Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
ACS Biomater Sci Eng ; 9(3): 1520-1540, 2023 03 13.
Article in English | MEDLINE | ID: mdl-36826450

ABSTRACT

Effective wound management imposes several challenges in clinical outcomes due to the complexity of the wound microenvironment, bacterial infections, impaired angiogenesis, aggravated inflammation, and enduring pain. In addition, adhesion on wet biological tissue is another extremely challenging task. Addressing all the issues is necessary for an effective wound healing process. Herein, we developed a unique multifunctional, adhesive composite hydrogel composed of gelatin, chitosan, polydopamine-coated bioactive glass (BG), and curcumin-capped silver nanoparticles (Cur-AgNPs) to target the multifaceted complexity of the wound. The PDA-coated BG serves multiple purposes: (1) adhesivity: catechol groups of PDA and Ca ion released from BG chelate the group present in the hydrogel network and surrounding tissues, (2) angiogenesis: promotes vascularization due to the release of Si from BG, and (3) BG also serves as the "reservoir" for the pain-relieving diclofenac sodium drug with a sustained-release behavior. Cur-AgNPs provide excellent bactericidal and anti-inflammatory properties to the composite hydrogel. In situ application of the composite hydrogel could serve the purpose of a "skin biomimetic" and work as a barrier along with bactericidal properties to inhibit the microbial growth. The multifunctional composite hydrogel (MCH) targeted multiple aspects of wound repair including pain alleviation, elimination of microbes (up to 99%), reduced inflammation, high adhesivity, and increased angiogenesis for effective skin regeneration. The MCH showed excellent wound healing potential as significant wound closure was observed at day 7 and also significantly upregulated the expression of crucial genes involved in the skin regeneration process along with increasing vascularization in the wound area.


Subject(s)
Hydrogels , Metal Nanoparticles , Humans , Adhesives/pharmacology , Silver/pharmacology , Wound Healing , Inflammation , Pain
2.
J Biomol Struct Dyn ; 41(10): 4405-4420, 2023 Jul.
Article in English | MEDLINE | ID: mdl-35491689

ABSTRACT

Despite substantial progress in surgery, managing multi-tissue injuries is strenuous to accomplish and requires a multi-staged serial treatment of individual tissues. Stimulated regeneration affects the complete structural and functional repair of both hard and soft tissues post-injury and thus serves as an attractive therapeutic option to target multi-tissue injuries. This study utilized data mining and structural analysis to identify a target that has the ability to evoke healing of the two most commonly injured tissues i.e., bone and muscle, and stimulate the inherent vascular connectivity between the tissues. To find out the multipotential molecule the gene expression profile from GSE34747 was extracted and processed to identify the differentially expressed genes (DEGs). The DEGs were then subjected to gene ontology enrichment analysis to filter out a target that is likely to regulate the multi-tissue regeneration. Further, STITCH and PubChem databases were screened to determine a stimulatory drug against the identified target molecule. Finally, the binding affinity and stability of the potential drug candidate(s) against the target were analysed by molecular docking and molecular dynamics simulation. The results revealed that bone morphogenetic protein-4 (BMP-4) was associated with the regulation of the multiple regeneration processes. The computational screening results suggested Retinoic acid and Torularhodin as potential drug candidates for the stimulation of BMP-4. Both drugs demonstrated slightly different but stable interactions with BMP-4, suggesting that the identified drug candidates are likely to serve as potential leads to further enhance tissues regeneration.Communicated by Ramaswamy H. Sarma.


Subject(s)
Molecular Docking Simulation
3.
Transplant Cell Ther ; 28(10): 667.e1-667.e10, 2022 10.
Article in English | MEDLINE | ID: mdl-35850428

