Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Adv Mater ; 30(35): e1802878, 2018 Aug.
Article in English | MEDLINE | ID: mdl-30003620

ABSTRACT

A nanoparticle system for systemic delivery of therapeutics is described, which incorporates a means of tracking the fate of the nanocarrier and its residual drug payload in vivo by photoluminescence (PL). Porous silicon nanoparticles (PSiNPs) containing the proapoptotic antimicrobial peptide payload, D [KLAKLAK]2 , are monitored by measurement of the intrinsic PL intensity and the PL lifetime of the nanoparticles. The PL lifetime of the PSiNPs is on the order of microseconds, substantially longer than the nanosecond lifetimes typically exhibited by conventional fluorescent tags or by autofluorescence from cells and tissues; thus, emission from the nanoparticles is readily discerned in the time-resolved PL spectrum. It is found that the luminescence lifetime of the PSiNP host decreases as the nanoparticle dissolves in phosphate-buffered saline solution (37 °C), and this correlates with the extent of release of the peptide payload. The time-resolved PL measurement allows tracking of the in vivo fate of PSiNPs injected (via tail vein) into mice. Clearance of the nanoparticles through the liver, kidneys, and lungs of the animals is observed. The luminescence lifetime of the PSiNPs decreases with increasing residence time in the mice, providing a measure of half-life for degradation of the drug nanocarriers.


Subject(s)
Nanoparticles , Animals , Luminescence , Mice , Peptides , Porosity , Silicon
2.
Nat Biomed Eng ; 2(2): 95-103, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29955439

ABSTRACT

Bacterial resistance to antibiotics has made it necessary to resort to antibiotics that have considerable toxicities. Here, we show that the cyclic 9-amino acid peptide CARGGLKSC (CARG), identified via phage display on Staphylococcus aureus (S. aureus) bacteria and through in vivo screening in mice with S. aureus-induced lung infections, increases the antibacterial activity of CARG-conjugated vancomycin-loaded nanoparticles in S. aureus-infected tissues and reduces the needed overall systemic dose, minimizing side effects. CARG binds specifically to S. aureus bacteria but not Pseudomonas bacteria in vitro, selectively accumulates in S. aureus-infected lungs and skin of mice but not in non-infected tissue and Pseudomonas-infected tissue, and significantly enhances the accumulation of intravenously injected vancomycin-loaded porous silicon nanoparticles bearing the peptide in S. aureus-infected mouse lung tissue. The targeted nanoparticles more effectively suppress staphylococcal infections in vivo relative to equivalent doses of untargeted vancomycin nanoparticles or of free vancomycin. The therapeutic delivery of antibiotic-carrying nanoparticles bearing peptides targeting infected tissue may help combat difficult-to-treat infections.

3.
Nat Commun ; 9(1): 1070, 2018 03 09.
Article in English | MEDLINE | ID: mdl-29523838

ABSTRACT

The original version of the Supplementary Information associated with this Article inadvertently omitted Supplementary Table 1. The HTML has now been updated to include a corrected version of the Supplementary Information.

4.
Nat Commun ; 8(1): 1403, 2017 11 10.
Article in English | MEDLINE | ID: mdl-29123083

ABSTRACT

Cerebrovascular changes occur in Alzheimer's disease (AD). Using in vivo phage display, we searched for molecular markers of the neurovascular unit, including endothelial cells and astrocytes, in mouse models of AD. We identified a cyclic peptide, CDAGRKQKC (DAG), that accumulates in the hippocampus of hAPP-J20 mice at different ages. Intravenously injected DAG peptide homes to neurovascular unit endothelial cells and to reactive astrocytes in mouse models of AD. We identified connective tissue growth factor (CTGF), a matricellular protein that is highly expressed in the brain of individuals with AD and in mouse models, as the target of the DAG peptide. We also showed that exogenously delivered DAG homes to the brain in mouse models of glioblastoma, traumatic brain injury, and Parkinson's disease. DAG may potentially be used as a tool to enhance delivery of therapeutics and imaging agents to sites of vascular changes and astrogliosis in diseases associated with neuroinflammation.


Subject(s)
Alzheimer Disease/metabolism , Peptides, Cyclic/metabolism , Alzheimer Disease/etiology , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/metabolism , Animals , Astrocytes/metabolism , Astrocytes/pathology , Brain/blood supply , Brain/metabolism , Brain/pathology , Connective Tissue Growth Factor/metabolism , Disease Models, Animal , Hippocampus/blood supply , Hippocampus/metabolism , Hippocampus/pathology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Nude , Mice, Transgenic , Peptide Library , Peptides, Cyclic/chemistry , Protein Binding
5.
Chem Sci ; 8(3): 2387-2395, 2017 Mar 01.
Article in English | MEDLINE | ID: mdl-28451344

