Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Pavimol Angsantikul is active.

Publication


Featured researches published by Pavimol Angsantikul.


Nature | 2015

Nanoparticle biointerfacing by platelet membrane cloaking

Che-Ming J. Hu; Ronnie H. Fang; Kuei-Chun Wang; Brian T. Luk; Soracha Thamphiwatana; Diana Dehaini; Phu Nguyen; Pavimol Angsantikul; Cindy Wen; Ashley V. Kroll; Cody W. Carpenter; Manikantan Ramesh; Vivian Qu; Sherrina Patel; Jie Zhu; William Shi; Florence M. Hofman; Thomas C. Chen; Weiwei Gao; Kang Zhang; Shu Chien; Liangfang Zhang

Development of functional nanoparticles can be encumbered by unanticipated material properties and biological events, which can affect nanoparticle effectiveness in complex, physiologically relevant systems. Despite the advances in bottom-up nanoengineering and surface chemistry, reductionist functionalization approaches remain inadequate in replicating the complex interfaces present in nature and cannot avoid exposure of foreign materials. Here we report on the preparation of polymeric nanoparticles enclosed in the plasma membrane of human platelets, which are a unique population of cellular fragments that adhere to a variety of disease-relevant substrates. The resulting nanoparticles possess a right-side-out unilamellar membrane coating functionalized with immunomodulatory and adhesion antigens associated with platelets. Compared to uncoated particles, the platelet membrane-cloaked nanoparticles have reduced cellular uptake by macrophage-like cells and lack particle-induced complement activation in autologous human plasma. The cloaked nanoparticles also display platelet-mimicking properties such as selective adhesion to damaged human and rodent vasculatures as well as enhanced binding to platelet-adhering pathogens. In an experimental rat model of coronary restenosis and a mouse model of systemic bacterial infection, docetaxel and vancomycin, respectively, show enhanced therapeutic efficacy when delivered by the platelet-mimetic nanoparticles. The multifaceted biointerfacing enabled by the platelet membrane cloaking method provides a new approach in developing functional nanoparticles for disease-targeted delivery.


Nano Letters | 2015

Modulating Antibacterial Immunity via Bacterial Membrane-Coated Nanoparticles

Weiwei Gao; Ronnie H. Fang; Soracha Thamphiwatana; Brian T. Luk; Jieming Li; Pavimol Angsantikul; Qiangzhe Zhang; Che-Ming J. Hu; Liangfang Zhang

Synthetic nanoparticles coated with cellular membranes have been increasingly explored to harness natural cell functions toward the development of novel therapeutic strategies. Herein, we report on a unique bacterial membrane-coated nanoparticle system as a new and exciting antibacterial vaccine. Using Escherichia coli as a model pathogen, we collect bacterial outer membrane vesicles (OMVs) and successfully coat them onto small gold nanoparticles (AuNPs) with a diameter of 30 nm. The resulting bacterial membrane-coated AuNPs (BM-AuNPs) show markedly enhanced stability in biological buffer solutions. When injected subcutaneously, the BM-AuNPs induce rapid activation and maturation of dendritic cells in the lymph nodes of the vaccinated mice. In addition, vaccination with BM-AuNPs generates antibody responses that are durable and of higher avidity than those elicited by OMVs only. The BM-AuNPs also induce an elevated production of interferon gamma (INFγ) and interleukin-17 (IL-17), but not interleukin-4 (IL-4), indicating its capability of generating strong Th1 and Th17 biased cell responses against the source bacteria. These observed results demonstrate that using natural bacterial membranes to coat synthetic nanoparticles holds great promise for designing effective antibacterial vaccines.


Wiley Interdisciplinary Reviews-nanomedicine and Nanobiotechnology | 2014

Nanoparticle approaches against bacterial infections.