ABSTRACT

Bone marrow (BM) transplantation has been used to treat malignant and nonmalignant BM-related disorders. Although an effective strategy, the procedure is associated with numerous complications, including graft rejection and nonspecific stem cell distribution. Avoidance of immune graft rejection has downsized the quantity of available stem cells, whereas nonspecific distribution necessitates the infusion of increased stem cell doses. A dual-purpose approach is needed to reduce the stem cell dose for transplantation. This used cell encapsulation and BM targeting to increase the quantity of transplanted stem cells reaching the BM. Cells were encapsulated with different biomaterials-chitosan (CS), polyallylamine hydrochloride (PAH), polystyrene sulfonate (PSS), and liposomes-and assessed for interaction with immune system components. The biomaterials were conjugated with vascular cell adhesion molecule 1 (VCAM1), a peptide that binds to BM-specific cell surface molecule, and evaluated for a successful transplantation. Encapsulated cells showed reduced interaction with antibody and cytokine without adverse effects on viability. Constitutive expression of VCAM1 was found to be specific on human bone marrow endothelial cells and was used for targeting by conjugating VCAM1-binding peptide to encapsulated cells. Peptide-conjugated-encapsulated stem cell formulations transplanted in irradiated mice with or without treatment with radioprotectant amifostine showed an increased percentage of cells reaching bone marrow. Post-transplantation survival along with blood count of neutrophils, platelets, and leukocytes was also enhanced for VCAM1-binding peptide-conjugated formulations with 100% survival demonstrated by peptide-conjugated CS/PSS/CS encapsulation formulation in irradiated mice for 4 weeks. Encapsulated cells retained their viability with increased shielding to the immune system, ensuring that graft rejection can be avoided. Transplanted encapsulated stem cells were distributed in a higher percentage to BM when conjugated to VCAM1-binding peptide, which could enable the use of lower stem cell doses. The peptide-conjugated CS/PSS/CS encapsulation system conferred 100% survival in irradiated mice, with increased regeneration demonstrating the ability to treat radiation-exposed victims.


Subject(s)
Amifostine , Chitosan , Animals , Biocompatible Materials , Bone Marrow , Cytokines , Endothelial Cells , Hematopoietic Stem Cells , Humans , Liposomes , Mice , Vascular Cell Adhesion Molecule-1
4.
Hum Cell ; 35(1): 37-50, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34800267

ABSTRACT

Reactive Oxygen Species are chemically unstable molecules generated during aerobic respiration, especially in the electron transport chain. ROS are involved in various biological functions; any imbalance in their standard level results in severe damage, for instance, oxidative damage, inflammation in a cellular system, and cancer. Oxidative damage activates signaling pathways, which result in cell proliferation, oncogenesis, and metastasis. Since the last few decades, mesenchymal stromal cells have been explored as therapeutic agents against various pathologies, such as cardiovascular diseases, acute and chronic kidney disease, neurodegenerative diseases, macular degeneration, and biliary diseases. Recently, the research community has begun developing several anti-tumor drugs, but these therapeutic drugs are ineffective. In this present review, we would like to emphasize MSCs-based targeted therapy against pathologies induced by ROS as cells possess regenerative potential, immunomodulation, and migratory capacity. We have also focused on how MSCs can be used as next-generation drugs with no side effects.


Subject(s)
Cardiovascular Diseases/etiology , Cardiovascular Diseases/therapy , Inflammation/etiology , Inflammation/therapy , Kidney Diseases/etiology , Kidney Diseases/therapy , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells , Neoplasms/etiology , Neoplasms/therapy , Neurodegenerative Diseases/etiology , Neurodegenerative Diseases/therapy , Oxidative Stress , Reactive Oxygen Species/adverse effects , Animals , Cardiovascular Diseases/pathology , Electron Transport , Humans , Inflammation/pathology , Kidney Diseases/pathology , Mice , Neoplasms/pathology , Neurodegenerative Diseases/pathology
5.
Mol Cell Biochem ; 476(11): 3987-4002, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34195882

ABSTRACT

In December 2019, the emergence and expansion of novel and infectious respiratory virus SARS-CoV-2 originated from Wuhan, China caused an unprecedented threat to the public health and became a global pandemic. SARS-CoV-2 is an enveloped, positive sense and single stranded RNA virus belonging to genera betacoronavirus, of Coronaviridae family. The viral genome sequencing studies revealed 75-80% similarity with SARS-CoV. SARS-CoV-2 mainly affects the lower respiratory system and may progress to pneumonia and Acute Respiratory Distress Syndrome (ARDS). Apart from life-threatening situations and burden on the global healthcare system, the COVID-19 pandemic has imposed several challenges on the worldwide economics and livelihood. The novel pathogen is highly virulent, rapidly mutating and has a tendency to cross the species boundaries such as from bats to humans through the evolution and natural selection from intermediate host. In this review we tried to summarize the overall picture of SARS-CoV-2 including origin/ emergence, epidemiology, pathogenesis, genome organization, comparative analysis with other CoVs, infection and replication mechanism along with cellular tropism and immunopathogenesis which will provide a brief panoramic view about the virus and disease.