ABSTRACT

Despite the advances in the design of antibody-drug conjugates (ADCs), the search is still ongoing for novel approaches that lead to increased stability and homogeneity of the ADCs. We report, for the first time, an ADC platform technology using a platinum(ii)-based linker that can re-bridge the inter-chain cysteines in the antibody, post-reduction. The strong platinum-sulfur interaction improves the stability of the ADC when compared with a standard maleimide-linked ADC thereby reducing the linker-drug exchange with albumin significantly. Moreover, due to the precise conserved locations of cysteines, both homogeneity and site-specificity are simultaneously achieved. Additionally, we demonstrate that our ADCs exhibit increased anticancer efficacy in vitro and in vivo. The Pt-based ADCs can emerge as a simple and exciting proposition to address the limitations of the current ADC linker technologies.

6.
ACS Appl Mater Interfaces ; 8(44): 30449-30457, 2016 Nov 09.
Article in English | MEDLINE | ID: mdl-27754645

ABSTRACT

A theranostic nanoparticle with biochemically triggered antibacterial activity is demonstrated. Metallic silver is deposited onto porous silicon nanoparticles (pSiNPs) by galvanic displacement. When aqueous diaminesilver ([Ag(NH3)2]+) is used as a silver source, the pSiNPs template the crystalline silver as small (mean diameter 13 nm) and well-dispersed nanoparticles embedded within and on the larger (100 nm) pSiNPs. The silver nanoparticles (AgNPs) quench intrinsic photoluminescence (PL) from the porous silicon (pSi) matrix. When exposed to an aqueous oxidant, the AgNPs are preferentially etched, Ag+ is released into solution, and PL from the pSi carrier is recovered. The released Ag+ results in 90% killing of (Gram-negative) Pseudomonas aeruginosa and (Gram-positive) Staphylococcus aureus within 3 h. When conjugated with the TAT peptide (sequence RKKRRQRRR), the silver-deposited porous silicon (pSi-Ag) nanocomposite shows distinct targeting toward S. aureus bacteria in vitro. Intravenously injected TAT-conjugated pSi-Ag nanoparticles accumulate in the liver, spleen, and lungs of mice, and the in vivo release of Ag+ and recovery of PL from pSi are demonstrated by the subsequent intraperitoneal administration of a hexacyanoferrate solution. The released Ag+ leads to a significant bacterial count reduction in liver tissue relative to the control. The data demonstrate the feasibility of the targeted and triggered delivery of antibacterial Ag+ ion in vivo, using a self-reporting and nontoxic nanocarrier.


Subject(s)
Metal Nanoparticles , Animals , Anti-Bacterial Agents , Mice , Microbial Sensitivity Tests , Silicon , Silver , Staphylococcus aureus , Theranostic Nanomedicine
7.
Adv Mater ; 28(36): 7962-7969, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27383373

ABSTRACT

Calcium ions react with silicic acid released from dissolving porous silicon nanoparticles to create an insoluble calcium silicate shell. The calcium silicate shell traps and protects an siRNA payload, which can be delivered to neuronal tissues in vitro or in vivo. Gene delivery is enhanced by the action of targeting and cell-penetrating peptides attached to the calcium silicate shell.


Subject(s)
Brain Injuries/metabolism , Calcium Compounds/chemistry , Nanoparticles/chemistry , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/pharmacokinetics , Silicates/chemistry , Silicon/chemistry , Animals , Brain/metabolism , Brain/pathology , Brain Injuries/pathology , Cell Line, Tumor , Disease Models, Animal , Mice , Porosity , RNA, Small Interfering/metabolism
8.
Nat Commun ; 7: 11980, 2016 06 28.
Article in English | MEDLINE | ID: mdl-27351915

ABSTRACT

Traumatic brain injury (TBI) is a major health and socio-economic problem, but no pharmacological agent is currently approved for the treatment of acute TBI. Thus, there is a great need for advances in this field. Here, we describe a short peptide (sequence CAQK) identified by in vivo phage display screening in mice with acute brain injury. The CAQK peptide selectively binds to injured mouse and human brain, and systemically injected CAQK specifically homes to sites of brain injury in mouse models. The CAQK target is a proteoglycan complex upregulated in brain injuries. Coupling to CAQK increased injury site accumulation of systemically administered molecules ranging from a drug-sized molecule to nanoparticles. CAQK-coated nanoparticles containing silencing oligonucleotides provided the first evidence of gene silencing in injured brain parenchyma by systemically administered siRNA. These findings present an effective targeting strategy for the delivery of therapeutics in clinical management of acute brain injuries.


Subject(s)
Brain Injuries, Traumatic/therapy , Drug Delivery Systems , Peptides , Aged , Animals , Brain Injuries, Traumatic/diagnostic imaging , Extracellular Matrix/metabolism , Humans , Male , Mice , Middle Aged
SELECTION OF CITATIONS
SEARCH DETAIL
...