/microbiology , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Middle Aged , Patient Safety , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa , Survival Rate , Therapeutics/methods , Young Adult
4.
Front Immunol ; 12: 661875, 2021.
Article in English | MEDLINE | ID: mdl-34054826

ABSTRACT

Regulatory T (Treg) cells are a heterogenous population of immunosuppressive T cells whose therapeutic potential for the treatment of autoimmune diseases and graft rejection is currently being explored. While clinical trial results thus far support the safety and efficacy of adoptive therapies using polyclonal Treg cells, some studies suggest that antigen-specific Treg cells are more potent in regulating and improving immune tolerance in a disease-specific manner. Hence, several approaches to generate and/or expand antigen-specific Treg cells in vitro or in vivo are currently under investigation. However, antigen-specific Treg cell therapies face additional challenges that require further consideration, including the identification of disease-relevant antigens as well as the in vivo stability and migratory behavior of Treg cells following transfer. In this review, we discuss these approaches and the potential limitations and describe prospective strategies to enhance the efficacy of antigen-specific Treg cell treatments in autoimmunity and transplantation.


Subject(s)
Autoimmune Diseases/immunology , Autoimmune Diseases/therapy , Cell- and Tissue-Based Therapy/methods , Immunotherapy, Adoptive/methods , Organ Transplantation , T-Lymphocytes, Regulatory/immunology , Animals , Autoimmunity , Drug Evaluation, Preclinical , Genetic Engineering , Humans , Immune Tolerance , Immunotherapy, Adoptive/statistics & numerical data , Mice
5.
Front Immunol ; 12: 664244, 2021.
Article in English | MEDLINE | ID: mdl-33841448

ABSTRACT

A number of immune regulatory cellular therapies, including regulatory T cells and mesenchymal stromal cells, have emerged as novel alternative therapies for the control of transplant alloresponses. Clinical studies have demonstrated their feasibility and safety, however developing our understanding of the impact of cellular therapeutics in vivo requires advanced immune monitoring strategies. To accurately monitor the immune response, a combination of complementary methods is required to measure the cellular and molecular phenotype as well as the function of cells involved. In this review we focus on the current immune monitoring strategies and discuss which methods may be utilized in the future.


Subject(s)
Cell Transplantation , Cell- and Tissue-Based Therapy , Clinical Trials as Topic , Monitoring, Immunologic/methods , Animals , Cell Transplantation/adverse effects , Cell Transplantation/methods , Cell- and Tissue-Based Therapy/adverse effects , Cell- and Tissue-Based Therapy/methods , Cell- and Tissue-Based Therapy/standards , Clinical Decision-Making , Clinical Trials as Topic/standards , Disease Management , Humans , Monitoring, Immunologic/standards , Organ Specificity , Treatment Outcome
6.
Cancer Discov ; 11(3): 560-574, 2021 03.
Article in English | MEDLINE | ID: mdl-33563662

ABSTRACT

Adoptive cell therapy (ACT) for cancer shows tremendous potential; however, several challenges preclude its widespread use. These include poor T-cell function in hostile tumor microenvironments, a lack of tumor-specific target antigens, and the high cost and poor scalability of cell therapy manufacturing. Creative genome-editing strategies are beginning to emerge to address each of these limitations, which has initiated the next generation of cell therapy products now entering clinical trials. CRISPR is at the forefront of this revolution, offering a simple and versatile platform for genetic engineering. This review provides a comprehensive overview of CRISPR applications that have advanced ACT. SIGNIFICANCE: The clinical impact of ACT for cancer can be expanded by implementing specific genetic modifications that enhance the potency, safety, and scalability of cellular products. Here we provide a detailed description of such genetic modifications, highlighting avenues to enhance the therapeutic efficacy and accessibility of ACT for cancer. Furthermore, we review high-throughput CRISPR genetic screens that have unveiled novel targets for cell therapy enhancement.


