Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 + 2639 word(s) 2639 2021-03-02 08:48:36 |
2 format correct Meta information modification 2639 2021-03-12 05:16:39 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Rádis-Baptista, G. Cell-Penetrating Peptides. Encyclopedia. Available online: https://encyclopedia.pub/entry/7968 (accessed on 19 July 2024).
Rádis-Baptista G. Cell-Penetrating Peptides. Encyclopedia. Available at: https://encyclopedia.pub/entry/7968. Accessed July 19, 2024.
Rádis-Baptista, Gandhi. "Cell-Penetrating Peptides" Encyclopedia, https://encyclopedia.pub/entry/7968 (accessed July 19, 2024).
Rádis-Baptista, G. (2021, March 12). Cell-Penetrating Peptides. In Encyclopedia. https://encyclopedia.pub/entry/7968
Rádis-Baptista, Gandhi. "Cell-Penetrating Peptides." Encyclopedia. Web. 12 March, 2021.
Cell-Penetrating Peptides
Edit

Cell-penetrating peptides (CPPs) comprise a class of short polypeptides that possess the ability to selectively interact with the cytoplasmic membrane of certain cell types, translocate across plasma membranes and accumulate in the cell cytoplasm, organelles (e.g., the nucleus and mitochondria), and other subcellular compartments. CPPs are either of natural origin or de novo designed and synthesized from segments and patches of larger proteins or designed by algorithms. With such intrinsic characteristics, along with membrane permeation, translocation, and cellular uptake properties, CPPs can intracellularly convey diverse substances and nanomaterials, such as hydrophilic organic compounds and drugs, macromolecules (nucleic acids and proteins), nanoparticles (nanocrystals and polyplexes), metals and radionuclides, which can be covalently attached via CPP N- and C-terminals or through the preparation of CPP complexes. A cumulative number of studies on animal toxins, primarily isolated from the venom of arthropods and snakes, have revealed the cell-penetrating activities of venom peptides and toxins, which can be harnessed for application in biomedicine and pharmaceutical biotechnology.

cell-penetrating peptide venom peptide arachnid venom peptide insect venom peptide snake venom peptide cellular uptake peptide chemical modification peptide engineering peptide design peptide carrier

1. Introduction

It has been established that the more diverse a biome is, the more intricate is the chemical-ecological relationships among organisms sharing the same niche and richer is the diversity of molecules and pharmacological activities that these co-inhabiting organisms contain. Indeed, a great probability of success in the drug discovery process from nature is connected to maximal biodiversity and chemical diversity [1].

Animal venoms and skin secretions are rich biological materials that contain a blend of biologically and pharmacologically active components, used by venomous and poisonous organisms for defense, predation and territorial disputes. The mechanisms via which these organisms deliver toxic components are related to how toxins have evolved and exerted their ecological functions [2]. As a result of millions of years of evolution, the diversity of molecular structures and activities is immeasurable, with hidden functions and structures only recently disclosed by modern omics techniques associated with classical pharmacological studies [3]. Thus, the holistic molecular genetic analyses of venomous animals utilizing, for example, transcriptome in combination with proteome studies of venom glands, have revealed not only the diversity of a set of substances produced by a given organism that inhabits a particular biome but also the uniqueness of such molecular repertoire expressed as active components of biological materials, such as venoms [4][5][6][7][8].

Although marine and terrestrial biological reservoirs of active venom peptides appear crucial sources for drug discovery, examples of basic and applied research should be provided to attract the attention of the scientific community both in academia and industry. Fundamental research, development and innovation of venom components as diagnostics, therapeutics or both are not always an obvious issue for consideration for the general audience. Furthermore, molecular diversity is intrinsically related to biodiversity and pharmaceutical success in drug discovery from nature is dependent on the abundance and variety of biological resources, thus highlighting arguments for policies of environmental conservation and economic sustainability worldwide [9].

A particular class of molecules from animal venom that has been under constant focus comprises biologically active (bioactive) peptides. Owing to their selectivity for corresponding cell receptors and target specificity, as well as the relative structural stability in body fluids and amenability to be genetically and synthetically engineered, natural venom peptides are promising scaffolds for conversion into biopharmaceuticals (biotherapeutics), as exemplified by cysteine-stabilized and linear helical peptides [10][11][12]. In this review, I discussed natural peptides from animal venom and their derivatives possessing intrinsic properties such as interaction with biomembranes of target cells, access to the cytoplasm through membrane translocation and eventual accumulation in distinct organelles such as the nucleus and mitochondria. This class of special peptides, collectively called cell-penetrating peptides (CPPs), comprise full sequences, peptide segments or patches of large proteins and even encrypted subcellular localization signals, as well as rationally designed peptide chimeras, cyclic, stapled, dimeric, multivalent and self-assembled peptides and peptoid foldamers [13][14][15].