Subject(s)
COVID-19/epidemiology , SARS-CoV-2/physiology , Animals , COVID-19/immunology , COVID-19/pathology , COVID-19/virology , Genome, Viral , Humans , Pandemics , SARS-CoV-2/genetics
6.
Int J Nanomedicine ; 16: 3833-3859, 2021.
Article in English | MEDLINE | ID: mdl-34113102

ABSTRACT

BACKGROUND: Hematopoietic stem cells' commitment to myelopoiesis builds immunity to prevent infection. This process is controlled through transcription factor, especially Purine rich box 1 (PU.1) protein, which plays a central role in regulating myelopoiesis. The ß3/ß4 region of PU.1 accommodates a coactivator transcription factor, c-Jun, to activate myelopoiesis. However, an erythroid transcription factor, GATA-1, competes with c-Jun for the ß3/ß4 region, abolishing myelopoiesis and promoting erythropoiesis. This competitive regulation decides the hematopoietic stem cells' commitment towards either erythroid or myeloid lineage. METHODS: Therefore, this study investigated the in vitro and in vivo effect of novel synthetic PU.1 ß3/ß4 mimic peptide analogs and peptide-loaded hydrophilic poly(D,L-lactide-co-glycolide) (PLGA) nanoparticles. RESULTS: The designed peptides significantly increase the expression of corresponding myeloid markers, specifically CD33 in vitro. However, the in vivo delivery of peptide-loaded PLGA nanoparticles, which have sustained release effect of peptides, increases 10.8% of granulocytes as compared to control. CONCLUSION: The observations showed that the fabricated nanoparticles protected the loaded peptides from the harsh intracellular environment for a longer duration without causing any toxicity. These findings highlight the possibility to use these peptides and peptide-loaded nanoparticles to increase hematopoietic stem cell commitment to myeloid cells in case of opportunistic infection.


Subject(s)
Erythropoiesis , GATA1 Transcription Factor/metabolism , Gene Expression Regulation , Hematopoietic Stem Cells/cytology , Myelopoiesis , Peptide Fragments/metabolism , Proto-Oncogene Proteins/metabolism , Trans-Activators/metabolism , Cell Differentiation , GATA1 Transcription Factor/genetics , Hematopoietic Stem Cells/metabolism , Humans , Proto-Oncogene Proteins/genetics , Trans-Activators/genetics
7.
Eur J Neurosci ; 2021 May 01.
Article in English | MEDLINE | ID: mdl-33932318

ABSTRACT

Cellular transplantation of stem cells can be a beneficial treatment approach for neurodegenerative diseases such as traumatic brain injury (TBI). In this study, we investigated the proliferation and differentiation potential of infused mesenchymal stem cells (MSCs) after localisation at the injury site. We evaluated the appropriate homing of infused MSCs through immunohistochemistry, followed by Y-chromosome-specific polymerase chain reaction and fluorescent in situ hybridisation analyses. The proliferation and differentiation of infused MSCs were analysed using exogenous cell tracer 5'-bromo-2'-deoxyuridine (BrdU) labelling and neuronal specific markers, respectively. Structural and functional recovery in TBI mice were examined by performing magnetic resonance imaging and different behavioural assessments, respectively. Results demonstrated a significantly high number of BrdU-positive cells in the lesion region in the MSC-infused group compared with control and TBI groups. Infused MSCs were well differentiated into neural-like cells and expressed significantly more neural markers (neuronal nuclear antigen [NeuN], microtubule-associated protein 2 [MAP2] and glial fibrillary acid protein [GFAP]). Improved tissue abnormalities as well as functional behaviours were observed in MSC-infused TBI mice, implying the substantial proliferation and differentiation of infused MSCs. Our findings support the neuroprotective response and efficacy of MSCs after transplantation in TBI mice, and MSCs may serve as potential therapeutic candidates in regenerative medicine.

