Network


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

Hotspot


Dive into the research topics where Ermei Mäkilä is active.

Publication


Featured researches published by Ermei Mäkilä.


ACS Nano | 2010

Biocompatibility of Thermally Hydrocarbonized Porous Silicon Nanoparticles and their Biodistribution in Rats

Luis M. Bimbo; Mirkka Sarparanta; Hélder A. Santos; Anu J. Airaksinen; Ermei Mäkilä; Timo Laaksonen; Leena Peltonen; Vesa-Pekka Lehto; Jouni Hirvonen; Jarno Salonen

Porous silicon (PSi) particles have been studied for the effects they elicit in Caco-2 and RAW 264.7 macrophage cells in terms of toxicity, oxidative stress, and inflammatory response. The most suitable particles were then functionalized with a novel (18)F label to assess their biodistribution after enteral and parenteral administration in a rat model. The results show that thermally hydrocarbonized porous silicon (THCPSi) nanoparticles did not induce any significant toxicity, oxidative stress, or inflammatory response in Caco-2 and RAW 264.7 macrophage cells. Fluorescently labeled nanoparticles were associated with the cells surface but were not extensively internalized. Biodistribution studies in rats using novel (18)F-labeled THCPSi nanoparticles demonstrated that the particles passed intact through the gastrointestinal tract after oral administration and were also not absorbed from a subcutaneous deposit. After intravenous administration, the particles were found mainly in the liver and spleen, indicating rapid removal from the circulation. Overall, these silicon-based nanosystems exhibit excellent in vivo stability, low cytotoxicity, and nonimmunogenic profiles, ideal for oral drug delivery purposes.


Biomaterials | 2011

Drug permeation across intestinal epithelial cells using porous silicon nanoparticles

Luis M. Bimbo; Ermei Mäkilä; Timo Laaksonen; Vesa-Pekka Lehto; Jarno Salonen; Jouni Hirvonen; Hélder A. Santos

Mesoporous silicon particles hold great potential in improving the solubility of otherwise poorly soluble drugs. To effectively translate this feature into the clinic, especially via oral or parenteral administration, a thorough understanding of the interactions of the micro- and nanosized material with the physiological environment during the delivery process is required. In the present study, the behaviour of thermally oxidized porous silicon particles of different sizes interacting with Caco-2 cells (both non-differentiated and polarized monolayers) was investigated in order to establish their fate in a model of intestinal epithelial cell barrier. Particle interactions and TNF-α were measured in RAW 264.7 macrophages, while cell viabilities, reactive oxygen species and nitric oxide levels, together with transmission electron microscope images of the polarized monolayers, were assessed with both the Caco-2 cells and RAW 264.7 macrophages. The results showed a concentration and size dependent influence on cell viability and ROS-, NO- and TNF-α levels. There was no evidence of the porous nanoparticles crossing the Caco-2 cell monolayers, yet increased permeation of the loaded poorly soluble drug, griseofulvin, was shown.


Acta Biomaterialia | 2010

In vitro cytotoxicity of porous silicon microparticles: Effect of the particle concentration, surface chemistry and size

Hélder A. Santos; Joakim Riikonen; Jarno Salonen; Ermei Mäkilä; Teemu Heikkilä; Timo Laaksonen; Leena Peltonen; Vesa-Pekka Lehto; Jouni Hirvonen

We report here the in vitro cytotoxicity of mesoporous silicon (PSi) microparticles on the Caco-2 cells as a function of particle size fractions (1.2-75 microm), particle concentration (0.2-4 mg ml(-1)) and incubation times (3, 11 and 24 h). The particle size (smaller PSi particles showed higher cytotoxicity) and the surface chemistry treatment of the PSi microparticles were considered to be the key factors regarding the toxicity aspects. These effects were significant after the 11 and 24 h exposure times, and were explained by cell-particle interactions involving mitochondrial disruption resulting from ATP depletion and reactive oxygen species production induced by the PSi surface. These events further induced an increase in cell apoptosis and consequent cell damage and cell death in a dose-dependent manner and as a function of the PSi particle size. These effects were, however, less pronounced with thermally oxidized PSi particles. Under the experimental conditions tested and at particle sizes >25 microm, the non-toxic threshold concentration for thermally hydrocarbonized and carbonized PSi particles was <2 mg ml(-1), and for thermally oxidized PSi microparticles was <4 mg ml(-1).