In the field of toxicology, membrane translocation and cell-penetration capabilities are well-recognized phenomena mainly associated with microbial and plant toxins, such as binary bacterial toxins (e.g., diphtheria toxin, Shiga toxin and cholera toxin) and plant ribosome inhibitor proteins (e.g., ricin and abrin), which bind to their respective receptors on the eukaryotic cytoplasmic membrane and enter cells by receptor-mediated endocytosis [16][17][18]. Less common but with a steeply increasing number of reports published over the years, animal toxins and venom peptides endowed with intrinsic properties of membrane permeation and cell internalization are currently being unraveled. These venom CPPs and derivatives, recapitulated herein, mechanistically penetrate cells by distinct pathways that involve receptor-dependent and/or receptor-independent endocytosis (non-disruptive peptides) or direct translocation through pore formation, followed by cellular uptake and intracellular compartmentalization (disruptive peptides). Hence, in this review, I collated current examples of cell-penetrating peptides derived from animal venoms and toxins, their basic research and advanced applications.

Cell-penetrating peptides (CPPs) consist of short sequences that range from few amino acids to less than 40 residues, which owing to their physicochemical and biological properties can cross lipid membranes of cells and intracellularly transport diverse types of molecular cargoes in the form of covalent conjugates or noncovalent complexes. The first CPP sequences were characterized and derived from the transactivator of transcription from HIV-1 (Tat protein), the antennapedia (Antp) homeodomain from Drosophila and VP22 from Herpes simplex virus type I, almost three decades ago. For instance, CPPs such as Tat48–60 (residues 48–60, GRKKRRQRRRPPQ) and penetratin (Antp43–58, RQIKIWFQNRRMKWKK), which are short-derived fragments from TAT protein and Drosophila Antp, respectively, were able to translocate across the plasma membrane of eukaryotic cells and set the stage for the development of a new class of non-viral vectors for the intracellular delivery of compounds and, therefore, converted into archetypal CPPs [19][20]. Since these initial discoveries, the field of CPPs has expanded into a prolific field of research. Databases of curated information about experimentally characterized CPPs and algorithms to predict CPPs have been created, along with the expansion of the field of CPPs [21][22][23][24]. It has become evident that CPPs are mostly linear sequences but cyclic sequences have also been detected and developed. They are usually composed of positively charged amino acids with or without distributed hydrophobic residues, conferring these peptide structures with a variable but a considerable level of amphipathic and cationicity and high affinity for negatively charged lipid membranes and their components, including phospholipids and proteoglycans. In particular, in the case of linear CPPs, studies investigating the relationship between CPP secondary structures and penetrability have demonstrated that cationic helical peptides possess superior cell permeation than amphipathic helical and amphipathic random peptides, as revealed through synthetic polyarginine CPPs stabilized with different proportion of α-aminoisobutyric acid [25]. Nevertheless, unstructured peptides in solution eventually form helices upon interaction with plasma membranes, promoting their efficient membrane insertion, translocation and cellular uptake [26]. Hydrophobic interaction followed by membrane insertion also plays a role in the cell penetration property demonstrated by some amphipathic CPPs [27]. Importantly, from the perspective of applications in biomedicine and pharmaceutical biotechnology, CPPs can simultaneously mediate the intracellular delivery of diverse molecular cargos, along with the membrane-crossing activity. These molecular cargoes include hydrophilic drugs, radionuclides, imaging agents (fluorescent dyes), biopolymers (nucleic acids, polypeptides), functionalized liposomes and nanoparticles (nanocrystals, light-sensitive and magnetic nanoparticles). Therefore, CPPs have attracted considerable attention not only for their application as vectors for drug delivery but also as agents for diagnostics and therapy (theranostics) in medicine [28][29][30][31].