Subject(s)
CRISPR-Cas Systems , Cell- and Tissue-Based Therapy/methods , Gene Editing/methods , Immunotherapy, Adoptive/methods , Neoplasms/therapy , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Cell- and Tissue-Based Therapy/adverse effects , Clinical Trials as Topic , Combined Modality Therapy , Disease Management , Drug Evaluation, Preclinical , Genetic Engineering , Genetic Therapy , Humans , Immunotherapy, Adoptive/adverse effects , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Treatment Outcome
7.
Cancer Immunol Immunother ; 70(1): 31-45, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32623477

ABSTRACT

Dendritic cell (DC)-based vaccines are recognized as a promising immunotherapeutic strategy against cancer; however, the efficacy of immunotherapy with DCs is controlled via immune checkpoints, such as programmed death-ligand 1 (PD-L1). PD-L1 expressed on DC and tumor cells binds to programmed death-1 (PD-1) receptors on the activated T cells, which leads to the inhibition of cytotoxic T cells. Blocking of PD-L1 on DC may lead to improve the efficacy of DC therapy for cancer. Here we demonstrated that DC vaccination in combination with pomalidomide and programmed death-ligand 1 (PD-L1) blockade inhibited tumor growth of a multiple myeloma (MM) mouse model. DCs + pomalidomide with dexamethasone + PD-L1 blockade significantly inhibited immune immunosuppressive factors and promoted proportions of immune effector cells in the spleen and tumor microenvironment. Additionally, functional activities of cytotoxic T lymphocytes and NK cells in spleen were enhanced by DCs + pomalidomide with dexamethasone + PD-L1 blockade. Taken together, this study identifies a potential new therapeutic approach for the treatment of MM. These results also provide a foundation for the future development of immunotherapeutic modalities to inhibit tumor growth and restore immune function in MM.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , B7-H1 Antigen/antagonists & inhibitors , Dendritic Cells/immunology , Immune Checkpoint Inhibitors/pharmacology , Multiple Myeloma/drug therapy , Multiple Myeloma/therapy , Thalidomide/analogs & derivatives , Animals , Cell Proliferation/drug effects , Cell- and Tissue-Based Therapy/methods , Cells, Cultured , Chemotherapy, Adjuvant/methods , Drug Evaluation, Preclinical/methods , Female , Immunosuppressive Agents/pharmacology , Immunotherapy/methods , Mice , Mice, Inbred BALB C , Multiple Myeloma/immunology , Spleen/drug effects , Spleen/immunology , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology , Thalidomide/pharmacology , Vaccination/methods
8.
Int J Mol Sci ; 21(23)2020 Nov 24.
Article in English | MEDLINE | ID: mdl-33255323

ABSTRACT

The translation of new therapies for spinal cord injury to clinical trials can be facilitated with large animal models close in morpho-physiological scale to humans. Here, we report functional restoration and morphological reorganization after spinal contusion in pigs, following a combined treatment of locomotor training facilitated with epidural electrical stimulation (EES) and cell-mediated triple gene therapy with umbilical cord blood mononuclear cells overexpressing recombinant vascular endothelial growth factor, glial-derived neurotrophic factor, and neural cell adhesion molecule. Preliminary results obtained on a small sample of pigs 2 months after spinal contusion revealed the difference in post-traumatic spinal cord outcomes in control and treated animals. In treated pigs, motor performance was enabled by EES and the corresponding morpho-functional changes in hind limb skeletal muscles were accompanied by the reorganization of the glial cell, the reaction of stress cell, and synaptic proteins. Our data demonstrate effects of combined EES-facilitated motor training and cell-mediated triple gene therapy after spinal contusion in large animals, informing a background for further animal studies and clinical translation.


Subject(s)
Electric Stimulation Therapy , Glial Cell Line-Derived Neurotrophic Factor/genetics , Neural Cell Adhesion Molecules/genetics , Spinal Cord Injuries/therapy , Vascular Endothelial Growth Factor A/genetics , Adenoviridae/genetics , Animals , Cell- and Tissue-Based Therapy/methods , Disease Models, Animal , Epidural Space , Genetic Therapy/methods , Genetic Vectors/therapeutic use , Glial Cell Line-Derived Neurotrophic Factor/therapeutic use , Humans , Motor Activity/genetics , Motor Activity/physiology , Neural Cell Adhesion Molecules/therapeutic use , Neuroglia/transplantation , Recovery of Function/genetics , Recovery of Function/radiation effects , Spinal Cord/physiopathology , Spinal Cord/radiation effects , Spinal Cord Injuries/genetics , Spinal Cord Injuries/physiopathology , Swine/genetics , Vascular Endothelial Growth Factor A/therapeutic use
9.
Nat Commun ; 11(1): 2758, 2020 06 02.
Article in English | MEDLINE | ID: mdl-32488069

