Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 65
Filter
Add more filters










Publication year range
1.
PLoS One ; 17(11): e0275245, 2022.
Article in English | MEDLINE | ID: mdl-36383519

ABSTRACT

We conducted the first profitability comparison study across health care industries in the United States, using the DuPont Analysis framework. The combination of Return on Equity (ROE) and ROE volatility was used to provide a comprehensive "risk-return" approach for profitability comparison. Based on the 2010-2019 financial disclosures of 1,231 publicly traded health care companies in the U.S. that reported positive assets and equity, we estimated the industry-specific fixed effects on ROE and its three components-profit margin, asset utilization, and financial leverage-for ten industries in the health care sector, classified by the Global Industry Classification Standard (GICS). For each industry, we also estimated its fixed effects on ROE volatility. We found that the pharmaceuticals industry and biotechnology industry have lower ROE-mainly driven by their relatively low profit margin and low assets utilization-and higher ROE volatility than other health care industries. We also found that the health care facilities industry relies most on debt financing. This study demonstrates a holistic approach for profitability comparison across industries.


Subject(s)
Drug Industry , Health Care Sector , United States
2.
J Immunotoxicol ; 19(1): 61-73, 2022 12.
Article in English | MEDLINE | ID: mdl-35901199

ABSTRACT

Nickel titanium (NiTi, or Nitinol) alloy is used in several biomedical applications, including cardiac, peripheral vascular, and fallopian tube stents. There are significant biocompatibility issues of metallic implants to nickel ions and nano-/micro-sized alloy particles. Our laboratories have recently shown that microscale CoCr wear particles from metal-on-metal hips crosslink with the innate immune signaling Toll-like receptor 4 (TLR4), prompting downstream signaling that results in interleukin (IL)-1ß and IL-8 gene expression. In vivo, NiTi alloy can also generate wear particles on the nanoscale (NP) that have thus far not been studied for their potential to induce inflammation and angiogenesis that can, in turn, contribute to implant (e.g. stent) failure. Earlier studies by others demonstrated that nickel could induce contact hypersensitivity by crosslinking the human, but not the mouse, TLR4. In the present work, it is demonstrated that NiCl2 ions and NiTi nanoparticles induce pro-inflammatory and pro-angiogenic cytokine/chemokine expression in human endothelial and monocyte cell lines in vitro. These observations prompt concerns about potential mechanisms for stent failure. The data here showed a direct correlation between intracellular uptake of Ni2+ and generation of reactive oxygen species. To determine a role for nickel and NiTi nanoparticles in inducing angiogenesis in vivo, 1-cm silicone angioreactors were implanted subcutaneously into athymic (T-cell-deficient) nude mice. The angioreactors contained Matrigel (a gelatinous protein mixture that resembles extracellular matrix) in addition to one of the following: PBS (negative control), VEGF/FGF-2 (positive control), NiCl2, or NiTi NP. The implantation of angioreactors represents a potential tool for quantification of angiogenic potentials of medical device-derived particles and ions in vivo. By this approach, NiTi NP were found to be markedly angiogenic, while Ni2+ was less-so. The angioreactors may provide a powerful tool to examine if debris shed from medical devices may promote untoward biological effects.


Subject(s)
Metal Nanoparticles , Nickel , Alloys , Animals , Humans , Inflammation , Ions , Mice , Mice, Nude , Nanoparticles , Nickel/pharmacology , Titanium/adverse effects , Toll-Like Receptor 4
3.
Int J Environ Health Res ; 32(6): 1403-1417, 2022 Jun.
Article in English | MEDLINE | ID: mdl-33573386

ABSTRACT

Chronic Obstructive Pulmonary Disease (COPD) is attributable to household air pollution and is known to increase the Disability Adjusted Life Years (DALYs), morbidity and mortality and women are most susceptible groups for the exposure. In order to understand the global risk among women with COPD due to exposure of household air pollutants, an evidence-based systematic review and meta-analysis was conducted. Meta regression analysis was carried out to identify potential sources of heterogeneity. The summary estimates of the included studies showed higher prevalence of COPD due to biomass fuel exposure in women. Clinical diagnosis has shown more risk of COPD prevalence compared to diagnosis based on spirometer test alone. However, the data between included studies for both clinical and spirometry-based studies showed higher heterogeneity. The present meta-data analysis has shown that household air pollutants may be a factor associated with increased risk of COPD in women.


Subject(s)
Air Pollutants , Air Pollution, Indoor , Air Pollution , Pulmonary Disease, Chronic Obstructive , Air Pollutants/analysis , Air Pollutants/toxicity , Air Pollution/analysis , Air Pollution, Indoor/adverse effects , Air Pollution, Indoor/analysis , Biomass , Female , Humans , Male , Prevalence , Pulmonary Disease, Chronic Obstructive/chemically induced , Pulmonary Disease, Chronic Obstructive/epidemiology , Risk Factors
4.
J Colloid Interface Sci ; 606(Pt 2): 2024-2037, 2022 Jan 15.
Article in English | MEDLINE | ID: mdl-34749449

