Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Type of study
Publication year range
1.
Nanomedicine ; 48: 102644, 2023 02.
Article in English | MEDLINE | ID: mdl-36549555

ABSTRACT

Porous polymer microspheres are employed in biotherapeutics, tissue engineering, and regenerative medicine. Porosity dictates cargo carriage and release that are aligned with the polymer physicochemical properties. These include material tuning, biodegradation, and cargo encapsulation. How uniformity of pore size affects therapeutic delivery remains an area of active investigation. Herein, we characterize six branched aliphatic hydrocarbon-based porogen(s) produced to create pores in single and multilayered microspheres. The porogens are composed of biocompatible polycaprolactone, poly(lactic-co-glycolic acid), and polylactic acid polymers within porous multilayered microspheres. These serve as controlled effective drug and vaccine delivery platforms.


Subject(s)
Drug Delivery Systems , Polymers , Porosity , Microspheres , Polymers/chemistry , Hydrocarbons , Particle Size
2.
Acta Biomater ; 158: 493-509, 2023 03 01.
Article in English | MEDLINE | ID: mdl-36581007

ABSTRACT

Effective antigen delivery facilitates antiviral vaccine success defined by effective immune protective responses against viral exposures. To improve severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) antigen delivery, a controlled biodegradable, stable, biocompatible, and nontoxic polymeric microsphere system was developed for chemically inactivated viral proteins. SARS-CoV-2 proteins encapsulated in polymeric microspheres induced robust antiviral immunity. The viral antigen-loaded microsphere system can preclude the need for repeat administrations, highlighting its potential as an effective vaccine. STATEMENT OF SIGNIFICANCE: Successful SARS-CoV-2 vaccines were developed and quickly approved by the US Food and Drug Administration (FDA). However, each of the vaccines requires boosting as new variants arise. We posit that injectable biodegradable polymers represent a means for the sustained release of emerging viral antigens. The approach offers a means to reduce immunization frequency by predicting viral genomic variability. This strategy could lead to longer-lasting antiviral protective immunity. The current proof-of-concept multipolymer study for SARS-CoV-2 achieve these metrics.


Subject(s)
COVID-19 , Vaccines , Humans , SARS-CoV-2 , COVID-19 Vaccines , Microspheres , Antiviral Agents/pharmacology
3.
Clin Transl Med ; 12(7): e958, 2022 07.
Article in English | MEDLINE | ID: mdl-35802825

ABSTRACT

BACKGROUND: Dysregulation of innate and adaptive immunity heralds both the development and progression of Parkinson's disease (PD). Deficits in innate immunity in PD are defined by impairments in monocyte activation, function, and pro-inflammatory secretory factors. Each influences disease pathobiology. METHODS AND RESULTS: To define monocyte biomarkers associated with immune transformative therapy for PD, changes in gene and protein expression were evaluated before and during treatment with recombinant human granulocyte-macrophage colony-stimulating factor (GM-CSF, sargramostim, Leukine® ). Monocytes were recovered after leukapheresis and isolation by centrifugal elutriation, before and 2 and 6 months after initiation of treatment. Transcriptome and proteome biomarkers were scored against clinical motor functions. Pathway enrichments from single cell-RNA sequencing and proteomic analyses from sargramostim-treated PD patients demonstrate a neuroprotective signature, including, but not limited to, antioxidant, anti-inflammatory, and autophagy genes and proteins (LRRK2, HMOX1, TLR2, TLR8, RELA, ATG7, and GABARAPL2). CONCLUSIONS: This monocyte profile provides an "early" and unique biomarker strategy to track clinical immune-based interventions, but requiring validation in larger case studies.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor , Parkinson Disease , Biomarkers , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , Humans , Monocytes/metabolism , Parkinson Disease/drug therapy , Parkinson Disease/metabolism , Proteomics , Recombinant Proteins , Treatment Outcome
4.
EBioMedicine ; 73: 103678, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34774454