ABSTRACT

Human beige adipocytes (BAs) have potential utility for the development of therapeutics to treat diabetes and obesity-associated diseases. Although several reports have described the generation of beige adipocytes in vitro, their potential utility in cell therapy and drug discovery has not been reported. Here, we describe the generation of BAs from human adipose-derived stem/stromal cells (ADSCs) in serum-free medium with efficiencies >90%. Molecular profiling of beige adipocytes shows them to be similar to primary BAs isolated from human tissue. In vitro, beige adipocytes exhibit uncoupled mitochondrial respiration and cAMP-induced lipolytic activity. Following transplantation, BAs increase whole-body energy expenditure and oxygen consumption, while reducing body-weight in recipient mice. Finally, we show the therapeutic utility of BAs in a platform for high-throughput drug screening (HTS). These findings demonstrate the potential utility of BAs as a cell therapeutic and as a tool for the identification of drugs to treat metabolic diseases.


Subject(s)
Adipocytes, Beige/metabolism , Cell- and Tissue-Based Therapy/methods , Drug Discovery/methods , Metabolic Diseases/metabolism , Adipocytes, Beige/cytology , Animals , Body Weight , Drug Evaluation, Preclinical , Energy Metabolism , Female , High-Throughput Screening Assays , Humans , Male , Mesenchymal Stem Cells , Metabolic Diseases/drug therapy , Mice , Mice, Inbred NOD , Mice, SCID , Mitochondria/metabolism , Oxygen Consumption , Stromal Cells , Transplantation
10.
Clin Microbiol Infect ; 26(8): 988-998, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32454187

ABSTRACT

BACKGROUND: As COVID-19 cases continue to rise globally, evidence from large randomized controlled trials is still lacking. Currently, numerous trials testing potential treatment and preventative options are being undertaken all over the world. OBJECTIVES: We summarized all registered clinical trials examining treatment and prevention options for COVID-19. Additionally, we evaluated the quality of the retrieved studies. DATA SOURCES: Clinicaltrials.gov, the Chinese Clinical Trial Registry and the European Union Clinical Trials Register were systematically searched. STUDY ELIGIBILITY CRITERIA: Registered clinical trials examining treatment and/or prevention options for COVID-19 were included. No language, country or study design restrictions were applied. We excluded withdrawn or cancelled studies and trials not reporting therapeutic or preventative strategies for COVID-19. PARTICIPANTS AND INTERVENTIONS: No restrictions in terms of participants' age and medical background or type of intervention were enforced. METHODS: The registries were searched using the term 'coronavirus' or 'COVID-19' from their inception until 26 March 2020. Additional manual search of the registries was also performed. Eligible studies were summarized and tabulated. Interventional trials were methodologically analysed, excluding expanded access studies and trials testing traditional Chinese medicine. RESULTS: In total, 309 trials evaluating therapeutic management options, 23 studies assessing preventive strategies and three studies examining both were retrieved. Finally, 214 studies were methodologically reviewed. Interventional treatment studies were mostly randomized (n = 150/198, 76%) and open label (n = 73/198, 37%) with a median number of planned inclusions of 90 (interquartile range 40-200). Major categories of interventions that are currently being investigated are discussed. CONCLUSIONS: Numerous clinical trials have been registered since the onset of the COVID-19 pandemic. Summarized data on these trials will assist physicians and researchers to promote patient care and guide future research efforts for COVID-19 pandemic containment.


Subject(s)
Antiviral Agents/therapeutic use , Betacoronavirus/drug effects , Cell- and Tissue-Based Therapy/methods , Coronavirus Infections/therapy , Pneumonia, Viral/therapy , Antiviral Agents/pharmacology , COVID-19 , Clinical Trials as Topic , Coronavirus Infections/virology , Humans , Pandemics , Pneumonia, Viral/virology , Randomized Controlled Trials as Topic , SARS-CoV-2 , Treatment Outcome
11.
J Cell Physiol ; 235(10): 6462-6495, 2020 10.
Article in English | MEDLINE | ID: mdl-32239727

