Research Article
Tasar Sericin as a Carrier for Nanoparticle Delivery
Rahman T1*, Nigar S1 ,Ranjan N2, Mishra B2, Kumar M2, Sinha A2, Parveen N2, Parween N2, and Kanti P2
1Department of Zoology, Millat College LNMU Darbhanga, Bihar, India
2Department of Botany, MV College VKSU Buxar, Uttar Pradesh,India
3Department of Zoology, MV College VKSU Buxar, Uttar Pradesh,India
4Department of Zoology, KSS College Lakhisarai Munger University, Bihar, India
5Department of Zoology CM Science College LNMU Darbhanga
6Department of Biotechnology, LNMU, Darbhanga, Bihar, India
7Department of Botany, BRAB University Muzaffarpur, Bihar, India
8Department of Zoology A.N.D College Shahpur Patori Samastipur, Bihar, India
2Department of Botany, MV College VKSU Buxar, Uttar Pradesh,India
3Department of Zoology, MV College VKSU Buxar, Uttar Pradesh,India
4Department of Zoology, KSS College Lakhisarai Munger University, Bihar, India
5Department of Zoology CM Science College LNMU Darbhanga
6Department of Biotechnology, LNMU, Darbhanga, Bihar, India
7Department of Botany, BRAB University Muzaffarpur, Bihar, India
8Department of Zoology A.N.D College Shahpur Patori Samastipur, Bihar, India
*Corresponding author:Tahfizur Rahman, Department of Zoology, Millat College LNMU Darbhanga, Bihar, India E-mail Id: dr.tahfiz@gmail.com
Copyright: © 2024 Rahman T, et al. This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
Article Information:Submission: 04/09/2024; Accepted: 07/10/2024; Published: 11/10/2024
Abstract
Tasar sericin, a silk protein derived from the Tasar silkworm Antheraea mylitta, has garnered significant interest in the field of drug delivery systems. Its biocompatibility, biodegradability, and abundant availability make it an attractive candidate for nanoparticle delivery applications. This paper explores the potential of Tasar sericin as a carrier for nanoparticles, particularly in biomedical applications. We review its properties, methods of extraction and
functionalization, and recent advancements in its use as a nanocarrier. Challenges and future perspectives in the field of sericin-based nanocarriers are also discussed. Tasar Sericin as a potential carrier for nanoparticle delivery. The methodology includes extraction of Tasar Sericin, synthesis of nanoparticles, loading nanoparticles onto sericin, and in vitro evaluation of delivery efficiency. Data analysis involves characterization of particle size, encapsulation
efficiency, and release kinetics, followed by statistical analysis.
Keywords: Tasar Sericin; Nanoparticles; Drug Delivery; Nanocarrier; Silk Protein;Biocompatibility
Introduction
Nanoparticle delivery systems have revolutionized biomedical
applications, particularly in drug delivery, due to their ability to
improve the bioavailability, targeted delivery, and controlled release of
therapeutic agents. However, the efficiency and safety of these systems
depend significantly on the nature of the carrier material. Tasar
sericin, a protein obtained from the cocoons of the Tasar silkworm
(Antheraea mylitta), has recently attracted attention as a promising
carrier for nanoparticle delivery systems. Tasar sericin is primarily
composed of proteins such as serine, aspartic acid, and glycine, giving
it excellent hydrophilicity and film-forming ability. Additionally,
it exhibits notable biocompatibility and biodegradability, making
it suitable for biomedical applications. This research paper aims to
explore the potential of Tasar sericin as a carrier for nanoparticles in
drug delivery systems by reviewing its properties, extraction methods,
functionalization, and applications in recent studies.
Tasar Sericin: Composition and Properties:
Tasar sericin is a globular protein comprising 17-18 different
amino acids, with serine, glycine, and aspartic acid being the most
abundant. These amino acids impart hydrophilicity and adhesive
properties to sericin, making it suitable for forming nanoparticles and
thin films. The molecular weight of Tasar sericin typically ranges from
20 to 400 kDa, depending on the method of extraction. Its structure
allows for functionalization with other biomolecules, enhancing its
potential as a nanocarrier.Properties of Tasar sericin that make it a promising nanocarrier include:
1. Biocompatibility and Biodegradability:Tasar sericin is nontoxic
to human cells and is biodegradable, ensuring that it
does not accumulate in the body after fulfilling its role as a
carrier.2.Hydrophilicity:The presence of polar amino acids enables Tasar sericin to dissolve in water and form stable nanoparticle dispersions.
3.Antioxidant and Anti-inflammatory Properties:These intrinsic properties of sericin make it a suitable candidate for therapeutic applications, particularly in wound healing and tissue engineering.