8.
Stem Cell Rev Rep ; 17(1): 113-131, 2021 02.
Article in English | MEDLINE | ID: mdl-32920752

ABSTRACT

The novel virus, Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) caused the Corona Virus Disease-2019 (COVID-19) outbreak in Wuhan, Hubei province of China. This virus disseminated rapidly and reached to an unprecedented pandemic proportion in more than 213 nations with a large number of fatalities. The hypersecretion of pro-inflammatory cytokines is the main cause of mortality and morbidity due to COVID-19, therefore strategies that avert the cytokine storm may play a crucial role in abating the severity of COVID-19. This review highlights the minute details of SARS-CoV-2, its genomic organization, genomic variations within structural and non-structural proteins and viral progression mechanism in human beings. The approaches like antiviral strategies are discussed, including drugs that obstruct viral propagation and suppress the pro-inflammatory cytokines. This compilation emphasizes Mesenchymal Stem Cells (MSCs) based therapy alone or in combination with other therapeutics as an attractive curative approach for COVID-19 pandemic. The MSCs and its secretome, including antimicrobial peptides (AMPs) have various capabilities, for instance, immunomodulation, regeneration, antimicrobial properties, potential for attenuating the cytokine storm and bare minimum chances of being infected with SARS-CoV-2 virus. The immunomodulatory property of MSCs affects inflammatory state and regulates immune response during SARS-CoV-2 infection. However, as of now, there is no WHO-approved MSCs based therapy for the treatment of COVID-19 infection. Graphical abstract.


Subject(s)
COVID-19/therapy , Mesenchymal Stem Cell Transplantation , Pandemics , SARS-CoV-2/pathogenicity , COVID-19/epidemiology , COVID-19/immunology , COVID-19/virology , Cytokines/immunology , Humans , Immunomodulation/immunology , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/immunology
9.
Gene Expr Patterns ; 37: 119128, 2020 09.
Article in English | MEDLINE | ID: mdl-32707324

ABSTRACT

Differentiation of the HSCs into myeloid lineage is primarily monitored by transcription factor PU.1. GATA1 acts as a negative regulator by antagonizing the function of PU.1 by bindings its ß3/ß4 domain. In this study, a mutation was induced in PU.1 which blocks its interaction with GATA1. The pure form of this mutant protein i.e Y244D was loaded on poly (lactic-co-glycolic acid) nanoparticles to transfect CD34+ cells, which act as a selective tool to enhance the myelopoiesis, as confirmed by flow cytometry analysis. Further, microarray data analysis was performed to gather information on myelopoiesis specific signaling pathways and genes involved in myelopoiesis like CCL2, S100A8, and S100A9, which were also found to be significantly upregulated in the mutant form. Different molecular functions like antioxidant activity, signal transduction, developmental processes, and biological adhesion were analyzed. This study potentially signifies that PU.1 mutant is highly efficient in myelopoiesis.


Subject(s)
Antigens, CD34/immunology , Gene Expression Profiling , Hematopoietic Stem Cells/metabolism , Myelopoiesis/genetics , Cell Differentiation , Cytokines/metabolism , Flow Cytometry , Hematopoietic Stem Cells/immunology , Humans , MAP Kinase Signaling System , Mutation , Nanoparticles/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Protein Binding , Proto-Oncogene Proteins/genetics , Trans-Activators/genetics
10.
Exp Cell Res ; 390(1): 111954, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32156602

ABSTRACT

Bone marrow failure is the primary cause of death after nuclear accidents or intentional exposure to high or low doses of ionizing radiation. Hematopoietic stem cell transplantation is the most potent treatment procedure for patients suffering from several hematopoietic malignancies arising after radiation injuries. Successful hematopoietic recovery after transplantation depends on efficient homing and subsequent engraftment of hematopoietic stem cells in specific niches within the bone marrow. It is a rapid and coordinated process in which circulating cells actively enter the bone marrow through the process known as transvascular migration, which involves the tightly regulated relay of events that finally leads to homing of cells in the bone marrow. Various adhesion molecules, chemokines, glycoproteins, integrins, present both on the surface of stem cells and sinusoidal endothelium plays a critical role in transvascular migration. But despite having an in-depth knowledge of homing and engraftment and the key events that regulate it, we are still not completely able to avoid graft failures and post-transplant mortalities. This deems it necessary to design a flawless plan for successful transplantation. Here, in this review, we will discuss the current clinical methods used to overcome graft failures and their flaws. We will also discuss, what are the new approaches developed in the past 10-12 years to selectively deliver the hematopoietic stem cells in the bone marrow by adopting proper targeting strategies that can help revolutionize the field of regenerative and translational medicine.