Molecular Pharmaceutics | 2012

Intravenous Delivery of Hydrophobin-Functionalized Porous Silicon Nanoparticles: Stability, Plasma Protein Adsorption and Biodistribution

Mirkka Sarparanta; Luis M. Bimbo; Jussi Rytkönen; Ermei Mäkilä; Timo Laaksonen; Päivi Laaksonen; Markus Nyman; Jarno Salonen; Markus B. Linder; Jouni Hirvonen; Hélder A. Santos; Anu J. Airaksinen

Rapid immune recognition and subsequent elimination from the circulation hampers the use of many nanomaterials as carriers to targeted drug delivery and controlled release in the intravenous route. Here, we report the effect of a functional self-assembled protein coating on the intravenous biodistribution of (18)F-labeled thermally hydrocarbonized porous silicon (THCPSi) nanoparticles in rats. (18)F-Radiolabeling enables the sensitive and easy quantification of nanoparticles in tissues using radiometric methods and allows imaging of the nanoparticle biodistribution with positron emission tomography. Coating with Trichoderma reesei HFBII altered the hydrophobicity of (18)F-THCPSi nanoparticles and resulted in a pronounced change in the degree of plasma protein adsorption to the nanoparticle surface in vitro. The HFBII-THCPSi nanoparticles were biocompatible in RAW 264.7 macrophages and HepG2 liver cells making their intravenous administration feasible. In vivo, the distribution of the nanoparticles between the liver and spleen, the major mononuclear phagocyte system organs in the body, was altered compared to that of uncoated (18)F-THCPSi. Identification of the adsorbed proteins revealed that certain opsonins and apolipoproteins are enriched in HFBII-functionalized nanoparticles, whereas the adsorption of abundant plasma components such as serum albumin and fibrinogen is decreased.


Journal of Controlled Release | 2013

Co-delivery of a hydrophobic small molecule and a hydrophilic peptide by porous silicon nanoparticles

Dongfei Liu; Luis M. Bimbo; Ermei Mäkilä; Francesca Villanova; Martti Kaasalainen; Bárbara Herranz-Blanco; Carla Caramella; Vesa-Pekka Lehto; Jarno Salonen; Karl-Heinz Herzig; Jouni Hirvonen; Hélder A. Santos

Nanoparticulate drug delivery systems offer remarkable opportunities for clinical treatment. However, there are several challenges when they are employed to deliver multiple cargos/payloads, particularly concerning the synchronous delivery of small molecular weight drugs and relatively larger peptides. Since porous silicon (PSi) nanoparticles (NPs) can easily contain high payloads of drugs with various properties, we evaluated their carrier potential in multi-drug delivery for co-loading of the hydrophobic drug indomethacin and the hydrophilic human peptide YY3-36 (PYY3-36). Sequential loading of these two drugs into the PSi NPs enhanced the drug release rate of each drug and also their amount permeated across Caco-2 and Caco-2/HT29 cell monolayers. Regardless of the loading approach used, dual or single, the drug permeation profiles were in good correlation with their drug release behaviour. Furthermore, the permeation studies indicated the critical role of the mucus intestinal layer and the paracellular resistance in the permeation of the therapeutic compounds across the intestinal wall. Loading with PYY3-36 also greatly improved the cytocompatibility of the PSi NPs. Conformational analysis indicated that the PYY3-36 could still display biological activity after release from the PSi NPs and permeation across the intestinal cell monolayers. These results are the first demonstration of the promising potential of PSi NPs for simultaneous multi-drug delivery of both hydrophobic and hydrophilic compounds.