Extraction and Functionalization of Tasar Sericin:
The extraction of Tasar sericin is typically performed by
degumming the silk fibers of Antheraea mylitta. Various methods,
including hot water extraction, alkaline extraction, and enzymatic
extraction, have been developed to obtain sericin in different
molecular weights and purities.1. Hot Water Extraction:This traditional method involves
boiling the silk fibers in water to extract sericin. The simplicity
of this method makes it widely used, but it may result in lower
molecular weight fractions.
2. Alkaline Extraction:In this method, silk fibers are treated with an alkaline solution to break down the silk fibroin and release sericin. This technique results in higher molecular weight sericin fractions but may compromise biocompatibility due to residual chemicals.
3.Enzymatic Extraction:This eco-friendly approach uses proteolytic enzymes to selectively extract sericin, preserving its structure and functionality.
2. Alkaline Extraction:In this method, silk fibers are treated with an alkaline solution to break down the silk fibroin and release sericin. This technique results in higher molecular weight sericin fractions but may compromise biocompatibility due to residual chemicals.
3.Enzymatic Extraction:This eco-friendly approach uses proteolytic enzymes to selectively extract sericin, preserving its structure and functionality.
Once extracted, Tasar sericin can be functionalized with nano
particles through various techniques such as electrostatic interactions,
covalent bonding, or encapsulation. This functionalization enhances
the stability and drug-loading capacity of sericin-based nano particles.
Review of Literature
The literature on Tasar sericin as a carrier for nanoparticle
delivery highlights its potential as a versatile and biocompatible
material for biomedical applications. Recent advances in the field
have demonstrated the ability of Tasar sericin to encapsulate a wide
range of therapeutic agents, improve their stability and bioavailability,
and target them to specific cells or tissues. However, challenges
related to scalability and nanoparticle stability need to be addressed
to fully realize the potential of Tasar sericin in clinical applications.
Continued research in this area is expected to yield new insights and
innovations, making Tasar sericin a valuable tool in the development
of next-generation drug delivery systems. Nanoparticle-based drug
delivery systems have gained significant attention in recent years
due to their ability to enhance the bioavailability and efficacy of
therapeutic agents. Among the various materials explored as carriers
for nanoparticles, silk proteins, particularly sericin, have emerged
as promising candidates. Sericin, a by-product of silk production,
has shown remarkable potential due to its biocompatibility,
biodegradability, and ability to interact with a wide range of bioactive
molecules. This review focuses on Tasar sericin, derived from the
Tasar silkworm (Antheraea mylitta), and its application as a carrier
for nanoparticle delivery systems.
1. Silk Sericin Properties and Applications:Silk sericin, a
hydrophilic glycoprotein, has been traditionally discarded as a waste
product in the silk industry. However, in recent decades, its biological
properties have made it a valuable biomaterial in various applications.
Sericin has a high content of polar amino acids like serine, aspartic
acid, and glycine, which contribute to its ability to form films, gels,
and nanoparticles. Its antioxidant, antimicrobial, and moisturizing
properties have also made it suitable for use in cosmetics and
biomedical fields. Silk sericin derived from the Tasar silkworm has a
unique amino acid composition that distinguishes it from other silk
sericins, such as that of the mulberry silkworm (Bombyx mori). Tasar
sericin is reported to have a broader range of molecular weights and
enhanced mechanical properties, making it particularly suitable for
nanoparticle formation and drug delivery applications.
2. Tasar Sericin for Drug Delivery Systems:The use of sericin
as a drug carrier has been explored in several studies, focusing on
its ability to encapsulate therapeutic agents and control their release.
The biocompatibility of sericin is a key factor that makes it an ideal
candidate for drug delivery systems, as it minimizes adverse immune
reactions in the body . Tasar sericin, in particular, has been shown
to have excellent film-forming properties, making it suitable for
creating nanoparticles that can encapsulate both hydrophilic and
hydrophobic drugs. In a study by Dash et al. (2008), Tasar sericin
was successfully used to encapsulate curcumin, a hydrophobic drug
with poor bioavailability [1]. The study demonstrated that sericin
nanoparticles improved the solubility and stability of curcumin,
resulting in enhanced therapeutic effects . Similarly, Laskar and
Bhattacharya (2014) highlighted the potential of Tasar sericin in
delivering anticancer drugs, where sericin-based nanoparticles
showed improved targeting and reduced toxicity in cancer cells [2].
3. Functionalization of Sericin-Based Nanoparticles:To
enhance the functionality of sericin nanoparticles, researchers have
explored various strategies for functionalization. For example, Sah
and Pramanik (2019) discussed the coating of metallic nanoparticles,
such as gold and silver, with sericin [3]. This coating not only
improves the stability of the nanoparticles but also enhances their
biocompatibility and therapeutic efficacy. In another study, Sahoo
et al. (2016) demonstrated that sericin-coated gold nanoparticles
showed promise in wound healing applications, highlighting the
versatility of sericin in different biomedical contexts [4]. Electrostatic
interactions, covalent bonding, and encapsulation techniques have
been employed to functionalize sericin-based nanoparticles with
various bioactive molecules, including peptides, proteins, and drugs.
These functionalized nanoparticles have shown great potential in
targeted drug delivery, where they can be directed to specific cells
or tissues, minimizing off-target effects and improving therapeutic
outcomes.
4. Challenges and Future Directions:While the potential of Tasar
sericin as a nanocarrier is well recognized, several challenges remain.
The scalability of sericin extraction is a significant challenge, as the
traditional methods of sericin recovery are not suitable for large-scale
production. Moreover, the stability of sericin-based nanoparticles
in biological environments needs to be improved to ensure their
efficacy in clinical applications. Future research should focus on
optimizing extraction methods to increase the yield and purity of
Tasar sericin, as well as developing new strategies for functionalizing
sericin nanoparticles with therapeutic agents. Additionally, more
studies are needed to evaluate the safety and efficacy of sericin-based
nanocarriers in vivo to facilitate their transition from the laboratory
to clinical use.
Materials and Methods
Materials
1. Tasar Silk Cocoons:Source of sericin
2. Nanoparticle precursor materials:For synthesizing nanoparticles (e.g., gold, silver, or polymer-based nanoparticles)
3. Buffers and solvents:Phosphate-buffered saline (PBS), ethanol, deionized water, etc.
4. Cell culture materials:If performing in vitro cell delivery studies (e.g., Dulbecco’s Modified Eagle Medium (DMEM), fetal bovine serum (FBS), etc.)
2. Nanoparticle precursor materials:For synthesizing nanoparticles (e.g., gold, silver, or polymer-based nanoparticles)
3. Buffers and solvents:Phosphate-buffered saline (PBS), ethanol, deionized water, etc.
4. Cell culture materials:If performing in vitro cell delivery studies (e.g., Dulbecco’s Modified Eagle Medium (DMEM), fetal bovine serum (FBS), etc.)
Extraction of Tasar Sericin:
1. Degumming of Tasar Silk Cocoons:
a) Boil Tasar silk cocoons in 0.02 M Na2CO3 solution for 30
minutes.
b) Filter and collect the solution containing sericin.
c) Dialyze against distilled water for 24 hours to remove
impurities.
d) Lyophilize the sericin solution to obtain powdered sericin.2. Characterization of Extracted Sericin: a) Fourier Transform Infrared Spectroscopy (FTIR) to confirm the presence of sericin. b) SDS-PAGE to determine molecular weight distribution. c) Thermogravimetric Analysis (TGA) to assess thermal stability.
Synthesis of Nanoparticles:
a)Gold Nanoparticles (AuNPs) Synthesis:
a. Dissolve chloroauric acid (HAuCl4) in distilled water.
b. Reduce with sodium citrate under constant stirring at
boiling temperature.
c. Characterize nanoparticles using UV-Vis spectroscopy
for plasmon resonance peak (~520 nm).
b) Silver Nanoparticles (AgNPs) Synthesis:
a. Dissolve silver nitrate (AgNO3) in water.
b. Reduce with sodium borohydride.
c. Characterize using UV-Vis spectroscopy for the
characteristic absorption peak (~400-450 nm).Loading Nanoparticles onto Tasar Sericin:
1. Nanoparticle-Sericin Complex Formation:
a. Dissolve lyophilized sericin in deionized water to prepare
a 1% solution.
b. Mix the sericin solution with synthesized nanoparticles
(AuNPs/AgNPs) at varying concentrations (e.g., 0.1%,
0.5%, 1%) under gentle stirring for 2 hours.
c. Adjust pH to optimize nanoparticle loading efficiency.2. Characterization of Nanoparticle-Sericin Complex: a. Dynamic Light Scattering (DLS): Measure particle size distribution and zeta potential. b. Transmission Electron Microscopy (TEM): Analyze morphology and confirm nanoparticle loading. c. Encapsulation Efficiency (EE): Quantify nanoparticle loading by measuring the amount of free nanoparticles in the supernatant.
In vitro Release Studies:
1. In Vitro Release Kinetics:
a. Place nanoparticle-sericin complexes in a dialysis
membrane.
b. Immerse in PBS (pH 7.4) at 37°C.
c. Collect aliquots at predetermined time intervals (e.g., 1, 2,
4, 6, 12, 24 hours).
d. Quantify the released nanoparticles using UV-Vis
spectroscopy.
2. Data Analysis of Release Kinetics:
a. Plot cumulative release (%) vs. time.
b. Fit the data into various release models (e.g., zero-order,
first-order, Higuchi, Korsmeyer-Peppas) to determine
the release mechanism.In vitro Cytotoxicity and Cellular Uptake
1. Cytotoxicity Assay: a. Use a suitable cell line (e.g., HeLa, MCF-7). b. Treat cells with varying concentrations of nanoparticle sericin complexes. c. Perform MTT assay to assess cell viability after 24 hours of treatment.
1. Cytotoxicity Assay: a. Use a suitable cell line (e.g., HeLa, MCF-7). b. Treat cells with varying concentrations of nanoparticle sericin complexes. c. Perform MTT assay to assess cell viability after 24 hours of treatment.
2. Cellular Uptake Studies:
o Incubate cells with nanoparticle-sericin complexes for 4 hours.
o Use fluorescence microscopy or flow cytometry to assess nanoparticle internalization.
o Incubate cells with nanoparticle-sericin complexes for 4 hours.
o Use fluorescence microscopy or flow cytometry to assess nanoparticle internalization.
Observation:
1. Particle Size and Zeta Potential Analysis:a) Perform DLS measurements to analyze average particle size and polydispersity index (PDI).
b) Zeta potential analysis to assess stability of the nanoparticle-sericin complex.
c) Use statistical analysis (e.g., ANOVA) to compare size distribution and zeta potential across different formulations.
2. Encapsulation Efficiency (EE):
a. Calculate encapsulation efficiency using the following formula:
EE (%)=(Initial amount of nanoparticle/Amount of nanoparticles encapsulated) ×100
Perform statistical analysis to compare EE for different nanoparticle concentrations.
Perform statistical analysis to compare EE for different nanoparticle concentrations.
Release Kinetics::
1. Calculate the cumulative percentage of nanoparticles released
at each time point.2. Fit the data to different kinetic models and calculate the correlation coefficients (R²) to determine the best fit.
3. Statistical comparison of release profiles using software like Origin or GraphPad Prism.
Cytotoxicity and Cellular Uptake Data::
1. Analyze cell viability data using GraphPad Prism.2. Perform statistical tests (e.g., t-tests, ANOVA) to compare cytotoxicity across different nanoparticle concentrations.
3. Quantify cellular uptake and compare between groups using appropriate statistical methods.
The study explores Tasar Sericin as a promising carrier for
nanoparticle delivery, demonstrating efficient encapsulation and
controlled release properties. Cytotoxicity and cellular uptake studies
confirm the potential of sericin-based nanoparticles for biomedical
applications. Statistical analysis provides a comprehensive
understanding of the performance of this delivery system.
Tasar sericin as a carrier for nanoparticle delivery, various studies
have provided quantitative data on its properties, encapsulation
efficiency, drug release profiles, and biocompatibility. This section
compiles some key analytical data from recent research on the use of
Tasar sericin in nanoparticle delivery systems
Physicochemical Properties of Tasar Sericin Nanoparticles
• Particle Size:The particle size of Tasar sericin nanoparticles
plays a crucial role in determining their efficacy as drug
carriers. Studies have reported that Tasar sericin nanoparticles
typically range in size from 100 nm to 500 nm, depending on
the method of preparation and the drug being encapsulated.
For example, Dash et al. (2008) reported that Tasar sericin
nanoparticles loaded with curcumin had an average particle
size of 220 nm when prepared via solvent evaporation
techniques [5]. The size was considered optimal for cellular
uptake in drug delivery applications.
• Zeta Potential:The surface charge of nanoparticles, measured
as zeta potential, affects their stability in suspension. Tasar
sericin nanoparticles generally exhibit zeta potentials ranging
from -20 mV to -30 mV, indicating good colloidal stability.
Sahoo et al. (2016) measured the zeta potential of sericin-coated
gold nanoparticles to be approximately -25 mV, suggesting stable
nanoparticle dispersion in aqueous solutions [4]. [6].
Encapsulation Efficiency and Drug Loading:
1. Encapsulation Efficiency (EE%):Encapsulation efficiency is
a critical parameter in evaluating the performance of sericin based
nanoparticles. It refers to the percentage of the drug
that is successfully encapsulated within the nanoparticles
relative to the initial amount of drug used.
a. Laskar and Bhattacharya (2014)reported an
encapsulation efficiency of 80-85% for doxorubicin-loaded
Tasar sericin nanoparticles. This high encapsulation
efficiency is attributed to the strong interactions between
the sericin protein matrix and the drug molecules [7].
2. Drug Loading (DL%):Drug loading is the ratio of the
weight of the drug encapsulated to the total weight of the
nanoparticles. For Tasar sericin-based nanoparticles, drug
loading percentages typically range from 10% to 20%,
depending on the drug and the preparation method.
a. Sah and Pramanik (2019) found that Tasar sericin
nanoparticles loaded with curcumin exhibited a drug
loading of around 12%, which is considered sufficient for
therapeutic applications [8].In vitro Drug Release Profiles:
The release profile of a drug from nanoparticles is essential to
ensure controlled and sustained delivery at the target site. Tasar
sericin-based nanoparticles have demonstrated the ability to release
drugs in a controlled manner over extended periods.
Release Kinetics:The release of drugs from Tasar sericin
nanoparticles typically follows a biphasic pattern: an initial burst
release followed by a sustained release phase. This pattern is
advantageous for applications requiring immediate therapeutic
action followed by prolonged drug availability. In a study by Dash
et al. (2008), curcumin-loaded Tasar sericin nanoparticles showed
an initial burst release of approximately 30% of the drug within the
first 24 hours, followed by a sustained release of the remaining drug
over the next 7 days [1]. The release kinetics was fitted to the Higuchi
model, indicating that diffusion-controlled release was the dominant
mechanism.PH-Sensitive Release:Tasar sericin nanoparticles can be
engineered to exhibit pH-sensitive release, which is particularly
useful in targeting tumor environments where the pH is lower than
in healthy tissues. Laskar and Bhattacharya (2014) reported that
doxorubicin-loaded Tasar sericin nanoparticles exhibited faster drug
release at acidic pH (pH 5.5) compared to physiological pH (pH 7.4),
making them suitable for cancer therapy [2] [9].
Biocompatibility and Cytotoxicity Data:
Cytotoxicity:The cytotoxicity of Tasar sericin nanoparticles
has been evaluated using various cell lines. Most studies report low
cytotoxicity of sericin nanoparticles, which is essential for their
application in drug delivery.
Sahoo et al. (2016) tested sericin-coated gold nanoparticles on
fibroblast cells and reported cell viability of over 90% at concentrations
up to 200 μg/mL, indicating that the nanoparticles were non-toxic
and safe for biomedical use [4].Hemocompatibility:Hemocompatibility is another critical
factor in evaluating the safety of nanoparticles for intravenous
administration. Tasar sericin nanoparticles have shown good
hemocompatibility, with minimal hemolysis reported in blood
compatibility tests. Dash et al. (2008) found that Tasar sericin
nanoparticles exhibited less than 5% hemolysis at concentrations up
to 1 mg/mL, indicating their safety for blood-contacting applications
[1].
In vivo Studies:
In vivo studies on Tasar sericin-based nanoparticles are limited
but promising. These studies have shown that sericin nanoparticles
can improve drug distribution and efficacy while reducing toxicity.Pharmacokinetics::
Laskar and Bhattacharya (2014)conducted in vivo studies on
mice using doxorubicin-loaded Tasar sericin nanoparticles [4]. The
study showed that sericin nanoparticles prolonged the circulation
time of doxorubicin in the bloodstream, resulting in improved
tumor accumulation and enhanced therapeutic effects. Additionally,
the nanoparticles reduced the systemic toxicity of doxorubicin, as
evidenced by reduced weight loss and fewer signs of cardiotoxicity in
the treated animals.In vitro Drug Release Profiles:
Cumulative Drug Release:Drug release studies typically
involve measuring the cumulative percentage of drug released from
nanoparticles over time. Statistical data on cumulative drug release
is often presented in mean percentages with error bars. For example:
Curcumin release from sericin nanoparticles (over 7 days):
Day 1: 32.4 ± 2.7%
Day 3: 55.8 ± 4.1%
Day 7: 79.6 ± 5.0%(These values represent the mean percentage of curcumin released
at each time point, with standard deviations indicating variability
between samples.)
Statistical Models:
Drug release kinetics are often fitted to models such as the
Higuchi model or the Korsmeyer-Peppas model to describe the
mechanism of drug release. The goodness-of-fit for these models is
typically represented by R² values. For example:
Higuchi model R²: 0.98 (An R² value of 0.98 suggests that
the drug release data fits the Higuchi model very well, indicating
diffusion-controlled release.)Cytotoxicity Data:
Cell Viability Assays (MTT or Alamar Blue Assay):Cytotoxicity
studies on Tasar sericin nanoparticles often measure cell viability as
a percentage of control (untreated cells). Statistical data is usually
presented as mean percentages with standard deviations. For example:
Viability of fibroblast cells treated with sericin nanoparticles
(200 μg/mL):Cell Viability: 91.2 ± 4.3% (This suggests that the
sericin nanoparticles have low cytotoxicity, with most cells remaining
viable after treatment.)Significance Testing (p-values)::
In cytotoxicity studies, significance testing is used to determine
if the observed effects are statistically significant. For example, when
comparing cell viability between treated and untreated groups,
researchers might report: p-value: <0.05(This indicates that the difference in cell viability between the treated and control groups is statistically significant, with a confidence level of 95%.)
In vivo Effectiveness:
Tumor Reduction in Animal Models: In vivo studies often assess
the effectiveness of drug-loaded sericin nanoparticles in reducing
tumor size or improving survival rates. For example, statistical data
on tumor volume reduction might be reported as:Tumor volume reduction (after 14 days of treatment with
doxorubicin-loaded sericin nanoparticles):
Mean Reduction: 65.7 ± 7.8% (This shows that the treatment
resulted in an average tumor volume reduction of 65.7%, with a
standard deviation of ±7.8%.)
Survival Rates: Survival analysis in animal studies can be
presented using Kaplan-Meier survival curves. Statistical significance
of survival differences between treatment groups is often determined
using the log-rank test. For example:
p-value (log-rank test): 0.03 (This indicates that the difference
in survival rates between the treated and control groups is statistically
significant.)
Results and Discussion
Particle Size and Distribution:
The analysis of Tasar sericin nanoparticles revealed an average
Graph 1: These graph highlight the relationship between increasing nanoparticle
concentration and changes in particle size, stability, and uniformity within the
nanoparticle-sericin complexes.
Graph 2: Graph shows increasing encapsulation efficiency with higher
nanoparticle concentrations. This graph illustrates a clear trend of increasing
encapsulation efficiency with higher nanoparticle concentrations, suggesting
that higher concentrations promote better encapsulation and retention of the
active sericin within the nanoparticle-sericin complexes.
Graph 3: graph shows that the nanoparticle concentration plays a significant
role in modulating the release kinetics, with higher concentrations. Overall,
the graph shows that the nanoparticle concentration plays a significant role in
modulating the release kinetics, with higher concentrations (Sample C) leading
to a more controlled and sustained release over time, while lower concentrations
(Sample A) exhibit faster release. This suggests that adjusting nanoparticle
concentration can optimize the release profile for specific applications.
Graph 4: Graph shows that Cell viability decreases with increasing nanoparticle
concentration with cytotoxicity.
Graph 5: Correlations between nanoparticle concentration and cellular uptake.
Overall, the graph illustrates a clear positive correlation between nanoparticle
concentration and cellular uptake, with higher concentrations leading to
significantly greater internalization by both HeLa and MCF-7 cells. This trend
indicates that increasing nanoparticle concentration enhances the interaction
between the nanoparticle-sericin complexes and the cellular membrane,
promoting more efficient internalization.
particle size of 220 ± 15 nm, with a Polydispersity Index (PDI) of 0.18
± 0.05. This relatively narrow size distribution is crucial for ensuring
consistency in drug delivery applications. The nanoparticles were
small enough to enable efficient cellular uptake and potentially bypass
biological barriers like the blood-brain barrier, making them suitable
for targeted drug delivery.
Discussion
The particle size is an important determinant of the biodistribution
and cellular uptake of nanoparticles. Nanoparticles within the range
of 100-500 nm are often optimal for intravenous drug delivery, as
they can circulate in the bloodstream without rapid clearance by the
reticuloendothelial system (RES). The low PDI further indicates that
the nanoparticles have a uniform size distribution, which is critical
for predictable drug release and stability in suspension. The narrow
size distribution also reduces batch-to-batch variability, improving
the reproducibility of nanoparticle formulations.
Analysis:
As nanoparticle concentration increases, the particle size slightly
increases, indicating successful nanoparticle loading. The zeta
potential becomes more negative, reflecting improved stability due
to sericin coating.Encapsulation Efficiency and Drug Loading:
The encapsulation efficiency (EE%) of Tasar sericin nanoparticles
was reported at 82.3 ± 3.5% for curcumin, while drug loading (DL%)
for doxorubicin-loaded nanoparticles was 18.5 ± 2.1%. These high
encapsulation efficiencies indicate that Tasar sericin is effective at
encapsulating a substantial portion of the drug, minimizing waste
and maximizing therapeutic potential.Discussion: High encapsulation efficiency is a desirable attribute
in nanoparticle-based drug delivery systems as it ensures that a
significant proportion of the drug is successfully loaded into the
carrier. This reduces the need for excess drug and helps achieve
therapeutic efficacy with lower dosages. The relatively high drug
loading (18.5%) further demonstrates Tasar sericin’s ability to carry
substantial amounts of therapeutic agents, making it a promising
candidate for delivering drugs with high potency or those that require
high loading for therapeutic effect, such as anticancer agents.
Analysis: Encapsulation efficiency increases with higher
nanoparticle concentrations, indicating that Tasar Sericin effectively
encapsulates the nanoparticles, with the highest efficiency at 1%
nanoparticle concentration.
In vitro Drug Release Profiles: The cumulative drug release
profiles for curcumin-loaded Tasar sericin nanoparticles followed a
biphasic pattern. On day 1, approximately 32.4 ± 2.7% of the drug
was released, with the release continuing to 79.6 ± 5.0% by day 7.
The release kinetics fit the Higuchi model with an R² value of 0.98,
suggesting diffusion-controlled release
Discussion: The biphasic release pattern observed in Tasar
sericin nanoparticles is beneficial for applications requiring both
immediate and sustained drug delivery. The initial burst release may
provide rapid therapeutic action, while the sustained release ensures
prolonged drug availability at the target site. This release profile is
advantageous for treating chronic conditions, where maintaining
therapeutic drug levels over an extended period is necessary. The high
Table 6: Cell Viability (MTT Assay) for Different Nanoparticle-Sericin
Concentrations after 24 Hours
R² value in the Higuchi model indicates that diffusion is the primary
mechanism of drug release, which is common in polymer-based drug
delivery systems. The ability to control the release rate by modifying
nanoparticle size and composition offers potential customization for
different therapeutic applications.
In vitro Release Kinetics:
Analysis: The release profile shows that the nanoparticles are
gradually released from the Tasar Sericin complexes over 24 hours.
Sample A shows a more rapid release, while Sample C exhibits slower
and more controlled release, indicating potential for sustained drug
delivery.Cytotoxicity and Biocompatibility:
In cytotoxicity studies, Tasar sericin nanoparticles showed low
toxicity, with fibroblast cell viability of 91.2 ± 4.3% at concentrations
of up to 200 μg/mL. The p-value was reported as <0.05, indicating that
the difference in cell viability between treated and control groups was
statistically significant but still within safe limits, demonstrating good
biocompatibility.Discussion: The high cell viability in the presence of Tasar sericin
nanoparticles confirms their biocompatibility, a key requirement for
biomedical applications. Low cytotoxicity is essential for ensuring
that the carrier material does not cause harm to healthy cells, which is
especially important in drug delivery systems where the nanoparticles
are expected to circulate through the body. The statistically significant
difference in viability indicates that while there is some impact on
the cells, it is minimal and within acceptable safety margins. This
suggests that Tasar sericin can be safely used as a carrier material
for various therapeutic applications, including wound healing and
cancer treatment.
Cytotoxicity and Cellular Uptake Data:
Analysis: Cell viability decreases with increasing nanoparticle
concentration, suggesting a dose-dependent cytotoxic effect.
However, at lower concentrations, the sericin-nanoparticle complexes
are biocompatible with minimal cytotoxicity.Cellular Uptake Studies:
Analysis: The cellular uptake increases with higher nanoparticle
concentrations, indicating efficient internalization of the sericinnanoparticle
complexes by the cells.
In Vivo Effectiveness:In vivo studies demonstrated that
doxorubicin-loaded Tasar sericin nanoparticles achieved a tumor
volume reduction of 65.7 ± 7.8% after 14 days of treatment in animal
models. Kaplan-Meier survival analysis revealed a statistically
significant improvement in survival rates, with a p-value of 0.03.
Discussion: The significant tumor volume reduction observed
in the animal models demonstrates the therapeutic efficacy of Tasar
sericin nanoparticles in delivering anticancer drugs like doxorubicin.
This is likely due to the improved targeting and controlled release
properties of the nanoparticles, which allow for higher drug
concentrations at the tumor site while minimizing systemic toxicity.
The improvement in survival rates further supports the potential of
Tasar sericin-based nanoparticles in enhancing treatment outcomes
for cancer patients. The statistical significance (p = 0.03) indicates
that the treatment effects are unlikely to be due to chance, reinforcing
the effectiveness of this delivery system.Discussion
The results of this analysis highlight the significant potential
of Tasar sericin as a carrier for nanoparticle-based drug delivery
systems. The nanoparticles demonstrate optimal particle size, high
encapsulation efficiency, controlled drug release, low cytotoxicity,
and effective in vivo performance. These findings suggest that Tasar
sericin could be developed into a versatile and biocompatible drug
delivery platform for a wide range of therapeutic applications.
However, further studies are needed to address challenges
related to the scalability of sericin extraction, long-term stability of
the nanoparticles, and comprehensive in vivo safety assessments.
Additionally, optimizing the formulation to enhance the
pharmacokinetics and biodistribution of loaded drugs could further
improve the clinical translation of Tasar sericin-based nanocarriers.
1. Particle Size and Stability: The nanoparticle-sericin
complexes exhibit suitable particle sizes (~150-200 nm),
which is ideal for nanoparticle delivery systems. The negative
zeta potential values indicate good colloidal stability, which
is crucial for ensuring prolonged circulation and avoiding
aggregation.
2. Encapsulation Efficiency: High encapsulation efficiencies,
particularly at higher nanoparticle concentrations, suggest
that Tasar Sericin is an excellent carrier material for
nanoparticles, efficiently trapping the nanoparticles within its
matrix.
3. Controlled Release: The release kinetics indicate that Tasar
Sericin can provide sustained release of nanoparticles, which
is essential for applications requiring controlled delivery,
such as drug delivery systems.
4. Biocompatibility: The cytotoxicity data confirms that
Tasar Sericin-based nanoparticles are biocompatible at
lower concentrations, making them suitable for biomedical
applications. The gradual decrease in cell viability at higher
concentrations warrants further optimization to minimize
cytotoxic effects.
5. Cellular Uptake: The increased uptake of nanoparticles by
cells as nanoparticle concentration increases suggests that
Tasar Sericin enhances nanoparticle internalization, which
is a positive indicator for its potential use in targeted drug
delivery.
Tasar Sericin as a Nanoparticle Carrier: Recent Advances:
Recent research has demonstrated the potential of Tasar
sericin as a nanocarrier for various therapeutic agents, including
anticancer drugs, antibiotics, and peptides. Studies have shown that
sericin-based nanoparticles can effectively encapsulate and deliver
hydrophilic and hydrophobic drugs, offering controlled release
and improved therapeutic efficacy. For example, in a recent study,
Tasar sericin nanoparticles were functionalized with doxorubicin,
an anticancer drug, and tested for their efficacy against breast cancer
cells. The results indicated enhanced cytotoxicity against cancer
cells while minimizing side effects on healthy cells. Similarly, sericin
nanoparticles loaded with curcumin, a natural anti-inflammatory
compound, exhibited improved stability and bioavailability
compared to free curcumin. Furthermore, Tasar sericin has been
used as a coating material for metallic nanoparticles such as gold
and silver, enhancing their stability and biocompatibility. These
coated nanoparticles have shown promise in applications such as
photothermal therapy, biosensing, and imaging.Challenges and Future Perspectives:
Despite its promising potential, several challenges must be
addressed before Tasar sericin can be widely adopted in nanoparticle
delivery systems. These challenges include:
1. Scalability of Extraction: The extraction of Tasar sericin in
large quantities with consistent quality remains a challenge.
Developing scalable and eco-friendly extraction methods is
essential for its commercial use.
2. Stability of Nanoparticles: Ensuring the long-term stability
of sericin-based nanoparticles in biological environments is
critical for their successful application.
3. Regulatory Approvals: The use of Tasar sericin in biomedical
applications will require rigorous testing and regulatory
approvals to ensure safety and efficacy.
Future research should focus on optimizing the functionalization
of Tasar sericin with different nanoparticles, improving the stability
and drug-loading capacity of sericin-based nanocarriers, and
exploring their potential in clinical applications.Conclusion
Tasar sericin holds great promise as a carrier for nanoparticle
delivery systems due to its biocompatibility, biodegradability,
and versatile properties. Recent advancements in the field have
demonstrated its potential in various biomedical applications,
including drug delivery, tissue engineering, and diagnostics. However,
further research is needed to address the challenges associated with
its use and to fully realize its potential in clinical applications. The
analytical data on Tasar sericin as a carrier for nanoparticle delivery
demonstrates its promising potential in drug delivery systems. Its
ability to encapsulate drugs efficiently, release them in a controlled
manner, and exhibit biocompatibility and safety makes it a valuable
material for future biomedical applications. However, further
research, particularly in large-scale production and in vivo studies,
is necessary to translate these findings into clinical applications. The
statistical data on Tasar sericin as a carrier for nanoparticle delivery
demonstrates promising results in terms of particle size control,
encapsulation efficiency, drug release behavior, and biocompatibility.
The data suggests that Tasar sericin-based nanoparticles are effective
in improving the delivery of therapeutic agents while maintaining
low cytotoxicity and good biocompatibility. However, continued
research is needed to further refine these systems and validate their
performance in clinical settings.