Regarding their mechanistic of cell penetrability, although not conclusively elucidated, it has been shown that CPPs traverse hydrophobic membranes and enter into cells through distinct routes, relying on both membrane-disruptive and non-disruptive processes, this later comprising energy-dependent, receptor-mediated and receptor-independent endocytosis, as well as energy independent, direct translocation strategies. Hence, pathways for CPP entry into cells involve: (I) non-disruptive endocytic routes, such as clathrin-mediated endocytosis, caveolae-mediated uptake, clathrin/caveolae-independent endocytosis and micropinocytosis; (II) non-disruptive membrane, direct translocation by alternative routes, like inverted micelle formation and “carpet” model penetration; (III) direct translocation by membrane-disruptive routes, such as pore-formation (toroid and barrel-stave pores) and electroporation-like permeabilization. Moreover, based on experimental evidence with numerous known CPPs, more than a single pathway of membrane permeation and cellular internalization may be involved, depending on the CPP physicochemical features, effective concentrations (i.e., the peptide-to-cell ratio) and targeted cell types [19][20][32]. Although distinct pathways are involved in membrane translocation and internalization of several distinct CPPs investigated, the mode of membrane interaction, the efficacy of CPP-mediated cargo-dependent transport, subcellular compartmentalization and eventual adverse cytotoxic effects, depend not only on their own CPP structures but also on the position and nature of the molecular cargoes and the types of cell targets, as observed with CPPs that were conjugated with fluorescent dyes and polypeptides [32][33][34][35][36].

2. Cell-Penetrating Peptides from Animal Venoms and Toxins

To date, native venom peptides with cell-penetrating properties have been identified in a limited number of species of insects (bees and wasps), arachnids (spiders and scorpions), fish, amphibians and snakes (elapids and pit vipers). These CPPs have dual or multiple biological activities and are derived from the venoms or skin secretions, which are toxigenic, cytotoxic or antimicrobials. These venom CPPs include melittin from the honeybee, anoplin and mastoparans from wasps, latarcin and lycosin from spiders, chlorotoxin, maurocalcine and imperatoxin from scorpions, cardiotoxin, crotamine, crotalicidin and elapid cathelicidin-related antimicrobial peptides (CRAMPs) from snakes, pardaxin from fish skin and bombesin from frog skin (Table 1).

Table 1. Poisonous and venomous organisms that produce toxins with cell penetrability that originate cell-penetrating peptides

Scientific name

Common name

Toxin or toxin template

Obs.

 

Insects

 

 

Table 2, below

Apis mellifera

European honey bee

Melittin

 

Anoplius samariensis

Japanese solitary wasp

Anoplin

 

Vespula lewisii

Korean leather-jacket

Mastoparan

 

 

 

 

 

Arachnids

 

 

Table 3

Lachesana tarabevi

Central Asian spider

Latarcin-1

 

Lycosa singorensis

Wolf spider

Lycosin-1

 

Leiurus quinquestriatus

Israeli yellow scorpion

Chlorotoxin

 

Scorpio maurus palmatus

Tunisian scorpion

Maurocalcine

 

Pandinus imperator

African scorpion

Imperatoxin-A

 

Hadrurus gertschi

Mexican scorpion

Hadrucalcin

 

Urodacus manicatus

Australian Black Rock scorpion

Wasabi receptor toxin

 

 

 

 

 

Fish

 

 

 

Pardachirus sp

Mole sole fishes

Pardaxin

Table 4

 

 

 

 

Amphibian

 

 

Table 4

Bombina bombina

European fire-bellied toad

Bombesin

 

 

 

 

 

Snakes

 

 

Table 4

Crotalus durissus terrificus

South American rattlesnake

crotamine

 

 

 

crotalicidin

 

Naja sp

Cobras

Cardiotoxin

 

 

The importance of this class of bioactive peptides relies on the fact that venom peptides and toxins, as well as related sequences, have evolved for millions of years of natural history and can be considered crucial improvements in peptide design that efficiently performs their functions. Importantly, native animal venom peptides and toxins with cell-penetrating activity have been characterized and CPPs derived from them conceived through a variety of structural modifications of their native sequences. These structural modifications of cell-penetrating venom peptides include peptide downsizing, amino acid residue replacement, structural stabilization and production of chimeras, which mostly reduced peptide cytotoxicity, maintained the target selectivity and enhanced the cellular penetrability and the capacity of intracellular cargo transport and delivery.

Tables 2 to 4 summarize examples of known venom peptides, toxins and derivatives with cell-penetrating properties, their overall structures, mode of cell entry, and conveyed compounds.