Weiwei Gao; Soracha Thamphiwatana; Pavimol Angsantikul; Liangfang Zhang

Despite the wide success of antibiotics, the treatment of bacterial infections still faces significant challenges, particularly the emergence of antibiotic resistance. As a result, nanoparticle drug delivery platforms including liposomes, polymeric nanoparticles, dendrimers, and various inorganic nanoparticles have been increasingly exploited to enhance the therapeutic effectiveness of existing antibiotics. This review focuses on areas where nanoparticle approaches hold significant potential to advance the treatment of bacterial infections. These areas include targeted antibiotic delivery, environmentally responsive antibiotic delivery, combinatorial antibiotic delivery, nanoparticle-enabled antibacterial vaccination, and nanoparticle-based bacterial detection. In each area we highlight the innovative antimicrobial nanoparticle platforms and review their progress made against bacterial infections.


Advanced Materials | 2017

Erythrocyte–Platelet Hybrid Membrane Coating for Enhanced Nanoparticle Functionalization

Diana Dehaini; Xiaoli Wei; Ronnie H. Fang; Sarah Masson; Pavimol Angsantikul; Brian T. Luk; Yue Zhang; Man Ying; Yao Jiang; Ashley V. Kroll; Weiwei Gao; Liangfang Zhang

Cell-membrane-coated nanoparticles have recently been studied extensively for their biological compatibility, retention of cellular properties, and adaptability to a variety of therapeutic and imaging applications. This class of nanoparticles, which has been fabricated with a variety of cell membrane coatings, including those derived from red blood cells (RBCs), platelets, white blood cells, cancer cells, and bacteria, exhibit properties that are characteristic of the source cell. In this study, a new type of biological coating is created by fusing membrane material from two different cells, providing a facile method for further enhancing nanoparticle functionality. As a proof of concept, the development of dual-membrane-coated nanoparticles from the fused RBC membrane and platelet membrane is demonstrated. The resulting particles, termed RBC-platelet hybrid membrane-coated nanoparticles ([RBC-P]NPs), are thoroughly characterized, and it is shown that they carry properties of both source cells. Further, the [RBC-P]NP platform exhibits long circulation and suitability for further in vivo exploration. The reported strategy opens the door for the creation of biocompatible, custom-tailored biomimetic nanoparticles with varying hybrid functionalities, which may be used to overcome the limitations of current nanoparticle-based therapeutic and imaging platforms.


ACS Nano | 2016

Enteric Micromotor Can Selectively Position and Spontaneously Propel in the Gastrointestinal Tract

Jinxing Li; Soracha Thamphiwatana; Wenjuan Liu; Berta Esteban-Fernández de Ávila; Pavimol Angsantikul; Elodie Sandraz; Jianxing Wang; Tailin Xu; Fernando Soto; Valentin Ramez; Xiaolei Wang; Weiwei Gao; Liangfang Zhang; Joseph Wang

The gastrointestinal (GI) tract, which hosts hundreds of bacteria species, becomes the most exciting organ for the emerging microbiome research. Some of these GI microbes are hostile and cause a variety of diseases. These bacteria colonize in different segments of the GI tract dependent on the local physicochemical and biological factors. Therefore, selectively locating therapeutic or imaging agents to specific GI segments is of significant importance for studying gut microbiome and treating various GI-related diseases. Herein, we demonstrate an enteric micromotor system capable of precise positioning and controllable retention in desired segments of the GI tract. These motors, consisting of magnesium-based tubular micromotors coated with an enteric polymer layer, act as a robust nanobiotechnology tool for site-specific GI delivery. The micromotors can deliver payload to a particular location via dissolution of their enteric coating to activate their propulsion at the target site toward localized tissue penetration and retention.


Advanced Materials | 2015

Hydrogel Retaining Toxin‐Absorbing Nanosponges for Local Treatment of Methicillin‐Resistant Staphylococcus aureus Infection

Fei Wang; Weiwei Gao; Soracha Thamphiwatana; Brian T. Luk; Pavimol Angsantikul; Qiangzhe Zhang; Che-Ming J. Hu; Ronnie H. Fang; Jonathan A. Copp; Dissaya Pornpattananangkul; Weiyue Lu; Liangfang Zhang