ABSTRACT

Mesenchymal stem cells (MSCs) are earmarked as perfect candidates for cell therapy and tissue engineering due to their capacity to differentiate into different cell types. However, their potential for application in regenerative medicine declines when the levels of the reactive oxygen and nitrogen species (RONS) increase from the physiological levels, a phenomenon which is at least inevitable in ex vivo cultures and air-exposed damaged tissues. Increased levels of RONS can alter the patterns of osteogenic and adipogenic differentiation and inhibit proliferation, as well. Besides, oxidative stress enhances senescence and cell death, thus lowering the success rates of the MSC engraftment. Hence, in this review, we have selected some representatives of antioxidants and newly emerged nano antioxidants in three main categories, including chemical compounds, biometabolites, and protein precursors/proteins, which are proved to be effective in the treatment of MSCs. We will focus on how antioxidants can be applied to optimize the clinical usage of the MSCs and their associated signaling pathways. We have also reviewed several paralleled properties of some antioxidants and nano antioxidants which can be simultaneously used in real-time imaging, scaffolding techniques, and other applications in addition to their primary antioxidative function.


Subject(s)
Antioxidants/pharmacology , Antioxidants/therapeutic use , Mesenchymal Stem Cells/drug effects , Protective Agents/pharmacology , Protective Agents/therapeutic use , Animals , Cell- and Tissue-Based Therapy/methods , Dietary Supplements , Humans , Oxidative Stress/drug effects , Signal Transduction/drug effects
12.
Nanomedicine ; 24: 102149, 2020 02.
Article in English | MEDLINE | ID: mdl-31927133

ABSTRACT

Alzheimer's disease (AD) is a multifactorial neurodegenerative disease which leads to progressive dysfunction of cognition, memory and learning in elderly people. Common therapeutic agents are not only inadequate to suppress the progression of AD pathogenesis but also produce deleterious side effects; hence, development of alternative therapies is required to specifically suppress complications of AD. The current review provides a commentary on conventional as well as novel therapeutic approaches with an emphasis on stem cell and nano-based therapies for improvement and management of AD pathogenesis. According to our overview of the current literature, AD is a multi-factorial disorder with various pathogenic trajectories; hence, a multifunctional strategy to create effective neuroprotective agents is required to treat this disorder.


Subject(s)
Alzheimer Disease/pathology , Cell- and Tissue-Based Therapy/methods , Neurodegenerative Diseases/pathology , Alzheimer Disease/therapy , Animals , Humans , Neurodegenerative Diseases/therapy
13.
Neuropharmacology ; 167: 107702, 2020 05 01.
Article in English | MEDLINE | ID: mdl-31301334

ABSTRACT

The epilepsies are a complex group of disorders that can be caused by a myriad of genetic and acquired factors. As such, identifying interventions that will prevent development of epilepsy, as well as cure the disorder once established, will require a multifaceted approach. Here we discuss the progress in scientific discovery propelling us towards this goal, including identification of genetic risk factors and big data approaches that integrate clinical and molecular 'omics' datasets to identify common pathophysiological signatures and biomarkers. We discuss the many animal and cellular models of epilepsy, what they have taught us about pathophysiology, and the cutting edge cellular, optogenetic, chemogenetic and anti-seizure drug screening approaches that are being used to find new cures in these models. Finally, we reflect on the work that still needs to be done towards identify at-risk individuals early, targeting and stopping epileptogenesis, and optimizing promising treatment approaches. Ultimately, developing and implementing cures for epilepsy will require a coordinated and immense effort from clinicians and basic scientists, as well as industry, and should always be guided by the needs of individuals affected by epilepsy and their families. This article is part of the special issue entitled 'New Epilepsy Therapies for the 21st Century - From Antiseizure Drugs to Prevention, Modification and Cure of Epilepsy'.


Subject(s)
Anticonvulsants/therapeutic use , Cell- and Tissue-Based Therapy/methods , Drug Discovery/methods , Epilepsy/therapy , Genetic Therapy/methods , Animals , Anticonvulsants/pharmacology , Cell- and Tissue-Based Therapy/trends , Drug Discovery/trends , Drug Evaluation, Preclinical/methods , Epigenesis, Genetic/drug effects , Epigenesis, Genetic/physiology , Epilepsy/diagnosis , Epilepsy/genetics , Genetic Therapy/trends , Humans
14.
J Drug Target ; 28(1): 11-22, 2020 01.
Article in English | MEDLINE | ID: mdl-30798636