ABSTRACT

The remediation of non-reactive phosphate pollutants in the aquatic system is essential for protecting the ecological niche. In this work, a highly robust protein nanoparticles networked rare-earth metal carbonate-grafted bio-composite membrane (abbreviated as REMC) was fabricated via chemical crosslinking of three-dimensional (3D) hierarchical lanthanum carbonate (mREM) and casein nanoparticles (CsNPs) for selective rejection of non-reactive phosphates. The main components of the REMC membrane are mREM and CsNPs, which were prepared via SDS/CTAB templated homogeneous precipitation and the coacervation/desolvation hybrid method, respectively. The active lanthanum ion (La3+) on the 3D spherulitic surface of mREM exhibited excellent phosphate adsorption capacity (maximum adsorption capacity was 358 mg.g-1) across a wide pH range and in a multi-ionic environment. A series of batch testing and characterizations revealed that the active La3+ and dominating phosphate centers in the REMC membrane framework enable non-enzymatic phosphatase-like activity, cleaving the phosphate ester bond of organic phosphates and releasing free phosphate anions. These released phosphate ions are retained in the REMC membrane via an ion exchange mechanism, where they contribute to improved phosphate removal capacities. Furthermore, CsNPs have a dual function in the membrane, acting as a matrix in the REMC membrane framework and contributing to phosphate ion sequestrations in a synergistic manner. The catalysis of para-nitrophenyl phosphates (pNPP) to paranitrophenol (pNP) in a sequential dephosphorylation by REMC offers an estimate of reaction kinetics and elucidates the underlying mechanism of improved phosphate selectivity in a multi-ionic environment. Furthermore, phosphate specificity, homogeneous binding capacity, reusability, and visual observation of REMC membrane saturation binding direct it's useful economic, industrial applications in aqueous phosphate contaminant removal, which could be beneficial for the active recovery of the aquatic ecosystem.


Subject(s)
Lanthanum , Water Pollutants, Chemical , Adsorption , Catalysis , Ecosystem , Hydrogen-Ion Concentration , Ion Exchange , Kinetics , Organophosphates , Phosphates
5.
Neuropharmacology ; 194: 108372, 2021 08 15.
Article in English | MEDLINE | ID: mdl-33157086

ABSTRACT

Epigenetic polycomb repressor complex-1 subunit BMI-1 plays a pivotal role in the process of gene repression to maintain the self-renewal and differentiation state of neurogenic tissues. Accumulating reports links lower expression of BMI-1 fails to regulate the repression of anti-oxidant response genes disrupt mitochondrial homeostasis underlying neurodegeneration. Interestingly, this negative relation between BMI-1 function and neurodegeneration is distinct but has not been generalized as a potential biomarker particularly in Parkinson's disease (PD). Hyperphosphorylated BMI-1 undergoes canonical polycomb E3 ligase function loss, thereby leads to reduce monoubiquitylation of histone 2A at lysine 119 (H2AK119ub) corroborates cellular accumulation of α-synuclein protein phosphorylated at serine 129 (pα-SYN (S129). In general, neuroprotectant suppressing pα-SYN (S129) level turns ineffective upon depletion of neuronal BMI-1. However, it has been observed that our neuroprotectant exposure suppresses the cellular pα-SYN (S129) and restore the the BMI-1 expression level in neuronal tissues. The pharmacological inhibition and activation of proteasomal machinery promote the cellular accumulation and degradation of neuronal pα-SYN (S129), respectively. Furthermore, our investigation reveals that accumulated pα-SYN (S129) are priorly complexed with BMI-1 undergoes ubiquitin-dependent proteasomal degradation and established as key pathway for therpeutic effect in PD. These findings linked the unestablished non-canonical role of BMI-1 in the clearance of pathological α-SYN and suspected to be a novel therapeutic target in PD.


Subject(s)
Parkinson Disease/metabolism , Polycomb Repressive Complex 1/metabolism , Ubiquitination/physiology , alpha-Synuclein/metabolism , Animals , Brain/metabolism , Cell Line, Tumor , Female , Humans , Melatonin/pharmacology , Mice , Mice, Inbred BALB C , Models, Animal , Neuroprotection , Phosphorylation , Polycomb-Group Proteins/metabolism , Proteasome Endopeptidase Complex/drug effects , Rats , Rotenone/pharmacology
6.
J Mater Chem B ; 8(37): 8658-8670, 2020 09 30.
Article in English | MEDLINE | ID: mdl-32844866

ABSTRACT

Acute myeloid leukemia (AML), which is common in the elderly population, accounts for poor long-term survival with a high possibility of relapse. The associated lack of currently developed therapeutics is directing the search for new therapeutic targets relating to AML. EZH2 (Enhancer of Zeste Homolog 2) is a histone methyltransferase member of the polycomb-group (PcG) family, and its significant overexpression in AML means it has emerged as a potential epigenetic target. Here, we propose the human serum albumin (HSA) nanoparticle based delivery of small interfering RNA (siRNA), which can target EZH2-expressing genes in AML. EZH2 specific siRNA loaded in a polyethyleneimine (PEI) conjugated HSA nanocarrier can overcome the systemic instability of siRNA and precisely target the AML cell population for increased EZH2 gene silencing. A stable nanosized complex (HSANPs-PEI@EZH2siRNA), achieved via the electrostatic interaction of PEI and EZH2 siRNA, shows increased systemic stability and hemocompatibility, and enhanced EZH2 gene silencing activity in vitro, compared to conventional transfection reagents. HSANPs-PEI@EZH2siRNA-treated AML cells showed downregulated EZH2, which is associated with a reduced level of Bmi-1 protein, and H3K27me3 and H2AK119ub modification. The ubiquitin-mediated proteasomal degradation pathway plays a critical role in the downregulation of associated proteins following HSANPs-PEI@EZH2siRNA exposure to AML cells. c-Myb is the AML-responsive transcription factor that directly binds on the EZH2 promoter and was downregulated in HSANPs-PEI@EZH2siRNA-treated AML cells. The systemic exposure to HSANPs-PEI@EZH2siRNA of AML engrafted immunodeficient nude mice displayed efficient EZH2 gene silencing and a reduced AML cell population in peripheral blood and bone marrow. The present study demonstrates a non-viral siRNA delivery system for epigenetic targeting based superior anti-leukemic therapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Drug Carriers/chemistry , Leukemia, Myeloid, Acute/drug therapy , Nanoparticles/chemistry , RNA, Small Interfering/therapeutic use , Animals , Down-Regulation , Drug Carriers/toxicity , Enhancer of Zeste Homolog 2 Protein/genetics , Female , Gene Knockdown Techniques , Gene Silencing , Humans , Leukemia, Myeloid, Acute/genetics , Mice, Inbred BALB C , Mice, Nude , Nanoparticles/toxicity , Polyethyleneimine/chemistry , Polyethyleneimine/toxicity , RNA, Small Interfering/genetics , Serum Albumin, Human/chemistry , Serum Albumin, Human/toxicity , Xenograft Model Antitumor Assays
7.
Blood ; 136(6): 749-754, 2020 08 06.
Article in English | MEDLINE | ID: mdl-32548640