Biomaterials | 2012

The mucoadhesive and gastroretentive properties of hydrophobin-coated porous silicon nanoparticle oral drug delivery systems

Mirkka Sarparanta; Luis M. Bimbo; Ermei Mäkilä; Jarno Salonen; Päivi Laaksonen; A.M. Kerttuli Helariutta; Markus B. Linder; Jouni Hirvonen; Timo Laaksonen; Hélder A. Santos; Anu J. Airaksinen

Impediments to intestinal absorption, such as poor solubility and instability in the variable conditions of the gastrointestinal (GI) tract plague many of the current drugs restricting their oral bioavailability. Particulate drug delivery systems hold great promise in solving these problems, but their effectiveness might be limited by their often rapid transit through the GI tract. Here we describe a bioadhesive oral drug delivery system based on thermally-hydrocarbonized porous silicon (THCPSi) functionalized with a self-assembled amphiphilic protein coating consisting of a class II hydrophobin (HFBII) from Trichoderma reesei. The HFBII-THCPSi nanoparticles were found to be non-cytotoxic and mucoadhesive in AGS cells, prompting their use in a biodistribution study in rats after oral administration. The passage of HFBII-THCPSi nanoparticles in the rat GI tract was significantly slower than that of uncoated THCPSi, and the nanoparticles were retained in stomach by gastric mucoadhesion up to 3 h after administration. Upon entry to the small intestine, the mucoadhesive properties were lost, resulting in the rapid transit of the nanoparticles through the remainder of the GI tract. The gastroretentive drug delivery system with a dual function presented here is a viable alternative for improving drug bioavailability in the oral route.


Journal of Pharmaceutical Sciences | 2011

Drug Delivery Formulations of Ordered and Nonordered Mesoporous Silica: Comparison of Three Drug Loading Methods

Tarja Limnell; Hélder A. Santos; Ermei Mäkilä; Teemu Heikkilä; Jarno Salonen; Dmitry Yu. Murzin; Narendra Kumar; Timo Laaksonen; Leena Peltonen; Jouni Hirvonen

A poorly soluble model drug, indomethacin (IMC), was loaded into two types of silica particles using three different loading methods. The loading efficiency and the extent/rate of drug release were evaluated. Widely used equipment in pharmaceutical laboratories, rotavapor and fluid bed, were used in the loading. The porous materials used were ordered mesoporous silica MCM-41 and nonordered silica gel Syloid 244 FP EU. The materials differ both in their pore properties and particle sizes. Tablets were successfully compressed from the IMC-loaded particles. Mechanical stability of the porous structures was studied with XRPD and nitrogen sorption after tableting and drug release was evaluated at pH 5.5 before and after tableting. The release of the poorly soluble IMC was faster from the Syloid than from the MCM-41, presumably due to the larger pore size and smaller particle size. Loading of IMC into the MCM-41 microparticles improved the drug dissolution, and blending the microparticles with pharmaceutical excipients improved the IMC release even further. The fast release was also maintained after tableting. Loading of IMC into the Syloid particles alone was sufficient to produce similar IMC release profiles, as in the case of MCM-41 with the excipients.


Small | 2014

Microfluidic Assembly of Monodisperse Multistage pH‐Responsive Polymer/Porous Silicon Composites for Precisely Controlled Multi‐Drug Delivery

Dongfei Liu; Hongbo Zhang; Bárbara Herranz-Blanco; Ermei Mäkilä; Vesa-Pekka Lehto; Jarno Salonen; Jouni Hirvonen; Hélder A. Santos