ABSTRACT

Neuropathic pain (NeP) is a complex chronic pain condition associated with nerve injury. Approximately, 7-10% of the general population across the globe is suffering from this traumatic condition, but the existing treatment strategies are inadequate to deliver pain relief and are associated with severe adverse effects. To overcome these limitations, lot of research is focussed on developing new molecules with high potency and fewer side effects, novel cell and gene-based therapies and modification of the previously approved drugs by different formulation aspects. Nanomedicine has attracted a lot of attention in the treatment of many diverse pathological conditions because of their unique physiochemical and biological properties. In this manuscript, we highlighted the emerging role of nanomedicine in different therapies (drug, cell and gene), also we emphasised on the challenges associated with nanomedicine such as development of well-characterised nanoformulation, scaling of batches with reproducible results and toxicity along with this we discussed about the future of nanomedicine in the treatment of neuropathic pain.


Subject(s)
Neuralgia/therapy , Cell- and Tissue-Based Therapy/methods , Central Nervous System Diseases/drug therapy , Central Nervous System Diseases/physiopathology , Chemotherapy, Adjuvant/methods , Drug Combinations , Drug Delivery Systems , Genetic Therapy/methods , Humans , Medical Marijuana/therapeutic use , Nanomedicine/methods , Neuralgia/drug therapy , Peripheral Nervous System Diseases/drug therapy , Peripheral Nervous System Diseases/physiopathology
15.
Life Sci ; 239: 116908, 2019 Dec 15.
Article in English | MEDLINE | ID: mdl-31610197

ABSTRACT

AIMS: An important obstacle on the way of cell-based therapy is the risk of tumorigenicity in the patients benefit from these transplanted cells due to undifferentiated cells which participate in transplantation. Curcumin, the main compound of spice turmeric -as one of the natural products-was demonstrated to possess effective anti-cancer properties, with no significant effect on normal cells in dose and/or time-dependent manner. Furthermore many studies have been accomplished using curcumin for diabetes treatment. Therefore in this study we examined the efficacy of IPCs treated with curcumin in vivo. MAIN METHODS: Differentiation efficiency investigated by flowcytometry. RNA extraction and real-time PCR performed for important genes in IPC differentiation and tumorigenesis including Insulin, Nestin, Ngn3, Pdx1, P21, and P53. Finally we investigated the efficiency of these differentiated and treated cells in diabetic rats. KEY FINDINGS: Our data indicates that nanocurcumin -in a specific dose-reduces the expression of Nestin with no significant effect on insulin expression in mRNA and protein level. Besides blood glucose level of diabetic rats which treated with DNC + cells, decreased from average 350 (mg/dI) to 100 (mg/dI). Checking out the pancreases of these rats, demonstrated that their endocrine segment was rebuilt. Moreover hematoxylin & eosin staining and IF results revealed that the Langerhans Islands were reformed. SIGNIFICANCE: IPCs' which treated with DNC were able to efficiently control the blood glucose level in diabetic rats which these cells were transplanted to them. Hence Curcumin has the potential to be employed in this kind of cell therapy.


Subject(s)
Curcumin/pharmacology , Diabetes Mellitus, Experimental/therapy , Animals , Blood Glucose/metabolism , Cell Differentiation/drug effects , Cell- and Tissue-Based Therapy/methods , Curcumin/metabolism , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 1/therapy , Disease Models, Animal , Humans , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Islets of Langerhans/metabolism , Male , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/metabolism , Pancreas/metabolism , Rats , Rats, Wistar
16.
Stem Cells Dev ; 28(21): 1424-1433, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31495275