Subject(s)
Cell Movement , Graft Survival , Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cells/physiology , Animals , Hematopoiesis , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Stem Cell Niche
11.
Sci Rep ; 10(1): 156, 2020 01 13.
Article in English | MEDLINE | ID: mdl-31932650

ABSTRACT

Platelet microvesicles (pMVs) are submicron-sized heterogeneous vesicles released upon activation and contain several membrane receptors and proteins (CD41, CD61, CD62, CXCR4, PAR-1, etc.). We have revealed their ability to adhere to the triblock copolymer pluronic-F127 (PF127) and form a platelet microvesicular nanocloud which has the potential to enhance the transvascular migration of hematopoietic stem cells across the sinusoidal endothelium to the bone marrow. Besides, the pMVs nanoclouds bestow survival benefits when present on the cells used for infusion, particularly with PF127-stabilized with chitosan-alginate (PF127-CA HSCs). The vesicles were found to be firmly associated with PF127 in the nanocloud, which was detected by confocal laser scanning microscopy. The abrogation of CXCR4/SDF-1 axis regulating the transmigration of the cells by antagonist AMD3100 revealed that the enriched CXCR4 receptors on pMVs robustize the transmigration of the infused cells. The homing of the cells led to effective engraftment and faster regeneration of the critical blood lineages, which elicited 100% survival of the mice receiving lethal doses of radiation. The Human Long-Term Culture Initiating Cells (LTC-ICs), Severe Combined Immunodeficient (SCID) - Repopulating Cells (SRCs) and Colony Forming Cells (CFCs) responsible for the regeneration, but present in extremely low numbers in the infused cell dose, have enabled the cells to reach the bone marrow in high numbers. This potential of the PF127 to sequester the pMVs and its application to achieve over 10-fold delivery of HSCs across the trans-endothelial checkpoint has so far not been reported. Thus, this mechanistic innovation is a potential post-exposure life-saving regimen capable of circumventing the irreparable damage to the bone marrow caused by lethal doses of radiation.


Subject(s)
Blood Platelets/chemistry , Bone Marrow Cells/cytology , Cell-Derived Microparticles/chemistry , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Nanocomposites/administration & dosage , Polyethylenes/chemistry , Polypropylenes/chemistry , Animals , Apoptosis , Bone Marrow Cells/metabolism , Bone Marrow Cells/radiation effects , Cell Movement , Cell Proliferation , Cells, Cultured , Female , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, Nude , Mice, SCID , Nanocomposites/chemistry
12.
Methods Mol Biol ; 2150: 93-111, 2020.
Article in English | MEDLINE | ID: mdl-31802432

ABSTRACT

Traumatic brain injury (TBI) is a leading cause of death and disability. The condition is difficult to treat owing to its heterogeneous nature and complex biological pathways. Stem cell transplantation is an emerging self-deliverable therapeutic modality which could immensely improve the invigorating management of the problem. The synergistic interaction of the stem cells with the paracrine niche molecules at the site of injury is an end point that decides the cells' effective tissue-forming regenerative response. Thus, noninvasive monitoring and tracking of the infused stem cells is quite decisive after transplantation. Here, we have designed and validated a distinctive in vivo magnetic resonance imaging protocol to monitor the transplanted mesenchymal stem cells (MSCs) longitudinally in TBI-induced mice. We have further described the synthesis of improved transverse relaxivity contrast agent, a protocol for the efficient labelling of MSCs, preparation of a TBI model system in mice, and the imaging and tracking of the implanted stem cells at the injury site through 7T MRI. MGE-T2∗ imaging in association with relaxometry-based quantitative assessment using absolute bias correction provided a suitable mechanism to monitor and track the infused labelled stem cells at the TBI site. High transverse relaxivity negative contrast agent synthesis, MSC labelling procedure, and quantitative T2∗ time measurement normalized with absolute bias correction are the key features of this protocol. This procedure has immense application potential and could therefore be extrapolated to stem cell tracking during the treatment of various diseases.


Subject(s)
Brain Injuries, Traumatic/diagnostic imaging , Brain Injuries, Traumatic/pathology , Cell Tracking/methods , Magnetic Resonance Imaging , Staining and Labeling , Animals , Cell Separation , Contrast Media/chemistry , Disease Models, Animal , Ferric Compounds/chemistry , Magnetite Nanoparticles/chemistry , Mice, Inbred BALB C
13.
Life Sci ; 238: 116981, 2019 Dec 01.
Article in English | MEDLINE | ID: mdl-31639394