ABSTRACT

BACKGROUND: A barrier to HIV-1 cure rests in the persistence of proviral DNA in infected CD4+ leukocytes. The high HIV-1 mutation rate leads to viral diversity, immune evasion, and consequent antiretroviral drug resistance. While CRISPR-spCas9 can eliminate latent proviral DNA, its efficacy is limited by HIV strain diversity and precision target cell delivery. METHODS: A library of guide RNAs (gRNAs) designed to disrupt five HIV-1 exons (tat1-2/rev1-2/gp41) was constructed. The gRNAs were derived from a conseensus sequence of the transcriptional regulator tat from 4004 HIV-1 strains. Efficacy was affirmed by gRNA cell entry through transfection, electroporation, or by lentivirus or lipid nanoparticle (LNP) delivery. Treated cells were evaluated for viral excision by monitoring HIV-1 DNA, RNA, protein, and progeny virus levels. FINDINGS: Virus was reduced in all transmitted founder strains by 82 and 94% after CRISPR TatDE transfection or lentivirus treatments, respectively. No recorded off-target cleavages were detected. Electroporation of TatDE ribonucleoprotein and delivery of LNP TatDE gRNA and spCas9 mRNA to latently infected cells resulted in up to 100% viral excision. Protection against HIV-1-challenge or induction of virus during latent infection, in primary or transformed CD4+ T cells or monocytes was achieved. We propose that multi-exon gRNA TatDE disruption delivered by LNPs enables translation for animal and human testing. INTERPRETATION: These results provide "proof of concept' for CRISPR gRNA treatments for HIV-1 elimination. The absence of full-length viral DNA by LNP delivery paired with undetectable off-target affirms the importance of payload delivery for effective viral gene editing. FUNDING: The work was supported by the University of Nebraska Foundation, including donations from the Carol Swarts, M.D. Emerging Neuroscience Research Laboratory, the Margaret R. Larson Professorship, and individual donor support from the Frances and Louie Blumkin Foundation and from Harriet Singer. The research received support from National Institutes of Health grants T32 NS105594, 5R01MH121402, 1R01Al158160, R01 DA054535, PO1 DA028555, R01 NS126089, R01 NS36126, PO1 MH64570, P30 MH062261, and 2R01 NS034239.


Subject(s)
CRISPR-Cas Systems , Exons , Gene Editing , HIV Infections/therapy , HIV Infections/virology , HIV-1/genetics , Cell Line , Conserved Sequence , Fluorescent Antibody Technique , Gene Targeting , Genes, Reporter , Genetic Therapy , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Genome, Viral , Humans , Liposomes , Macrophages/metabolism , Macrophages/virology , Nanoparticles , Proviruses/genetics , RNA Interference , RNA, Messenger/administration & dosage , RNA, Messenger/genetics , rev Gene Products, Human Immunodeficiency Virus/genetics , tat Gene Products, Human Immunodeficiency Virus/genetics , RNA, Guide, CRISPR-Cas Systems
5.
Front Immunol ; 12: 741502, 2021.
Article in English | MEDLINE | ID: mdl-34671355

ABSTRACT

Host innate immune response follows severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, and it is the driver of the acute respiratory distress syndrome (ARDS) amongst other inflammatory end-organ morbidities. Such life-threatening coronavirus disease 2019 (COVID-19) is heralded by virus-induced activation of mononuclear phagocytes (MPs; monocytes, macrophages, and dendritic cells). MPs play substantial roles in aberrant immune secretory activities affecting profound systemic inflammation and end-organ malfunctions. All follow the presence of persistent viral components and virions without evidence of viral replication. To elucidate SARS-CoV-2-MP interactions we investigated transcriptomic and proteomic profiles of human monocyte-derived macrophages. While expression of the SARS-CoV-2 receptor, the angiotensin-converting enzyme 2, paralleled monocyte-macrophage differentiation, it failed to affect productive viral infection. In contrast, simple macrophage viral exposure led to robust pro-inflammatory cytokine and chemokine expression but attenuated type I interferon (IFN) activity. Both paralleled dysregulation of innate immune signaling pathways, specifically those linked to IFN. We conclude that the SARS-CoV-2-infected host mounts a robust innate immune response characterized by a pro-inflammatory storm heralding end-organ tissue damage.


Subject(s)
COVID-19/virology , Immunity, Innate , Macrophages/virology , SARS-CoV-2/pathogenicity , Angiotensin-Converting Enzyme 2/genetics , Angiotensin-Converting Enzyme 2/metabolism , COVID-19/immunology , COVID-19/metabolism , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , Gene Expression Profiling , Gene Regulatory Networks , Host-Pathogen Interactions , Humans , Immunity, Innate/genetics , Inflammation Mediators/metabolism , Macrophages/immunology , Macrophages/metabolism , Proteome , Proteomics , Receptors, Virus/genetics , Receptors, Virus/metabolism , SARS-CoV-2/immunology , Signal Transduction , Transcriptome
6.
bioRxiv ; 2021 Jul 15.
Article in English | MEDLINE | ID: mdl-34268510