ABSTRACT

Several studies demonstrate that hemolysis and free heme in circulation cause endothelial barrier dysfunction and are associated with severe pathological conditions such as acute respiratory distress syndrome, acute chest syndrome, and sepsis. However, the precise molecular mechanisms involved in the pathology of heme-induced barrier disruption remain to be elucidated. In this study, we investigated the role of free heme in the endothelial barrier integrity and mechanisms of heme-mediated intracellular signaling of human lung microvascular endothelial cells (HLMVECs). Heme, in a dose-dependent manner, induced a rapid drop in the endothelial barrier integrity of HLMVECs. An investigation into barrier proteins revealed that heme primarily affected the tight junction proteins zona occludens-1, claudin-1, and claudin-5, which were significantly reduced after heme exposure. The p38MAPK/HSP27 pathway, involved in the regulation of endothelial cytoskeleton remodeling, was also significantly altered after heme treatment, both in HLMVECs and mice. By using a knockout (KO) mouse for MKK3, a key regulator of the p38MAPK pathway, we showed that this KO effectively decreased heme-induced endothelial barrier dysfunction. Taken together, our results indicate that targeting the p38MAPK pathway may represent a crucial treatment strategy in alleviating hemolytic diseases.


Subject(s)
Capillary Permeability/drug effects , Endothelial Cells/drug effects , Heme/pharmacology , MAP Kinase Kinase 3/physiology , MAP Kinase Signaling System/drug effects , Animals , Antigens, CD/analysis , Cadherins/analysis , Capillary Permeability/physiology , Cells, Cultured , Claudins/analysis , Endothelial Cells/physiology , HSP27 Heat-Shock Proteins/physiology , Heat-Shock Proteins/physiology , Hemolysis , Humans , Lung/blood supply , MAP Kinase Kinase 3/deficiency , MAP Kinase Signaling System/physiology , Mice , Mice, Knockout , Microvessels/cytology , Molecular Chaperones/physiology , Tight Junctions/drug effects , Zonula Occludens-1 Protein/analysis , p38 Mitogen-Activated Protein Kinases
8.
Redox Biol ; 36: 101593, 2020 09.
Article in English | MEDLINE | ID: mdl-32554303

ABSTRACT

Transforming growth factor beta-1 (TGF-ß1) signaling is increased and mitochondrial function is decreased in multiple models of pulmonary hypertension (PH) including lambs with increased pulmonary blood flow (PBF) and pressure (Shunt). However, the potential link between TGF-ß1 and the loss of mitochondrial function has not been investigated and was the focus of our investigations. Our data indicate that exposure of pulmonary arterial endothelial cells (PAEC) to TGF-ß1 disrupted mitochondrial function as determined by enhanced mitochondrial ROS generation, decreased mitochondrial membrane potential, and disrupted mitochondrial bioenergetics. These events resulted in a decrease in cellular ATP levels, decreased hsp90/eNOS interactions and attenuated shear-mediated NO release. TGF-ß1 induced mitochondrial dysfunction was linked to a nitration-mediated activation of Akt1 and the subsequent mitochondrial translocation of endothelial NO synthase (eNOS) resulting in the nitration of carnitine acetyl transferase (CrAT) and the disruption of carnitine homeostasis. The increase in Akt1 nitration correlated with increased NADPH oxidase activity associated with increased levels of p47phox, p67phox, and Rac1. The increase in NADPH oxidase was associated with a decrease in peroxisome proliferator-activated receptor type gamma (PPARγ) and the PPARγ antagonist, GW9662, was able to mimic the disruptive effect of TGF-ß1 on mitochondrial bioenergetics. Together, our studies reveal for the first time, that TGF-ß1 can disrupt mitochondrial function through the disruption of cellular carnitine homeostasis and suggest that stimulating carinitine homeostasis may be an avenue to treat pulmonary vascular disease.