ABSTRACT

AIM: Endothelial cell damage is critical to understand since its presence in the entire body makes the damage widespread instead of being localized. Being a major component of stem cell niche in bone marrow, deems it essential to gain knowledge of the damage to endothelium associated with bone marrow. Since radiation exposure has become common to numerous therapeutic modalities, its effects on bone marrow and its endothelial cells are crucial to understand. MATERIAL & METHODS: Microarray analysis was performed on irradiated human bone marrow endothelial cells (hBMECs) with and without prior treatment with radioprotectant amifostine to assess the effects of radiation on signalling pathways and the subsequent changes in pathways when treated with radioprotectant prior to radiation exposure. KEY FINDINGS: It was seen that adhesion pathways that were usually inactivated under normal circumstances were stimulated post radiation. However, where in the case of radiation exposure, these adhesion pathways included leukocyte adhesion and migration; in the case of radioprotected conditions the pathways revolve around cell-substrate adhesion and cell spreading. Genes like ROCK1, FLNA, RAC1, PRKCZ and MAP3K8 were seen to regulate the molecular switch between leukocyte-cell adhesion to cell-substrate adhesion. SIGNIFICANCE: Our study demonstrated that irradiated endothelium supports leukocyte adhesion and migration but shifts to substrate adhesion dependent cell spreading under radioprotected conditions in order to repair the monolayer damage from the radiation. The genes responsible for the shift were identified and can be employed to manipulate cell adhesion characteristics for the treatment of diseases caused by radiation or inflammation.


Subject(s)
Amifostine/pharmacology , Biomarkers/metabolism , Bone Marrow/metabolism , Cell Adhesion , Endothelium, Vascular/metabolism , Gamma Rays , Leukocytes/metabolism , Bone Marrow/drug effects , Bone Marrow/radiation effects , Cells, Cultured , Endothelium, Vascular/drug effects , Endothelium, Vascular/radiation effects , Gene Expression Regulation/drug effects , Gene Expression Regulation/radiation effects , Gene Regulatory Networks/drug effects , Gene Regulatory Networks/radiation effects , Humans , Leukocytes/drug effects , Leukocytes/radiation effects , Radiation-Protective Agents/pharmacology
14.
Front Genet ; 10: 554, 2019.
Article in English | MEDLINE | ID: mdl-31263479

ABSTRACT

The nuclear receptors known as peroxisome proliferator activated receptor gamma (PPARG) are lipid-activated transcription factors that have emerged as key regulators of inflammation. PPARG ligands have been shown to have an anti-proliferative effect on a variety of cancers. These ligands can induce apoptosis via TP53 (Tumor protein p53) or ERK1/2 (Extracellular signal-regulated kinases 1/2) (EPHB2) pathways. However, the exact mechanism is not known. PPAR, a type II nuclear hormone receptor deserves attention as a selective target for radiotherapy. Our study examines the potential of selective agonism of PPARG for radiation therapy in non-small cell lung carcinoma (NSCLC). We found that the overexpression of PPARG protein as well as its induction using the agonist, rosiglitazone was able to stimulate radiation-induced cell death in otherwise radio resistant NSCLC A549 cell line. This cell death was apoptotic and was found to be BAX (BCL2 associated X) mediated. The treatment also inhibited radiation-induced AKT (Protein Kinase B) phosphorylation. Interestingly, the ionising radiation (IR) induced apoptosis was found to be inversely related to TP53 levels. A relatively significant increase in the levels of radiation induced apoptosis was observed in H1299 cells (TP53 null) under PPARG overexpression condition further supporting the inverse relationship between apoptosis and TP53 levels. The combination of PPARG agonist and radiation was able to induce apoptosis at a radiation dose at which A549 and H1299 are radioresistant, thus confirming the potential of the combinatorial strategy. Taken together, PPARG agonism was found to invigorate the radiosensitising effect and hence its use in combination with radiotherapy is expected to enhance sensitivity in otherwise resistant cancer types.

15.
PLoS One ; 13(12): e0210133, 2018.
Article in English | MEDLINE | ID: mdl-30592765

ABSTRACT

[This corrects the article DOI: 10.1371/journal.pone.0206364.].