Targeting virulence factors such as bacterial toxins represents an attractive antimicrobial approach with potential advantages of expanding the repertoire of bacterial targets, preserving the host endogenous microbiome, and lowering selective pressure for resistance development.[1, 2] Among various toxins, pore-forming toxins (PFTs) are the most common class of bacterial protein toxins and constitute important bacterial virulence factors.[3] These toxins disrupt cells by forming pores on cellular membranes and altering their permeability for bioactivity.[4] However, the majority of current toxin targeting strategies, such as antisera,[5] monoclonal antibodies,[6, 7] small-molecule inhibitors,[8, 9] and molecularly imprinted polymers,[10] relies primarily on structure-specific epitopic binding and custom synthesis is required to match specific toxins. As a result, the enormous diversity of PFTs presents a serious challenge to devise an effective detoxification platform against bacterial infections. To address this challenge, a unique red blood cell (RBC) membrane-coated nanoparticle system has been recently developed by wrapping intact RBC membrane onto polymeric nanoparticles (denoted ‘nanosponges’) for broad detoxification applications.[11, 12] The term ‘nanosponges’ is used to describe the unique capability of the RBC membrane-coated nanoparticles for non-specifically ‘soaking up’ a broad spectrum of PFTs. Different from existing detoxification strategies, the nanosponges target the membrane-disrupting mechanism common to PFTs; thereby offering an all-purpose toxin decoy strategy to absorb various types of PFTs regardless of their molecular structures.[12]


ACS Nano | 2015

Detoxification of Organophosphate Poisoning Using Nanoparticle Bioscavengers

Zhiqing Pang; Che-Ming J. Hu; Ronnie H. Fang; Brian T. Luk; Weiwei Gao; Fei Wang; Erdembileg Chuluun; Pavimol Angsantikul; Soracha Thamphiwatana; Weiyue Lu; Xinguo Jiang; Liangfang Zhang

Organophosphate poisoning is highly lethal as organophosphates, which are commonly found in insecticides and nerve agents, cause irreversible phosphorylation and inactivation of acetylcholinesterase (AChE), leading to neuromuscular disorders via accumulation of acetylcholine in the body. Direct interception of organophosphates in the systemic circulation thus provides a desirable strategy in treatment of the condition. Inspired by the presence of AChE on red blood cell (RBC) membranes, we explored a biomimetic nanoparticle consisting of a polymeric core surrounded by RBC membranes to serve as an anti-organophosphate agent. Through in vitro studies, we demonstrated that the biomimetic nanoparticles retain the enzymatic activity of membrane-bound AChE and are able to bind to a model organophosphate, dichlorvos, precluding its inhibitory effect on other enzymatic substrates. In a mouse model of organophosphate poisoning, the nanoparticles were shown to improve the AChE activity in the blood and markedly improved the survival of dichlorvos-challenged mice.


Nature Communications | 2017

Micromotor-enabled active drug delivery for in vivo treatment of stomach infection

Berta Esteban-Fernández de Ávila; Pavimol Angsantikul; Jinxing Li; Miguel Angel Lopez-Ramirez; Doris E. Ramírez-Herrera; Soracha Thamphiwatana; Chuanrui Chen; Jorge Delezuk; Richard Samakapiruk; Valentin Ramez; Liangfang Zhang; Joseph Wang

Advances in bioinspired design principles and nanomaterials have led to tremendous progress in autonomously moving synthetic nano/micromotors with diverse functionalities in different environments. However, a significant gap remains in moving nano/micromotors from test tubes to living organisms for treating diseases with high efficacy. Here we present the first, to our knowledge, in vivo therapeutic micromotors application for active drug delivery to treat gastric bacterial infection in a mouse model using clarithromycin as a model antibiotic and Helicobacter pylori infection as a model disease. The propulsion of drug-loaded magnesium micromotors in gastric media enables effective antibiotic delivery, leading to significant bacteria burden reduction in the mouse stomach compared with passive drug carriers, with no apparent toxicity. Moreover, while the drug-loaded micromotors reach similar therapeutic efficacy as the positive control of free drug plus proton pump inhibitor, the micromotors can function without proton pump inhibitors because of their built-in proton depletion function associated with their locomotion.Nano- and micromotors have been demonstrated in vitro for a range of applications. Here the authors demonstrate the in-vivo therapeutic use of micromotors to treat H. pylori infection.