Subject(s)
Endothelial Cells , Hypertension, Pulmonary , Animals , Carnitine/pharmacology , Endothelial Cells/metabolism , Energy Metabolism , Homeostasis , Hypertension, Pulmonary/metabolism , Mitochondria/metabolism , Sheep , Transforming Growth Factor beta1/metabolism
9.
Biomater Sci ; 8(5): 1345-1363, 2020 Mar 07.
Article in English | MEDLINE | ID: mdl-31912833

ABSTRACT

Parkinson's disease (PD) is characterized by the loss of dopaminergic neurons in the substantia nigra and localized deposition of cytoplasmic fibrillary inclusions as Lewy bodies in the brain. The aberrant phosphorylation of α-synuclein at serine 129 is the key process on its early onset, which alters the cellular conformation to oligomers and insoluble aggregates, underpinning cellular oxidative stress and mitochondrial dysfunction, leading to devastating PD synucleinopathy. The multiple neuroprotective roles of dopamine and melatonin are often demonstrated separately; however, this approach suffers from low and short bioavailability and is associated with side-effects upon overdosing. Herein, highly pleiotropic melatonin-enriched polydopamine nanostructures were fabricated, which showed efficient brain tissue retention, sustainable and prolonged melatonin release, and prevented neuroblastoma cell death elicited by Parkinson's disease-associated and mitochondrial damaging stimuli. The synergistic neuroprotection re-established the mitochondrial membrane potential, reduced the generation of cellular reactive oxygen species (ROS), inhibited the activation of both the caspase-dependent and independent apoptotic pathways, and exhibited an anti-inflammatory effect. At the molecular level, it suppressed α-synuclein phosphorylation at Ser 129 and reduced the cellular deposition of high molecular weight oligomers. The therapeutic assessment on ex vivo organotypic brain slice culture, and in vivo experimental PD model confirmed the superior brain targeting, collective neuroprotection on dopaminergic neurons with reduced alpha-synuclein phosphorylation and deposition in the hippocampal and substantia nigra region of the brain. Thus, nature-inspired melatonin-enriched polydopamine nanostructures conferring collective neuroprotective effects attributes activation of anti-oxidative, anti-inflammatory, and anti-apoptotic pathways may be superior for application in a nanomedicine-based PD therapy.


Subject(s)
Indoles/pharmacology , Melatonin/pharmacology , Nanostructures/chemistry , Neuroprotective Agents/pharmacology , Parkinson Disease/drug therapy , Polymers/pharmacology , Apoptosis/drug effects , Cell Proliferation/drug effects , Humans , Indoles/chemistry , Melatonin/chemistry , Membrane Potential, Mitochondrial/drug effects , Neuroprotective Agents/chemistry , Parkinson Disease/pathology , Polymers/chemistry , Reactive Oxygen Species/analysis , Reactive Oxygen Species/metabolism , Tumor Cells, Cultured
10.
ACS Appl Mater Interfaces ; 12(5): 5658-5670, 2020 Feb 05.
Article in English | MEDLINE | ID: mdl-31986005

ABSTRACT

Alzheimer's disease (AD) is one of the common causes of dementia and mild cognitive impairments, which is progressively expanding among the elderly population worldwide. A short Amyloid-ß (Aß) peptide generated after amyloidogenic processing of amyloid precursor protein exist as intermolecular ß-sheet rich oligomeric, protofibriler, and fibrillar structures and believe to be toxic species which instigate neuronal pathobiology in the brain and deposits as senile plaque. Enormous efforts are being made to develop an effective anti-AD therapy that can target Aß processing, aggregation, and propagation and provide a synergistic neuroprotective effect. However, a nanodrug prepared from natural origin can confer a multimodal synergistic chemo/photothermal inhibition of Aß pathobiology is not yet demonstrated. In the present work, we report a dopamine-melatonin nanocomposite (DM-NC), which possesses a synergistic near-infrared (NIR) responsive photothermal and pharmacological modality. The noncovalent interaction-mediated self-assembly of melatonin and dopamine oxidative intermediates leads to the evolution of DM-NCs that can withstand variable pH and peroxide environment. NIR-activated melatonin release and photothermal effect collectively inhibit Aß nucleation, self-seeding, and propagation and can also disrupt the preformed Aß fibers examined using in vitro Aß aggregation and Aß-misfolding cyclic amplification assays. The DM-NCs display a higher biocompatibility to neuroblastoma cells, suppress the AD-associated generation of intracellular reactive oxygen species, and are devoid of any negative impact on the axonal growth process. In okadaic acid-induced neuroblastoma and ex vivo midbrain slice culture-based AD model, DM-NCs exposure suppresses the intracellular Aß production, aggregation, and accumulation. Therefore, this nature-derived nanocomposite demonstrates a multimodal NIR-responsive synergistic photothermal and pharmacological modality for effective AD therapy.


Subject(s)
Amyloid beta-Peptides/chemistry , Dopamine/chemistry , Melatonin/chemistry , Nanocomposites/radiation effects , Neurons/drug effects , Alzheimer Disease/metabolism , Amyloid/drug effects , Amyloid/metabolism , Amyloid beta-Peptides/metabolism , Animals , Brain/drug effects , Brain/metabolism , Brain Chemistry , Cell Line, Tumor , Dopamine/pharmacology , Female , Humans , Infrared Rays , Melatonin/pharmacology , Mice , Mice, Inbred BALB C , Nanocomposites/chemistry , Neuroblastoma , Neurons/metabolism , Reactive Oxygen Species/metabolism
11.
Mol Pharm ; 17(2): 604-621, 2020 02 03.
Article in English | MEDLINE | ID: mdl-31904978