ABSTRACT

Dome formation can occur in cultured tubular epithelial cells originating from various tissues, including the mammary gland and the kidney. The isolation and characterization of normal kidney epithelial stem cells that give rise to dome-forming tubular cells have never been reported. We attempted to isolate and characterize canine kidney epithelial stem cells using a simple cell culture method that we have previously used to isolate other adult human stem cells. Dome-forming kidney epithelial cells were derived from dissociated adult canine kidney tissues that were cultured in a modified keratinocyte serum-free medium supplemented with N-acetyl-l-cysteine, l-ascorbic acid 2-phosphate, nicotinamide, and fetal bovine serum. These cells exhibited high self-renewal capacity in long-term culture (growth for >13 months and 30 cumulative population doublings) and exhibited characteristics of stem cells, including (1) deficiency in gap junctional intercellular communication, (2) anchorage-independent growth, (3) expression of stem cell markers octamer-binding transcription factor 4 and SRY (sex determining region Y)-box 2, (4) expression of cell surface markers CD24 and CD133, and (5) multipotent differentiation into osteoblasts, adipocytes, chondrocytes, and dome-forming tubular cells. Most of these characteristics are shared by the well-known canine renal tubule-derived immortalized Madin-Darby Canine Kidney cell line. Furthermore, the putative canine kidney stem cells developed in this study formed budding tubule-like organoids on Matrigel and required high cell density (>4,000 cells/cm2) for sustained growth and confluency for dome formation. The signal transducer and activator of transcription-3 (STAT3) phosphorylation inhibitor, AG490, inhibited colony-forming efficiency and dome formation, whereas lipopolysaccharide, an activator of STAT3, increased colony-forming efficiency in a dose-dependent manner. These results are consistent with the hypothesis that high cell density induces STAT3 expression, which promotes both stem cell self-renewal and differentiation into tubular cells. Our novel cell culture method should be useful for the future development of normal human kidney stem cells for clinical applications and for studying mechanisms of nephrotoxicity.


Subject(s)
Epithelial Cells/cytology , Kidney Tubules/cytology , Multipotent Stem Cells/cytology , STAT3 Transcription Factor/metabolism , AC133 Antigen/metabolism , Animals , CD24 Antigen/metabolism , Cell Line , Cell- and Tissue-Based Therapy/methods , Dogs , Enzyme Inhibitors/pharmacology , Kidney Failure, Chronic/therapy , Lipopolysaccharides , Madin Darby Canine Kidney Cells , Octamer Transcription Factor-3/metabolism , SOXB1 Transcription Factors/metabolism , STAT3 Transcription Factor/antagonists & inhibitors , Tyrphostins/pharmacology
17.
Curr Opin Organ Transplant ; 24(5): 598-603, 2019 10.
Article in English | MEDLINE | ID: mdl-31397728

ABSTRACT

PURPOSE OF REVIEW: It has been increasingly common to use adipose tissue for regenerative and reconstructive purposes. Applications of autologous fat transfer and different stem cell therapies have significant limitations and adipose tissue engineering may have the potential to be an important strategy in the reconstruction of large tissue defects. A better understanding of adipogenesis will help to develop strategies to make adipose tissue more effective for repairing volumetric defects. RECENT FINDINGS: We provide an overview of the current applications of adipose tissue transfer and cellular therapy methods for soft tissue reconstruction, cellular physiology, and factors influencing adipogenesis, and adipose tissue engineering. Furthermore, we discuss mechanical properties and vascularization strategies of engineered adipose tissue, and its potential applications in the clinical settings. SUMMARY: Autologous fat tissue transfer is the standard of care technique for the majority of surgeons; however, high resorption rates, poor perfusion within a large volume fat graft and widely inconsistent graft survival are the main limitations. Adipose tissue engineering is a promising field to reach the first goal of producing adipose tissue which has more predictable survival and higher graft retention rates. Advancements of scaffold and vascularization strategies will contribute to metabolically and functionally more relevant adipose tissue engineering.


Subject(s)
Adipogenesis/physiology , Adipose Tissue/transplantation , Cell- and Tissue-Based Therapy/methods , Therapy, Soft Tissue , Tissue Engineering/methods , Humans , Tissue Scaffolds , Transplantation, Autologous
18.
Phytother Res ; 33(11): 2927-2937, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31452263

ABSTRACT

Curcumin is a dietary polyphenol and a bioactive phytochemical agent that possesses anti-inflammatory, antioxidant, anticancer, and chemopreventive properties. Some of the predominant activities of stem cells include regeneration of identical cells and the ability to maintain the proliferation and multipotentiality. However, these cells could be stimulated to differentiate into specific cell types. Curcumin protects some stem cells from toxicity and can stimulate proliferation and differentiation of stem cells. In the present review, we summarize the antioxidant, stemness activity, antiaging, and neuroprotective as well as wound healing and regenerative effects of curcumin.