ABSTRACT

Host innate immune response follows severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, and it is the driver of the acute respiratory distress syndrome (ARDS) amongst other inflammatory end-organ morbidities. Such life-threatening coronavirus disease 2019 (COVID-19) is heralded by virus-induced activation of mononuclear phagocytes (MPs; monocytes, macrophages, and dendritic cells). MPs play substantial roles in aberrant immune secretory activities affecting profound systemic inflammation and end organ malfunctions. All follow an abortive viral infection. To elucidate SARS-CoV-2-MP interactions we investigated transcriptomic and proteomic profiles of human monocyte-derived macrophages. While expression of the SARS-CoV-2 receptor, the angiotensin-converting enzyme 2, paralleled monocyte-macrophage differentiation it failed to affect productive viral infection. In contrast, simple macrophage viral exposure led to robust pro-inflammatory cytokine and chemokine expression but attenuated type I interferon (IFN) activity. Both paralleled dysregulation of innate immune signaling pathways specifically those linked to IFN. We conclude that the SARS-CoV-2-infected host mounts a robust innate immune response characterized by a pro-inflammatory storm heralding consequent end-organ tissue damage.

7.
FEMS Microbiol Rev ; 45(6)2021 11 23.
Article in English | MEDLINE | ID: mdl-34160586

ABSTRACT

Infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) can lead to coronavirus disease 2019 (COVID-19). Virus-specific immunity controls infection, transmission and disease severity. With respect to disease severity, a spectrum of clinical outcomes occur associated with age, genetics, comorbidities and immune responses in an infected person. Dysfunctions in innate and adaptive immunity commonly follow viral infection. These are heralded by altered innate mononuclear phagocyte differentiation, activation, intracellular killing and adaptive memory, effector, and regulatory T cell responses. All of such affect viral clearance and the progression of end-organ disease. Failures to produce effective controlled antiviral immunity leads to life-threatening end-organ disease that is typified by the acute respiratory distress syndrome. The most effective means to contain SARS-CoV-2 infection is by vaccination. While an arsenal of immunomodulators were developed for control of viral infection and subsequent COVID-19 disease, further research is required to enable therapeutic implementation.


Subject(s)
COVID-19 , Adaptive Immunity , Humans , Immunity, Innate , SARS-CoV-2
8.
J Neuroimmune Pharmacol ; 16(2): 270-288, 2021 06.
Article in English | MEDLINE | ID: mdl-33544324

ABSTRACT

Extracellular vesicles (EVs) are the common designation for ectosomes, microparticles and microvesicles serving dominant roles in intercellular communication. Both viable and dying cells release EVs to the extracellular environment for transfer of cell, immune and infectious materials. Defined morphologically as lipid bi-layered structures EVs show molecular, biochemical, distribution, and entry mechanisms similar to viruses within cells and tissues. In recent years their functional capacities have been harnessed to deliver biomolecules and drugs and immunological agents to specific cells and organs of interest or disease. Interest in EVs as putative vaccines or drug delivery vehicles are substantial. The vesicles have properties of receptors nanoassembly on their surface. EVs can interact with specific immunocytes that include antigen presenting cells (dendritic cells and other mononuclear phagocytes) to elicit immune responses or affect tissue and cellular homeostasis or disease. Due to potential advantages like biocompatibility, biodegradation and efficient immune activation, EVs have gained attraction for the development of treatment or a vaccine system against the severe acute respiratory syndrome coronavirus 2 (SARS CoV-2) infection. In this review efforts to use EVs to contain SARS CoV-2 and affect the current viral pandemic are discussed. An emphasis is made on mesenchymal stem cell derived EVs' as a vaccine candidate delivery system.


Subject(s)
COVID-19 Drug Treatment , Drug Delivery Systems/trends , Extracellular Vesicles , SARS-CoV-2/drug effects , Animals , Antiviral Agents/administration & dosage , Antiviral Agents/metabolism , COVID-19/immunology , COVID-19/metabolism , Drug Delivery Systems/methods , Extracellular Vesicles/immunology , Extracellular Vesicles/metabolism , Humans , Immunity, Cellular/drug effects , Immunity, Cellular/physiology , Immunologic Factors/administration & dosage , Immunologic Factors/metabolism , SARS-CoV-2/immunology , SARS-CoV-2/metabolism
9.
Adv Drug Deliv Rev ; 171: 215-239, 2021 04.
Article in English | MEDLINE | ID: mdl-33428995

ABSTRACT

The SARS-CoV-2 global pandemic has seen rapid spread, disease morbidities and death associated with substantive social, economic and societal impacts. Treatments rely on re-purposed antivirals and immune modulatory agents focusing on attenuating the acute respiratory distress syndrome. No curative therapies exist. Vaccines remain the best hope for disease control and the principal global effort to end the pandemic. Herein, we summarize those developments with a focus on the role played by nanocarrier delivery.