16.
PLoS One ; 13(11): e0206364, 2018.
Article in English | MEDLINE | ID: mdl-30388134

ABSTRACT

Enhancement of hematopoietic stem cells (HSCs) proliferation is a central aim in bone marrow transplantation (BMT). A stem cell factor (SCF) and c-Kit mediated extracellular signaling trigger proliferation of HSCs. This signaling is negatively regulated by protein tyrosine phosphatases (PTPs), SHP-1 and SHP-2. Although NSC87877 (N) is known to inhibit SHP-1/SHP-2, c-Kit-mediated HSCs proliferation by inhibiting SHP-1/SHP-2 has not been reported. This study investigated the combined effect of SCF (S) and N in c-Kit mediated proliferation and underlying mechanisms. The growth of human megakaryoblastic cell line, MO7e and HSCs, upon treatment with S and N alone, and in combination was assessed by PrestoBlue staining. The expression of c-Kit, phosphorylated c-Kit, SHP-1/SHP-2 and HePTP inhibition using S and N treatment were evaluated in the MO7e cells. Megakaryoblast cell proliferation was determined by quantification of Ki-67+, S-phase, BrdU+ and CFDA-SE+ cells using flow cytometry. The combination of S and N leads to enhanced cell growth compared with either S or N alone. Collectively, the results reveal a novel mechanism by which S in combination with N significantly enhances proliferation of human megakaryoblast cells. The pretreatment of N before S enhances proliferation of cells than S alone. This promising combination would likely play an essential role in enhancing the proliferation of cells.


Subject(s)
Megakaryocyte Progenitor Cells/cytology , Megakaryocyte Progenitor Cells/drug effects , Proto-Oncogene Proteins c-kit/metabolism , Quinolines/pharmacology , Stem Cell Factor/pharmacology , Cell Proliferation/drug effects , Drug Interactions , Gene Expression Regulation/drug effects , Humans , Megakaryocyte Progenitor Cells/metabolism , Phosphorylation/drug effects , Signal Transduction/drug effects
17.
Life Sci ; 214: 84-97, 2018 Dec 01.
Article in English | MEDLINE | ID: mdl-30308182

ABSTRACT

The biological mechanisms underlying the effects of stem cell factor (SCF) and an inhibitor, NSC87877 (N) of the c-Kit negative regulator (SHP-1 and SHP-2) on cell proliferation are different. Therefore, we compared the cell's response to these two either alone or in combination in K562 cells. Binding of SCF (S) to c-Kit induces dimerization that activates its kinase activity. The activated c-Kit undergoes autophosphorylation at tyrosine residues that serve as a docking site for signal transduction molecules containing SH2 domains. Predominantly, the phosphotyrosine 568 (pY568) in Juxtamembrane (JM) region of c-Kit interacts with adaptor protein APS, Src family kinase, and SHP-2, while phosphotyrosine 570 (pY570) interacts with the SHP-1 and the adaptor protein Shc. The dephosphorylation of phosphotyrosine residues by SHP-1/SHP-2 leads to inhibition of c-Kit proliferative signaling. A chemical molecule, N is reported to inhibit the enzymatic activity of SHP-1/SHP-2, but its effect on c-Kit-mediated proliferation has not been studied yet. Thus, this work aims at examining the effect of the combination of S and N on cells growth as compared to individual treatment. The present study is performed with erythroleukemic K562 cells, chosen for its mRNA expression concerning the c-Kit, and SHP-1/SHP-2. Interestingly, proliferation assay showed that combination significantly increased proliferation when G1 sorted K562 cells were used. These changes were significantly higher when K562 cells were initially treated with N followed by S treatment. Collectively, these results give mechanistic insight into the proliferation enhancement of bone marrow transplantation through the synergistic effect of S and N by inhibiting SHP-1/SHP-2. The study gives solid evidence that S and N combination can be used to enhance cell proliferation/growth.


Subject(s)
Erythroid Cells/drug effects , Proto-Oncogene Proteins c-kit/antagonists & inhibitors , Stem Cell Factor/pharmacology , Cell Cycle/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Proliferation/physiology , Drug Synergism , Erythroid Cells/cytology , Erythroid Cells/metabolism , Humans , K562 Cells , Ki-67 Antigen/metabolism , Phosphorylation , Proliferating Cell Nuclear Antigen/metabolism , Proto-Oncogene Proteins c-kit/metabolism , Stem Cell Factor/metabolism
18.
J Microencapsul ; 35(6): 522-532, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30146922

ABSTRACT

Natural killer and cytotoxic CD8+ T cells are involved in the rapid clearance of cells which express foreign antigens. Hence, these cells are crucial elements of the vertebrate immune system. However, these benefits turn problematic when they cause transplant rejection through their direct cytotoxic effects on donor organs/cells, which is attributed to the human leukocyte antigen disparity. To overcome these limitations, a strategy has been devised wherein the above effects can be minimised by shielding the cells through encapsulation. The layer-by-layer approach was employed for encapsulation using chitosan and alginate. Confocal microscopy, scanning electron microscopy confirmed the complete shielding of cells. Encapsulation did not affect cell viability as no toxicity was discerned. Calcein release assay was applied for assessing cell-mediated cytotoxicity. It was observed that the encapsulated cells underwent lesser lysis, thereby revealing the potential that this approach offers in reducing conditions such as graft failure and hypersensitivity.