ABSTRACT

Acute myeloid leukemia (AML) is a malignant disorder of hematopoietic progenitor cells with a poor prognosis of 26% of patients surviving 5 years after diagnosis. Poor bioavailability and solubility are significant factors limiting the efficacy of chemopreventive agents. In AML, the epigenetic regulator polycomb group of protein member EZH2 is highly expressed and is essential for the survival of leukemic cells. An EZH2-specific inhibitor, EPZ011989, encapsulated in human serum albumin nanoparticles (HSANPs) was synthesized for the first time via the desolvation method. The noncovalent interactions between EPZ011989 and HSANPs in nanocomposites facilitating the efficient loading and sustainable release of the drug showed enhanced cellular uptake and nuclear localization of EPZ011989-loaded HSANPs in human AML cell lines. The reduction of cell viability, colony formation inhibition, cell cycle arrest at the G2/M phase, and cell proliferation assay promoting apoptosis through the loss of mitochondrial homeostasis exerting antileukemic activity were evident. The real-time polymerase chain reaction (PCR) and western blot-based studies showed that the present nanoformulation reduces the level of PcG proteins, including EZH2, BMI-1, etc. This downregulation is associated with reduced H3K27me3 and H2AK119ub modifications conferring chromatin compaction. The immunoprecipitation study showed the physical interaction of EZH2 and c-Myb can be linked to the regulation of leukemogenesis. Further investigation revealed the mechanism of EZH2 and c-Myb downregulation via ubiquitination and proteasomal degradation pathway, confirmed by using proteasome inhibitor, suggesting the key role of proteasomal degradation machinery. Moreover, c-Myb interacted with the EZH2 promoter, which is evident by the chromatin immunoprecipitation assay and siRNA silencing. Furthermore, the formulation of EPZ011989 in HSANPs improved its biodistribution in vivo and showed excellent aqueous dispersibility and biocompatibility. In vivo studies further showed that EPZ011989-loaded HSANPs reduce the expression of CD11b+ and CD45+ markers in immunophenotyping from peripheral blood and bone marrow in engrafted nude mice. Targeted depletion of EZH2 alleviated the disease progression in nude mice and prolonged their survival. The findings provide valuable experimental evidence for the targeted epigenetic therapy of AML. The present results demonstrate an epigenetic regulation-based superior antileukemic therapy.


Subject(s)
Drug Delivery Systems/methods , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Enhancer of Zeste Homolog 2 Protein/genetics , Epigenesis, Genetic/drug effects , Leukemia, Myeloid, Acute/drug therapy , Nanoparticles/administration & dosage , Proteasome Endopeptidase Complex/metabolism , Proteolysis/drug effects , Proto-Oncogene Proteins c-myb/genetics , Animals , Cell Survival/drug effects , Drug Compounding/methods , Female , Gene Expression Regulation, Neoplastic , HEK293 Cells , HL-60 Cells , Humans , Leukemia, Myeloid, Acute/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Nanoparticles/chemistry , Serum Albumin, Human/chemistry , Tissue Distribution , Transfection , U937 Cells , Xenograft Model Antitumor Assays
12.
Nanomedicine ; 24: 102088, 2020 02.
Article in English | MEDLINE | ID: mdl-31476446

ABSTRACT

Posttranslational modification and agglomeration of α-synuclein (α-Syn), mitochondrial dysfunction, oxidative stress and loss of dopaminergic neurons are hallmark of Parkinson's disease (PD). This paper evaluates neuroprotection efficacy of nature inspired biocompatible polydopamine nanocarrier for metformin delivery (Met encapsulated PDANPs) by crossing blood brain barrier in in vitro, 3D and in vivo experimental PD models. The neuroprotective potential was arbitrated by downregulation of phospho-serine 129 (pSer129) α-Syn, with reduction in oxidative stress, prevention of apoptosis and anti-inflammatory activities. The neuroprotective mechanism proved novel interaction of epigenetic regulator EZH2 mediated ubiquitination and proteasomal degradation of aggregated pSer129 α-Syn. In summary, this study divulges the neuroprotective role of Met loaded PDANPs by reversing the neurochemical deficits by confirming an epigenetic mediated nanotherapeutic approach for the PD prevention.


Subject(s)
Enhancer of Zeste Homolog 2 Protein/metabolism , Metformin , Models, Biological , Nanostructures , Parkinson Disease/drug therapy , Proteasome Endopeptidase Complex/metabolism , Proteolysis/drug effects , alpha-Synuclein/metabolism , Cell Line, Tumor , Humans , Indoles/chemistry , Indoles/pharmacology , Metformin/chemistry , Metformin/pharmacology , Nanostructures/chemistry , Nanostructures/therapeutic use , Parkinson Disease/metabolism , Parkinson Disease/pathology , Polymers/chemistry , Polymers/pharmacology
13.
Sci Rep ; 9(1): 18347, 2019 12 04.
Article in English | MEDLINE | ID: mdl-31797958

ABSTRACT

We investigated biomarker CEACAM6, a highly abundant cell surface adhesion receptor that modulates the extracellular matrix (ECM) in pancreatic ductal adenocarcinoma (PDA). The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) RNA-Seq data from PDA patients were analyzed for CEACAM6 expression and evaluated for overall survival, association, enrichment and correlations. A CRISPR/Cas9 Knockout (KO) of CEACAM6 in PDA cell line for quantitative proteomics, mitochondrial bioenergetics and tumor growth in mice were conducted. We found CEACAM6 is over-expressed in primary and metastatic basal and classical PDA subtypes. Highest levels are in classical activated stroma subtype. CEACAM6 over-expression is universally a poor prognostic marker in KRAS mutant and wild type PDA. High CEACAM6 expression is associated with low cytolytic T-cell activity in both basal and classical PDA subtypes and correlates with low levels of T-REG markers. In HPAF-II cells knockout of CEACAM6 alters ECM-cell adhesion, catabolism, immune environment, transmembrane transport and autophagy. CEACAM6 loss increases mitochondrial basal and maximal respiratory capacity. HPAF-II CEACAM6-/- cells are growth suppressed by >65% vs. wild type in mice bearing tumors. CEACAM6, a key regulator affects several hallmarks of PDA including the fibrotic reaction, immune regulation, energy metabolism and is a novel therapeutic target in PDA.


Subject(s)
Adenocarcinoma/genetics , Antigens, CD/genetics , Carcinoma, Pancreatic Ductal/genetics , Cell Adhesion Molecules/genetics , T-Lymphocytes/metabolism , Adenocarcinoma/pathology , Adenocarcinoma/therapy , Animals , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/therapy , Cell Line, Tumor , Cell Proliferation/genetics , Energy Metabolism/genetics , GPI-Linked Proteins/genetics , Gene Expression Regulation, Neoplastic/genetics , Heterografts , Humans , Mice , Mitochondria/genetics , Molecular Targeted Therapy , Proto-Oncogene Proteins p21(ras)/genetics , T-Lymphocytes/pathology
14.
ACS Chem Neurosci ; 10(8): 3375-3385, 2019 08 21.
Article in English | MEDLINE | ID: mdl-31244053

ABSTRACT

Optogenetics have evolved as a promising tool to control the processes at a cellular level via photons. Specially, it confers a specific control over cellular function through real-time cytomodulation even in freely moving animals. Neuronal stimulation is prerequisite for deep tissue light penetration or insertion of optrode for light illumination to the neurons that have been proven to be compromised due to poor light penetration and invasiveness of the procedure, respectively. In this review, the application of nanotechnology is being elaborated by the use of metal nanoparticles (AuNPs), upconversion nanocrystals (UCNPs), and quantum dots (CdSe) for targeting particular organs or tissues, and their potential to emit a specific light on excitation to overcome the limitations associated with earlier methods has been elucidated. The optothermal and magnetothermal properties, photoluminescence, and higher photostability of nanomaterials are explored in context of therapeutic applicability of optogenetics. The nanostructure characteristics and specific ion channel targeting have shown promising therapeutic potential against neurodegenerative disorders (Alzheimer's, Parkinson's, Huntington's), epilepsy, and blindness. This review compiles mechanical and optical characteristics of nanomaterials that endow superior optogenetic therapeutic potentials to cure immedicable infirmities.


Subject(s)
Nanoparticles/therapeutic use , Nanotechnology/methods , Nanotechnology/trends , Optogenetics/methods , Optogenetics/trends , Animals , Humans , Neurology/methods , Neurology/trends
15.
Antioxid Redox Signal ; 30(15): 1775-1796, 2019 05 20.
Article in English | MEDLINE | ID: mdl-30187766

ABSTRACT

AIMS: Oxidant-induced endothelial injury plays a critical role in the pathogenesis of acute lung injury (ALI) and subsequent respiratory failure. Our previous studies revealed an endogenous antioxidant and protective pathway in lung endothelium mediated by heat shock protein 70 (Hsp70)-toll-like receptor 4 (TLR4) signaling. However, the downstream effector mechanisms remained unclear. Stanniocalcin 1 (STC1) has been reported to mediate antioxidant responses in tissues such as the lungs. However, regulators of STC1 expression as well as its physiological function in the lungs were unknown. We sought to elucidate the relationship between TLR4 and STC1 in hyperoxia-induced lung injury in vitro and in vivo and to define the functional role of STC1 expression in lung endothelium. RESULTS: We identified significantly decreased STC1 expression in TLR4 knockout mouse lungs and primary lung endothelium isolated from TLR4 knockout mice. Overexpression of STC1 was associated with endothelial cytoprotection, whereas decreased or insufficient expression was associated with increased oxidant-induced injury and death. An Hsp70-TLR4-nuclear factor kappa-light-chain-enhancer of activated B cells (NFκB) signal mediates STC1 induction in the lungs and endothelial cells. We also demonstrated a previously unrecognized role for mitochondrial-associated STC1, via TLR4, in maintaining normal glycolysis, mitochondrial bioenergetics, and mitochondrial calcium levels. INNOVATION: To date, a physiological role for STC1 in oxidant-induced ALI has not been identified. In addition, our studies show that STC1 is regulated by TLR4 and exerts lung and endothelial protection in response to sterile oxidant-induced lung injury. CONCLUSIONS: Our studies reveal a novel TLR4-STC1-mediated mitochondrial pathway that has homeostatic as well as oxidant-induced cytoprotective functions in lung endothelium.


Subject(s)
Acute Lung Injury/etiology , Acute Lung Injury/metabolism , Endothelial Cells/metabolism , Glycoproteins/genetics , Mitochondria/metabolism , Oxidants/metabolism , Toll-Like Receptor 4/metabolism , Acute Lung Injury/pathology , Animals , Antioxidants/metabolism , Calcium/metabolism , Disease Models, Animal , Fluorescent Antibody Technique , Gene Expression , Glycolysis , Glycoproteins/metabolism , HSP70 Heat-Shock Proteins/metabolism , Mice , Mice, Knockout , NF-kappa B/metabolism , Toll-Like Receptor 4/genetics
16.
Nanoscale Adv ; 1(6): 2188-2207, 2019 Jun 11.
Article in English | MEDLINE | ID: mdl-36131972

ABSTRACT

Combinatorial photodynamics and chemotherapy have drawn enormous attention as therapeutic modalities via precise stimuli-responsive drug delivery for glioblastoma, which can overcome the limitations associated with conventional therapies. Herein, we have prepared an indocyanine green tagged, genistein encapsulated casein nanoformulation (ICG-Gen@CasNPs) that exhibits the near infra-red region responsive controlled release of genistein and enhanced cellular uptake in the human glioblastoma monolayer and a three-dimensional raft culture model via the enhanced retention effect. ICG-Gen@CasNPs, with the integrated photosensitizer indocyanine green within the nanoformulation, triggered oxidative stress, activating the apoptosis cascade, promoting cell cycle arrest and damaging the mitochondrial membrane potential, collectively directing glioblastoma cell death. The suppression of the polycomb group of proteins in the glioblastoma upon ICG-Gen@CasNPs/NIR exposure revealed the involvement of the epigenetic repression complex machinery in the regulation. Furthermore, ICG-Gen@CasNPs/PDT/PTT directed ubiquitination and proteasomal degradation of EZH2 and BMI1 indicates the implication of the polycomb in conferring glioblastoma survival. The increased activation of the apoptotic pathways and the generation of cellular reactive oxygen species upon inhibiting the expression of EZH2 and BMI1 strengthen our observations. It is worth noting that ICG-Gen@CasNPs robustly accumulated in the brain after crossing the blood-brain barrier, which represents the eminent biocompatibility and means that the system is devoid of any nonspecific toxicity in vivo. Moreover, a superior anti-tumor effect was demonstrated on a three-dimensional glioma spheroid model. Thus, this combinatorial chemo/photodynamic therapy revealed that ICG-Gen@CasNPs mediated epigenetic regulation, which is a crucial molecular mechanism of GBM suppression.

17.
J Neurochem ; 148(2): 238-251, 2019 01.
Article in English | MEDLINE | ID: mdl-30308090

ABSTRACT

Brain-derived neurotrophic factor (BDNF) which is primarily associated with neuronal survivability, differentiation and synaptic plasticity has been reported to mediate neurodegeneration in hypoxia through its p75 Neurotrophin receptors (p75NTR). The molecular events promoting BDNF-mediated pro-death signalling in hypoxia, however, still remain an enigma. This study attempts towards deciphering the signalling cascades involved in alteration of BDNF isoforms and its cognate receptor subtypes leading to neurodegeneration in hypoxia. Adult Sprague-Dawley rats were exposed to global hypobaric hypoxia simulating an altitude of 7620 m at standard temperature and humidity. Chronic hypoxic exposure for 7 days resulted in higher expression of pro-BDNF and alteration in N-linked glycosylation in hippocampus along with increased expression of endoplasmic reticulum stress markers viz., glucose-regulated protein (Grp78), calnexin and changes in the endoplasmic reticulum morphology. Our findings reveal enriched expression of p75NTR in lipid rafts and higher expression of tyrosine receptor kinase ß (Trkß) in non-raft regions following hypoxic exposure. Further investigations on membrane properties revealed decline in membrane fluidity along with increased cholesterol oxidation resulting in reduced translocation of Trkß from non-raft to raft regions. Supplementation of vitamin E during hypoxic exposure on the other hand reduced cholesterol oxidation and increased translocation of Trkß from non-raft to raft regions and promoted neuronal survival. Hence, our findings suggest a novel mechanism of cholesterol oxidation-induced alteration in raft dynamics which is promotes p75 receptor-mediated death signalling in hippocampal neurons during chronic hypoxia.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Cholesterol/metabolism , Hippocampus/physiopathology , Hypoxia/physiopathology , Nerve Degeneration/physiopathology , Animals , Apoptosis/physiology , Hippocampus/metabolism , Male , Membrane Microdomains/metabolism , Membrane Microdomains/pathology , Nerve Tissue Proteins , Neurons/metabolism , Neurons/pathology , Oxidation-Reduction , Protein Isoforms/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Growth Factor , Receptors, Nerve Growth Factor/metabolism , Signal Transduction/physiology
18.
Respir Res ; 19(1): 69, 2018 04 23.
Article in English | MEDLINE | ID: mdl-29685148

ABSTRACT

BACKGROUND: Pulmonary arterial hypertension (PAH) is a fatal disease; however, the mechanisms directly involved in triggering and the progression of PAH are not clear. Based on previous studies that demonstrated a possible role of mitochondrial dysfunction in the pathogenesis of PAH, we investigated the effects of chronic inhibition of mitochondrial function in vivo in healthy rodents. METHODS: Right ventricle systolic pressure (RVSP) was measured in female rats at baseline and up to 24 days after inhibition of mitochondrial respiratory Complex III, induced by Antimycin A (AA, 0.35 mg/kg, given three times starting at baseline and then days 3 and 6 as a bolus injection into the right atrial chamber). RESULTS: Rodents exposed to AA demonstrated sustained increases in RVSP from days 6 through 24. AA-exposed rodents also possessed a progressive increase in RV end-diastolic pressure but not RV hypertrophy, which may be attributed to either early stages of PAH development or to reduced RV contractility due to inhibition of myocardial respiration. Protein nitration levels in plasma were positively correlated with PAH development in AA-treated rats. This finding was strongly supported by results obtained from PAH humans where plasma protein nitration levels were correlated with markers of PAH severity in female but not male PAH patients. Based on previously reported associations between increased nitric oxide production levels with female gender, we speculate that in females with PAH mitochondrial dysfunction may represent a more deleterious form, in part, due to an increased nitrosative stress development. Indeed, the histological analysis of AA treated rats revealed a strong perivascular edema, a marker of pulmonary endothelial damage. Finally, AA treatment was accompanied by a severe metabolic shift toward glycolysis, a hallmark of PAH pathology. CONCLUSIONS: Chronic mitochondrial dysfunction induces the combination of vascular damage and metabolic reprogramming that may be responsible for PAH development. This mechanism may be especially important in females, perhaps due to an increased NO production and nitrosative stress development.


Subject(s)
Electron Transport Complex III/antagonists & inhibitors , Electron Transport Complex III/metabolism , Glycolysis/physiology , Hypertension, Pulmonary/metabolism , Lung/metabolism , Mitochondria/metabolism , Vasoconstriction/physiology , Animals , Antimycin A/toxicity , Female , Glycolysis/drug effects , Humans , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/physiopathology , Lung/drug effects , Lung/physiopathology , Male , Mitochondria/drug effects , Monocrotaline/toxicity , Rats , Rats, Sprague-Dawley , Vasoconstriction/drug effects , Ventricular Dysfunction, Right/chemically induced , Ventricular Dysfunction, Right/metabolism , Ventricular Dysfunction, Right/physiopathology
19.
Am J Respir Cell Mol Biol ; 59(3): 334-345, 2018 09.
Article in English | MEDLINE | ID: mdl-29652520

ABSTRACT

Although hemolytic anemia-associated pulmonary hypertension (PH) and pulmonary arterial hypertension (PAH) are more common than the prevalence of idiopathic PAH alone, the role of hemolysis in the development of PAH is poorly characterized. We hypothesized that hemolysis independently contributes to PAH pathogenesis via endothelial barrier dysfunction with resulting perivascular edema and inflammation. Plasma samples from patients with and without PAH (both confirmed by right heart catheterization) were used to measure free hemoglobin (Hb) and its correlation with PAH severity. A sugen (50 mg/kg)/hypoxia (3 wk)/normoxia (2 wk) rat model was used to elucidate the role of free Hb/heme pathways in PAH. Human lung microvascular endothelial cells were used to study heme-mediated endothelial barrier effects. Our data indicate that patients with PAH have increased levels of free Hb in plasma that correlate with PAH severity. There is also a significant accumulation of free Hb and depletion of haptoglobin in the rat model. In rats, perivascular edema was observed at early time points concomitant with increased infiltration of inflammatory cells. Heme-induced endothelial permeability in human lung microvascular endothelial cells involved activation of the p38/HSP27 pathway. Indeed, the rat model also exhibited increased activation of p38/HSP27 during the initial phase of PH. Surprisingly, despite the increased levels of hemolysis and heme-mediated signaling, there was no heme oxygenase-1 activation. This can be explained by observed destabilization of HIF-1a during the first 2 weeks of PH regardless of hypoxic conditions. Our data suggest that hemolysis may play a significant role in PAH pathobiology.


Subject(s)
Hemoglobins/metabolism , Hemolysis/physiology , Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/pathology , Lung/blood supply , Adult , Aged , Animals , Disease Models, Animal , Female , Humans , Hypoxia/complications , Lung Diseases/pathology , Male , Middle Aged , Rats , Vascular Remodeling/physiology
20.
Nat Med ; 24(1): 39-49, 2018 01.
Article in English | MEDLINE | ID: mdl-29200204

ABSTRACT

Thyroid hormone (TH) is critical for the maintenance of cellular homeostasis during stress responses, but its role in lung fibrosis is unknown. Here we found that the activity and expression of iodothyronine deiodinase 2 (DIO2), an enzyme that activates TH, were higher in lungs from patients with idiopathic pulmonary fibrosis than in control individuals and were correlated with disease severity. We also found that Dio2-knockout mice exhibited enhanced bleomycin-induced lung fibrosis. Aerosolized TH delivery increased survival and resolved fibrosis in two models of pulmonary fibrosis in mice (intratracheal bleomycin and inducible TGF-ß1). Sobetirome, a TH mimetic, also blunted bleomycin-induced lung fibrosis. After bleomycin-induced injury, TH promoted mitochondrial biogenesis, improved mitochondrial bioenergetics and attenuated mitochondria-regulated apoptosis in alveolar epithelial cells both in vivo and in vitro. TH did not blunt fibrosis in Ppargc1a- or Pink1-knockout mice, suggesting dependence on these pathways. We conclude that the antifibrotic properties of TH are associated with protection of alveolar epithelial cells and restoration of mitochondrial function and that TH may thus represent a potential therapy for pulmonary fibrosis.


Subject(s)
Mitochondria/physiology , Pulmonary Fibrosis/prevention & control , Thyroid Hormones/physiology , Animals , Cells, Cultured , Epithelium/physiology , Female , Humans , Iodide Peroxidase/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Mimicry , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Protein Kinases/genetics , Pulmonary Fibrosis/physiopathology , Iodothyronine Deiodinase Type II
SELECTION OF CITATIONS
SEARCH DETAIL
...