Subject(s)
COVID-19 Vaccines/administration & dosage , COVID-19/prevention & control , Drug Carriers/administration & dosage , Nanocapsules/administration & dosage , SARS-CoV-2/drug effects , Animals , COVID-19/immunology , COVID-19 Vaccines/immunology , Drug Delivery Systems/methods , Drug Delivery Systems/trends , Humans , SARS-CoV-2/immunology , Viral Vaccines/administration & dosage , Viral Vaccines/immunology
10.
Front Cell Dev Biol ; 8: 149, 2020.
Article in English | MEDLINE | ID: mdl-32226787

ABSTRACT

Stem cell therapy has garnered much attention and application in the past decades for the treatment of diseases and injuries. Mesenchymal stem cells (MSCs) are studied most extensively for their therapeutic roles, which appear to be derived from their paracrine activity. Recent studies suggest a critical therapeutic role for extracellular vesicles (EV) secreted by MSCs. EV are nano-sized membrane-bound vesicles that shuttle important biomolecules between cells to maintain physiological homeostasis. Studies show that EV from MSCs (MSC-EV) have regenerative and anti-inflammatory properties. The use of MSC-EV, as an alternative to MSCs, confers several advantages, such as higher safety profile, lower immunogenicity, and the ability to cross biological barriers, and avoids complications that arise from stem cell-induced ectopic tumor formation, entrapment in lung microvasculature, and immune rejection. These advantages and the growing body of evidence suggesting that MSC-EV display therapeutic roles contribute to the strong rationale for developing EV as an alternative therapeutic option. Despite the success in preclinical studies, use of MSC-EV in clinical settings will require careful consideration; specifically, several critical issues such as (i) production methods, (ii) quantification and characterization, (iii) pharmacokinetics, targeting and transfer to the target sites, and (iv) safety profile assessments need to be resolved. Keeping these issues in mind, the aim of this mini-review is to shed light on the challenges faced in MSC-EV research in translating successful preclinical studies to clinical platforms.

11.
J Neuroimmune Pharmacol ; 15(3): 443-458, 2020 09.
Article in English | MEDLINE | ID: mdl-31485884

ABSTRACT

Effective drug delivery to the CNS to achieve the desired therapeutic response is a significant challenge in the field of drug delivery. In central nervous system (CNS), blood brain barrier (BBB) restricts the desired therapeutic responses due to inefficient targeting, release kinetics, and failure to reach therapeutic concentrations in the brain. Therefore, most potentially beneficial diagnostic and therapeutic agents are not able to reach to the brain upon systemic administration. Despite the existence of many invasive techniques to promote drug deliveries across BBB, novel strategies of drug delivery system which can cross BBB effectively are required, otherwise translation of novel neurotherapeutics from bench to bedside will be difficult to achieve. In this review, we briefly outline the existing and emerging strategies for CNS drug deliveries with a focus on potential and challenges of using extracellular vesicles (EVs) in CNS drug delivery system. EVs are emerging as a promising tool for therapeutic delivery owing to its favorable intrinsic features of biocompatibility, stability, stealth capacity, ability to overcome natural barriers and inherent homing capability. EVs are nanovesicles that allow cell-cell communication. The EVs-cargo reflects the physiological as well as the pathophysiological state of a cell. EVs are shown to play a role in human immunodeficiency virus (HIV) infection and dissemination, which contributes to acquired immune deficiency syndrome (AIDS). In the context of HIV-1 infection, this review also outlines the role of EVs in dissemination, challenges faced in EVs research in HIV-1 co-morbid conditions and potential of nanotechnologies, especially EVs in Neuro-AIDS. Graphical Abstract EVs are used for the delivery of small molecule drugs, protein, and nucleic acid to the CNS as well as imaging molecules for in vivo tracking. For the purpose of delivery, EVs may or may not be subjected to membrane modification. The advantages of EVs, including its biocompatibility, low immunogenicity, and low toxicity profiles, can be exploited to potentially devise novel therapeutic delivery system for CNS drug targeting. This article outlines the challenges in potential EV-based therapeutic delivery.