Subject(s)
Cell- and Tissue-Based Therapy/methods , Curcumin/pharmacology , Stem Cells/drug effects , Animals , Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Humans , Regeneration/drug effects , Stem Cells/physiology , Wound Healing/drug effects
19.
J BUON ; 24(2): 615-621, 2019.
Article in English | MEDLINE | ID: mdl-31128014

ABSTRACT

PURPOSE: To explore the therapeutic efficacy and safety of the combination treatment of dendritic cells and cytokine-induced killers (DC-CIK) and sorafenib in patients with advanced hepatocellular carcinoma (HCC). METHODS: Patients diagnosed with advanced HCC and treated with DC-CIK and/or sorafenib in the Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University from January 2015 to January 2016 were retrospectively analyzed. HCC patients were divided into (A): control group (oral administration of sorafenib) and (B): observation group (oral administration of sorafenib combined with DC-CIK). Patients were followed up every 4-8 weeks. Overall survival and adverse events of each patient were recorded. Therapeutic efficacy was evaluated using the modified RECIST criteria. RESULTS: After treatment, ALT and TBIL were remarkably elevated in the control group and decreased in the observation group. No significant change in AFP level was seen in the control group after treatment, whereas it was remarkably decreased in the observation group. The efficacy rate was 16.7% and 51.4% in the control and observation group, respectively. Clinical benefit rate (CBR) was 41.9% and 88.6% in the control group and observation group, respectively. The median survival time of the control and observation group was 13.8 and 18.6 months, respectively. In the observation group there was a significant difference in the survival time between patients with Child-Pugh A and Child-Pugh B, respectively. CONCLUSIONS: DC-CIK combined with sorafenib could improve the tumor response rate and prolong overall survival of advanced HCC without increasing the incidence of adverse events. HCC patients achieve a more stable disease condition and longer overall survival with DC-CIK combined with sorafenib than those with individual sorafenib treatment.


Subject(s)
Carcinoma, Hepatocellular/therapy , Cytokine-Induced Killer Cells/transplantation , Liver Neoplasms/therapy , Sorafenib/administration & dosage , Aged , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/pathology , Cell- and Tissue-Based Therapy/methods , Combined Modality Therapy , Cytokine-Induced Killer Cells/immunology , Dendritic Cells/immunology , Dendritic Cells/transplantation , Female , Humans , Immunotherapy, Adoptive , Liver Neoplasms/pathology , Male , Middle Aged , Neoplasm Staging , Retrospective Studies , Sorafenib/adverse effects , Treatment Outcome
20.
Brain Res ; 1717: 147-159, 2019 08 15.
Article in English | MEDLINE | ID: mdl-30998931

ABSTRACT

Cortical injury elicits long-term cytotoxic and cytoprotective mechanisms within the brain and the balance of these pathways can determine the functional outcome for the individual. Cytotoxicity is exacerbated by production of reactive oxygen species, accumulation of iron, and peroxidation of cell membranes and myelin. There are currently no neurorestorative treatments to aid in balancing the cytotoxic and cytoprotective mechanisms following cortical injury. Cell based therapies are an emerging treatment that may function in immunomodulation, reduction of secondary damage, and reorganization of surviving structures. We previously evaluated human umbilical tissue-derived cells (hUTC) in our non-human primate model of cortical injury restricted to the hand area of primary motor cortex. Systemic hUTC treatment resulted in significantly greater recovery of fine motor function compared to vehicle controls. Here we investigate the hypothesis that hUTC treatment reduces oxidative damage and iron accumulation and increases the extent of the microglial response to cortical injury. To test this, brain sections from these monkeys were processed using immunohistochemistry to quantify oxidative damage (4-HNE) and activated microglia (LN3), and Prussian Blue to quantify iron. hUTC treated subjects exhibited significantly reduced oxidative damage in the sublesional white matter and iron accumulation in the perilesional area as well as a significant increase in the extent of activated microglia along white matter pathways. Increased perilesional iron accumulation was associated with greater perilesional oxidative damage and larger reconstructed lesion volume. These findings support the hypothesis that systemic hUTC administered 24 h after cortical damage decreases the cytotoxic response while increasing the extent of microglial activation.


Subject(s)
Brain Injuries/therapy , Cell- and Tissue-Based Therapy/methods , Motor Cortex/metabolism , Animals , Brain/metabolism , Cord Blood Stem Cell Transplantation/methods , Humans , Iron/metabolism , Macaca mulatta , Macrophage Activation/physiology , Male , Microglia/metabolism , Myelin Sheath/metabolism , Oxidation-Reduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL