Skip to main content

Table 1 SER-based nano-formulations for biological applications

From: Sericin based nanoformulations: a comprehensive review on molecular mechanisms of interaction with organisms to biological applications

Nanomaterial

Active compound

Method of synthesis

NPs Properties

Main characteristics/advantages

References

Sericin-chitosan

Doxorubicin

Cross-linking method

MD: 231 ± 7.3 nm; ZP: − 6.25 ± 1.2; PDI: 0.067

pH-dependent release of doxorubicin and increased cellular uptake in tumor acid environment. Self-stabilization and increased colloidal stability after freeze-drying and resistance of plasma protein adsorption. In vivo assays showed significant anti-tumoral activity with reduced doxorubicin cardiotoxicity

[40]

Sericin- poly(ethylene glycol) (PEG) nanoparticles

-

Self-assembly

MD: 197.3 ± 46 nm

Spherical nanoparticles are formed by hydrophobic interaction between sericin and PEG

[78]

Sericin nanoparticles coated with poly-L-lysine

Plasmid encoding a green fluorescent protein

Desolvation method

MD: 244.00 ± 8.48 nm; ZP: -29.7 ± 2.54 MV; PDI: 0.35 ± 0.07

Plasmid DNA was strongly packed to the NPs surface, which leads to significant transfection of mouse fibroblast after 72 h. No toxicity was observed

[79]

Sericin-Poly(ethylcyanoacrylate) Nanospheres

Fenofibrate

Interfacial polymerization

MD: 175 ± 12 nm; ZP: − 32.8 ± 1.0 mV; PDI: 0.201

Increased ~ twofold the drug absorption in the gastrointestinal tract compared to a non-encapsulated drug, reducing the levels of total cholesterol (TC), triacylglycerols (TG), very low-density lipoproteins (VLDL), and low-density lipoproteins (LDL)

[80]

Sericin-genipin nanoparticles

Atorvastatin

Desolvation-genipin crosslinking method

DM: 166 ± 0.30 nm; ZP: − 38.28 mV; PDI: 0.225

Nanoparticles (10 mg.kg−1) enhanced the antihyperlipidemic activity in mouse models in comparison to a non-encapsulated drug at the same concentration. No cytotoxicity was observed to murine (J774) cells

[81]

Sericin-folate nanoparticles

Doxorrubicin (DOX)

Self-assembly and chemical crosslinking

DM: 53.8 ± 17.6 nm; ZP: -15.15 ± 1.51 mV

The release of DOX is ~ fivefold higher in an acid environment than in a neutral condition. NPs were target to folate receptors of cancer cells and displayed good hemo-compatibility

[7]

Sericin-chitosan nanoparticles

Doxorrubicin (DOX)

Physical and chemical crosslinking

DM: 231 ± 7.3 nm; ZP: − 6.25 ± 1.2 mV; PDI: 0.067

pH-dependent release of DOX and increased cellular uptake in tumor acid environment (pH 6)

[82]

Sericin-silver nanoparticles

-

Green reduction using sericin as reductant

DM: 4 – 20 nm

Significant inhibition of S. aureus growth at 25 mg.L−1 of nanoparticles. No cytotoxicity was observed to murine cells (3T3) under this concentration

[57]

Sericin nanoparticles

Resveratrol

Desolvation method

DM: 183.43 ± 7.23 nm; ZP: ~ -20 mV; PDI: 0.2

Significant inhibition of cancerous cell growth (human colorectal adenocarcinoma, Caco2) while no cytotoxicity was observed to normal skin fibroblasts (CRL-2522)

[18]

Sericin-gellan gum-rice bran albumin nanocomposites

Doxorubicin

Self-assembly

DM: 218 nm; ZP: -7.43 mV

IC50 of DOX for breast cancer cell line (MCF-7) was reduced 1.8-fold after encapsulation. The survival rate of cancer cells was reduced by 42% after treatment with nanocomposites

[83]

Sericin poly(γ-benzyl-L-glutamate) (PBLG) micelles

Doxorubicin

Self-assembly

DM: 111.1 ± 0.7 nm; ZP: -25.4 ± 0.76 mV; PDI: 0.17 ± 0.01

The drug release was accelerated in the lysosomes under acid pH. Micelles induced 1.5-fold higher apoptosis in the breast cancer cell line (MCF-7) concerning the non-encapsulated drug. Suppression of tumor growth in vivo was 70% higher in mice-model treated with micelles in comparison to control

[84]

Sericin-cholesterol-folate nano micelles

IR780

Self-assembly

DM: 140 nm

Cancerous cells (Human papillomavirus-related endocervical adenocarcinoma, BGC-823) containing folate receptors efficiently internalized the nanoparticles, which results in cell toxicity after laser irradiation at 808 nm

[85]

Sericin nanoparticles

Curcumin

Desolvation method

DM: 278.15 ± 53 nm; ZP: -23.0 ± 3.59 mV; PDI: 0.54 ± 0.09

The best nanoparticle physicochemical characteristic was achieved with 1 mg.mL−1 of sericin, which results in 84.7% of encapsulation entrapment

[86]

Sericin nanoparticles coated or not with poly-L-Lysine

-

Desolvation method

DM: 16 to 156 nm; ZP: -34 to + 36.2 mV; PDI: 0.8 to 1

NPs produced with sericin (30–50 kDa) positively charged showed the strongest antibacterial effects in S. aureus and E. coli strains by enhancing ROS production

[60]

Sponge nanocomposite of sericin-silver nanoparticles

-

Green reduction using sericin as reductant

DM: 111.1 to 175 nm

Both nanoparticles and sponge nanocomposites induced antibacterial activity against S. aureus and E. coli. Antibacterial activity of sponge nanocomposites was concentration-dependent, being the best results achieved at 100 ppm of AgNPs

[87]

  1. MD mean diameter, ZP Zeta potential, PDI polydispersity index