Subject(s)
Central Nervous System Agents/administration & dosage , Central Nervous System , Drug Delivery Systems , Extracellular Vesicles/chemistry , Extracellular Vesicles/metabolism , Animals , Blood-Brain Barrier , Central Nervous System Agents/therapeutic use , Drug Carriers , Humans , Nanotechnology
12.
Cells ; 9(1)2019 12 19.
Article in English | MEDLINE | ID: mdl-31861723

ABSTRACT

Oxycodone (oxy) is a semi-synthetic opioid commonly used as a pain medication that is also a widely abused prescription drug. While very limited studies have examined the effect of in utero oxy (IUO) exposure on neurodevelopment, a significant gap in knowledge is the effect of IUO compared with postnatal oxy (PNO) exposure on synaptogenesis-a key process in the formation of synapses during brain development-in the exposed offspring. One relatively unexplored form of cell-cell communication associated with brain development in response to IUO and PNO exposure are extracellular vesicles (EVs). EVs are membrane-bound vesicles that serve as carriers of cargo, such as microRNAs (miRNAs). Using RNA-Seq analysis, we identified distinct brain-derived extracellular vesicle (BDEs) miRNA signatures associated with IUO and PNO exposure, including their gene targets, regulating key functional pathways associated with brain development to be more impacted in the IUO offspring. Further treatment of primary 14-day in vitro (DIV) neurons with IUO BDEs caused a significant reduction in spine density compared to treatment with BDEs from PNO and saline groups. In summary, our studies identified for the first time, key BDE miRNA signatures in IUO- and PNO-exposed offspring, which could impact their brain development as well as synaptic function.


Subject(s)
Brain/growth & development , Extracellular Vesicles/genetics , Gene Expression Profiling/methods , MicroRNAs/genetics , Oxycodone/adverse effects , Animals , Animals, Newborn , Brain/cytology , Brain/drug effects , Brain/metabolism , Cell Communication , Disease Models, Animal , Extracellular Vesicles/drug effects , Female , Gene Expression Regulation, Developmental/drug effects , Humans , Male , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Pregnancy , Primary Cell Culture , Rats , Sequence Analysis, RNA
13.
Stem Cells Int ; 2019: 8710180, 2019.
Article in English | MEDLINE | ID: mdl-31191687

ABSTRACT

The identification of several evolutionary young miRNAs, which arose in primates, raised several possibilities for the role of such miRNAs in human-specific disease processes. We previously have identified an evolutionary young miRNA, miR-1290, to be essential in neural stem cell proliferation and neuronal differentiation. Here, we show that miR-1290 is significantly downregulated during neuronal differentiation in reprogrammed induced pluripotent stem cell- (iPSC-) derived neurons obtained from idiopathic autism spectrum disorder (ASD) patients. Further, we identified that miR-1290 is actively released into extracellular vesicles. Supplementing ASD patient-derived neural stem cells (NSCs) with conditioned media from differentiated control-NSCs spiked with "artificial EVs" containing synthetic miR-1290 oligonucleotides significantly rescued differentiation deficits in ASD cell lines. Based on our earlier published study and the observations from the data presented here, we conclude that miR-1290 regulation could play a critical role during neuronal differentiation in early brain development.

14.
Pak J Pharm Sci ; 30(2): 341-346, 2017 03.
Article in English | MEDLINE | ID: mdl-28649054

ABSTRACT

Warfarin, an oral anticoagulant is one of the most widely prescribed drugs in modern medicine. Large inter-individuals variability due to age, gender, diet, concurrent drug interactions and variations in CYP2C9 and VKORC1 genes make the management of warfarin therapy challenging and yet no study has been conducted on the Bangladeshi population. The aim of the study was to identify the role of VKORC1 and CYP2C9 polymorphisms in Bangladeshi population in dose requirement of warfarin. We studied 87 heart valve replacement patients who were prescribed warfarin for minimum of 6 months with a target International normalized ratio of 2.0-3.5. Genotyping of VKORC1rs9923231 (-1639 G>A), CYP2C9*2 and CYP2C9*3 was performed by Polymerase Chain Reaction- Restriction Fragment Length Polymorphism. The frequencies of GG, AG and AA genotypes of VKORC1rs9923231 in the studied population were 87.4%, 8%, and 4.6% respectively whereas the frequencies of the CYP2C9*1/3 and CYP2C9*3/3 were 4.6% and 3.4% respectively. The CYP2C9*2 was not found in the studied population. The results of this study indicate that comparatively higher daily maintenance doses of warfarin were required to achieve the target INR for patients carrying both GG genotype of VKORC1rs9923231 and wild type variant of CYP2C9*3 whereas minimum dose were required for patient having AA genotype of VKORC1rs9923231 and *3/*3 variant of CYP2C9.


Subject(s)
Cytochrome P-450 CYP2C9/genetics , Vitamin K Epoxide Reductases/genetics , Warfarin/pharmacokinetics , Anticoagulants/pharmacokinetics , Bangladesh , Gene Frequency , Genotype , Humans , Polymorphism, Single Nucleotide/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...