Subject(s)
Alginates/chemistry , Biocompatible Materials/chemistry , CD8-Positive T-Lymphocytes/immunology , Chitosan/chemistry , Killer Cells, Natural/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Line , Cell Survival , Cytotoxicity, Immunologic , Drug Compounding , Fluoresceins/metabolism , Humans , Killer Cells, Natural/metabolism , Receptors, Cell Surface/immunology , Static Electricity
19.
Stem Cell Rev Rep ; 14(6): 888-900, 2018 Dec.
Article in English | MEDLINE | ID: mdl-29911289

ABSTRACT

Stem cells transplantation has emerged as a promising alternative therapeutic due to its potency at injury site. The need to monitor and non-invasively track the infused stem cells is a significant challenge in the development of regenerative medicine. Thus, in vivo tracking to monitor infused stem cells is especially vital. In this manuscript, we have described an effective in vitro labelling method of MSCs, a serial in vivo tracking of implanted stem cells at traumatic brain injury (TBI) site through 7 T magnetic resonance imaging (MRI). Proper homing of infused MSCs was carried out at different time points using histological analysis and Prussian blue staining. Longitudinal in vivo tracking of infused MSCs were performed up to 21 days in different groups through MRI using relaxometry technique. Results demonstrated that MSCs incubated with iron oxide-poly-L-lysine complex (IO-PLL) at a ratio of 50:1.5 µg/ml and a time period of 6 h was optimised to increase labelling efficiency. T2*-weighted images and relaxation study demonstrated a significant signal loss and effective decrease in transverse relaxation time on day-3 at injury site after systemic transplantation, revealed maximum number of stem cells homing to the lesion area. MRI results further correlate with histological and Prussian blue staining in different time periods. Decrease in negative signal and increase in relaxation times were observed after day-14, may indicate damage tissue replacement with healthy tissue. MSCs tracking with synthesized negative contrast agent represent a great advantage during both in vitro and in vivo analysis. The proposed absolute bias correction based relaxometry analysis could be extrapolated for stem cell tracking and therapies in various neurodegenerative diseases.


Subject(s)
Brain Injuries, Traumatic/therapy , Cell Tracking/methods , Ferric Compounds/chemistry , Magnetic Resonance Imaging , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Animals , Brain Injuries, Traumatic/pathology , Cell Survival , Limit of Detection , Male , Mice, Inbred BALB C , Phenotype
20.
Toxicol In Vitro ; 50: 179-189, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29577976

ABSTRACT

The use of iron oxide nanoparticles for different biomedical applications, hold immense promise to develop negative tissue contrast in magnetic resonance imaging (MRI). Previously, we have optimized the labelling of mesenchymal stem cells (MSCs) with iron oxide nanoparticles complexed to different transfection agents like poly-l-lysine (IO-PLL) and protamine sulfate (Fe-Pro) on the basis of relaxation behaviour and its biological expressions. However, there is a distinct need to investigate the biocompatibility and biosafety concerns coupled with its cytotoxicity and genotoxicity. This study was prepared to evaluate the viability of cells, generation of ROS, changes in actin cytoskeleton, investigation of cell death, level of GSH and TAC, activities of SOD and GPx, and stability of DNA in MSCs after labelling. Results demonstrated a marginal alteration in toxicological parameters like ROS generation, cell length, actin cytoskeleton, total apoptosis and DNA damage was detected after stem cell labelling. Insignificant depletion of GSH and SOD level, and increase in GPx and TAC level in MSCs were measured after labelling with IO-PLL and Fe-Pro complexes, which later on recovered and normalized to its baseline. This MSCs labelling could provide a reference guideline for toxicological analysis and relaxometry based in vivo MRI detection.


Subject(s)
Contrast Media/toxicity , Ferric Compounds/toxicity , Mesenchymal Stem Cells/drug effects , Metal Nanoparticles/toxicity , Animals , Apoptosis/drug effects , Cells, Cultured , Comet Assay , Glutathione/metabolism , Glutathione Peroxidase/metabolism , Male , Mesenchymal Stem Cells/metabolism , Mice, Inbred BALB C , Necrosis/chemically induced , Reactive Oxygen Species/metabolism , Staining and Labeling , Superoxide Dismutase/metabolism , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...