We report an advanced drug delivery platform for combination chemotherapy by concurrently incorporating two different drugs into microcompoistes with ratiometric control over the loading degree. Atorvastatin and celecoxib were selected as model drugs due to their different physicochemical properties and synergetic effect on colorectal cancer prevention and inhibition. To be effective in colorectal cancer prevention and inhibition, the produced microcomposite contained hypromellose acetate succinate, which is insoluble in acidic conditions but highly dissolving at neutral or alkaline pH conditions. Taking advantage of the large pore volume of porous silicon (PSi), atorvastatin was firstly loaded into the PSi matrix, and then encapsulated into the pH-responsive polymer microparticles containing celecoxib by microfluidics in order to obtain multi-drug loaded polymer/PSi microcomposites. The prepared microcomposites showed monodisperse size distribution, multistage pH-response, precise ratiometric controlled loading degree towards the simultaneously loaded drug molecules, and tailored release kinetics of the loaded cargos. This attractive microcomposite platform protects the payloads from being released at low pH-values, and enhances their release at higher pH-values, which can be further used for colon cancer prevention and treatment. Overall, the pH-responsive polymer/PSi-based microcomposite can be used as a universal platform for the delivery of different drug molecules for combination therapy.


Langmuir | 2012

Amine Modification of Thermally Carbonized Porous Silicon with Silane Coupling Chemistry

Ermei Mäkilä; Luis M. Bimbo; Martti Kaasalainen; Barbara Herranz; Anu J. Airaksinen; Markku Heinonen; Edwin Kukk; Jouni Hirvonen; Hélder A. Santos; Jarno Salonen

Thermally carbonized porous silicon (TCPSi) microparticles were chemically modified with organofunctional alkoxysilane molecules using a silanization process. Before the silane coupling, the TCPSi surface was activated by immersion in hydrofluoric acid (HF). Instead of regeneration of the silicon hydride species, the HF immersion of silicon carbide structure forms a silanol termination (Si-OH) on the surface required for silanization. Subsequent functionalization with 3-aminopropyltriethoxysilane provides the surface with an amine (-NH(2)) termination, while the SiC-type layer significantly stabilizes the functionalized structure both mechanically and chemically. The presence of terminal amine groups was verified with FTIR, XPS, CHN analysis, and electrophoretic mobility measurements. The overall effects of the silanization to the morphological properties of the initial TCPSi were analyzed and they were found to be very limited, making the treatment effects highly predictable. The maximum obtained number of amine groups on the surface was calculated to be 1.6 groups/nm(2), corresponding to 79% surface coverage. The availability of the amine groups for further biofunctionalization was confirmed by successful biotinylation. The isoelectric point (IEP) of amine-terminated TCPSi was measured to be at pH 7.7, as opposed to pH 2.6 for untreated TCPSi. The effects of the surface amine termination on the cell viability of Caco-2 and HT-29 cells and on the in vitro fenofibrate release profiles were also assessed. The results indicated that the surface modification did not alter the loading of the drug inside the pores and also retained the beneficial enhanced dissolution characteristics similar to TCPSi. Cellular viability studies also showed that the surface modification had only a limited effect on the biocompatibility of the PSi.


Advanced Materials | 2014

Fabrication of a Multifunctional Nano‐in‐micro Drug Delivery Platform by Microfluidic Templated Encapsulation of Porous Silicon in Polymer Matrix

Hongbo Zhang; Dongfei Liu; Mohammad-Ali Shahbazi; Ermei Mäkilä; Bárbara Herranz-Blanco; Jarno Salonen; Jouni Hirvonen; Hélder A. Santos

A multifunctional nano-in-micro drug delivery platform is developed by conjugating the porous silicon nanoparticles with mucoadhesive polymers and subsequent encapsulation into a pH-responsive polymer using microfluidics. The multistage platform shows monodisperse size distribution and pH-responsive payload release, and the released nanoparticles are mucoadhesive. Moreover, this platform is capable of simultaneously loading and releasing multidrugs with distinct properties.

Collaboration


Dive into the Ermei Mäkilä's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hongbo Zhang

Åbo Akademi University

View shared research outputs
Top Co-Authors

Avatar

Mohammad-Ali Shahbazi

Technical University of Denmark

View shared research outputs
Top Co-Authors

Avatar

Dongfei Liu

University of Helsinki

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Vesa-Pekka Lehto

University of Eastern Finland

View shared research outputs
Researchain Logo
Decentralizing Knowledge