ACS Nano | 2017

Nanomotor-Enabled pH-Responsive Intracellular Delivery of Caspase-3: Toward Rapid Cell Apoptosis

Berta Esteban-Fernández de Ávila; Doris E. Ramírez-Herrera; Susana Campuzano; Pavimol Angsantikul; Liangfang Zhang; Joseph Wang

Direct and efficient intracellular delivery of enzymes to cytosol holds tremendous therapeutic potential while remaining an unmet technical challenge. Herein, an ultrasound (US)-propelled nanomotor approach and a high-pH-responsive delivery strategy are reported to overcome this challenge using caspase-3 (CASP-3) as a model enzyme. Consisting of a gold nanowire (AuNW) motor with a pH-responsive polymer coating, in which the CASP-3 is loaded, the resulting nanomotor protects the enzyme from release and deactivation prior to reaching an intracellular environment. However, upon entering a cell and exposure to the higher intracellular pH, the polymer coating is dissolved, thereby directly releasing the active CASP-3 enzyme to the cytosol and causing rapid cell apoptosis. In vitro studies using gastric cancer cells as a model cell line demonstrate that such a motion-based active delivery approach leads to remarkably high apoptosis efficiency within a significantly shorter time and with a lower amount of CASP-3 compared to other control groups not involving US-propelled nanomotors. For instance, the reported nanomotor system can achieve 80% apoptosis of human gastric adenocarcinoma cells within only 5 min, which dramatically outperforms other CASP-3 delivery approaches. These results indicate that the US-propelled nanomotors may act as a powerful vehicle for cytosolic delivery of active therapeutic proteins, which would offer an attractive means to enhance the current landscape of intracellular protein delivery and therapy. While CASP-3 is selected as a model protein in this study, the same nanomotor approach can be readily applied to a variety of different therapeutic proteins.


Journal of Controlled Release | 2017

Erythrocyte membrane-coated nanogel for combinatorial antivirulence and responsive antimicrobial delivery against Staphylococcus aureus infection

Yue Zhang; Jianhua Zhang; Wansong Chen; Pavimol Angsantikul; Kevin Spiekermann; Ronnie H. Fang; Weiwei Gao; Liangfang Zhang

ABSTRACT We reported an erythrocyte membrane‐coated nanogel (RBC‐nanogel) system with combinatorial antivirulence and responsive antibiotic delivery for the treatment of methicillin‐resistant Staphylococcus aureus (MRSA) infection. RBC membrane was coated onto the nanogel via a membrane vesicle templated in situ gelation process, whereas the redox‐responsiveness was achieved by using a disulfide bond‐based crosslinker. We demonstrated that the RBC‐nanogels effectively neutralized MRSA‐associated toxins in extracellular environment and the toxin neutralization in turn promoted bacterial uptake by macrophages. In intracellular reducing environment, the RBC‐nanogels showed an accelerated drug release profile, which resulted in more effective bacterial inhibition. When added to the macrophages infected with intracellular MRSA bacteria, the RBC‐nanogels significantly inhibited bacterial growth compared to free antibiotics and non‐responsive nanogel counterparts. These results indicate the great potential of the RBC‐nanogel system as a new and effective antimicrobial agent against MRSA infection. Graphical abstract We synthesized redox‐responsive hydrogel nanoparticles cloaked with red blood cell membranes, which integrate antivirulence therapy with responsive antimicrobial delivery for effective treatment against Staphylococcus aureus infection. Figure. No Caption available.

Collaboration


Dive into the Pavimol Angsantikul's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Weiwei Gao

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ronnie H. Fang

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Joseph Wang

University of California

View shared research outputs
Top Co-Authors

Avatar

Brian T. Luk

University of California

View shared research outputs
Top Co-Authors

Avatar

Jinxing Li

University of California

View shared research outputs
Top Co-Authors

Avatar

Che-Ming J. Hu

University of California

View shared research outputs
Top Co-Authors

Avatar

Diana Dehaini

University of California

View shared research outputs
Researchain Logo
Decentralizing Knowledge