Table 2. Examples of peptides from insect (honey bee and wasp) venom and derivatives with cell-penetrating property.

Peptide

Sequence a

Size

Major Structural Features b

Mechanism(s) of Cell Penetration c

Cargo Delivery d

Ref.

Honey bee

 

 

 

 

 

 

Mellitin (MEL)

GIGAVLKVLTTGLPALISWIKRKRQQ-NH2

26

Amphipathic α-helix

DT, PF

FD

[49]

Tat/CM18 hybrid

 

 

Chimera

DT, MD, ND

CI

[52]

MEL-d(KLAKLAK)2

GIGAVLKVLTTGLPALISWIKRKRQQGGGGS-d[KLAKLAKKLAKLAK]

45

Chimera

DT (?)

AP

[53]

p5RHH

VLTTGLPALISWIRRRHRRHC

21

C-terminal fragment

Endocytosis (?)

DNA polyplexes

[54,55]

p5RWR

VLTTGLPALISWIKRKRQQRWRRRR

25

C-terminal fragment

Endocytosis (?)

DNA polyplexes

[54,55]

MT20

GIGAVLKVLTTGLPALISWI

20

Hydrophobic helix

DT, MD

DNA polyplexes

[56]

FL20

GIGAILKVLATGLPTLISWI

20

Hydrophobic helix

DT, MD

DNA polyplexes

[56]

Melittin [1–14]

GIGAVLKVLTTGLP

14

N-terminal fragment

DT

NC, LP

[57]

 

 

 

 

 

 

 

Wasp

 

 

 

 

 

 

Anoplin

GLLKRIKTLL-NH2

10

Amphipathic α-helix

DT, PF

FD

[60]

Anoplin dimer

(GLLKRIKTLL-NH2)2

20

C-N terminal dimer

DT, PF

-

[61]

Mastoparan

INLKALAALAKKIL-NH2

14

Amphipathic α-helix

DT, PF

-

[64]

Mastoparan-X

INWKGIAAMAKKLL-NH2

14

Amphipathic α-helix

DT, PF

-

[65]

Transportan

GWTLNSAGYLLGKINLKALAALAKKIL-NH2

27

Chimera

multiple

CI

[66]

TP10

AGYLLGKINLKALAALAKKIL-NH2

21

Shorter transportan

multiple

CI

[70,77]

TP10-5

AGYLLGKINLKKLAKL(Aib)KKIL-NH2

21

Helical stabilized

DT

FD

[71]

Transportan 9dR

GWTLNSAGYLLGKINLKALAALAKKIL(dR)9

36

Chimera

Endocytosis (?)

siRNA

[73]

Mitoparan

INLKKLAKL(Aib)KKIL-NH2

14

Mastoparan analog

DT

FD, AP

[78–80]

Notes: a-NH2, amidated peptide; anoplin C-N terminal dimer, as prepared by intermolecular triazole bridge; Aib, α-aminoisobutyric acid; bParticular structural characteristics of native, designed or analogous peptides; C-N terminal dimer, intermolecular dimerization involving the carboxyl-terminus of one peptide and the amino-terminus of the other; cDT, direct translocation; PF, pore-forming; MD, membrane-disruptive cell uptake; NR, non-disruptive penetration; RM, receptor-mediated endocytosis; HI, hydrophobic insertion; multiple, more than a single mechanism of cell entry, that can involve direct translocation and endocytosis; dFD, fluorescent dyes; CI, diverse types of cell impermeant cargoes; AP, apoptotic peptides; NC, nanocrystals; LP, large proteins; RD, radionuclides; EZ, enzymes; IR, infrared; “-,” not applicable.

Table 3. Examples of peptides and derivatives from arachnid venoms with cell-penetrating and cargo delivery capacity.

Peptide

Sequence

Size

Major structural features

Mechanism(s) of cell Penetration

Cargo delivery

Ref.

Spider

 

 

 

 

 

 

Latarcin-1 (Ltc1)

SMWSGMWRRKLKKLRNALKKKLKGE

25

cationic α-helix

DT, PF

FD

[81]

Ltc1-decapeptide (LDP)

KWRRKLKKLR

10

Designed

DT

FD

[82]

LDP-NLS

KWRRKLKKLRPKKKRKV

17

Chimera

ED

FD, EZ

[83]

Lycosin-I

RKGWFKAMKSIAKFIAKEKLKEHL

24

Amphipathic α-helix

DT, ND

FD, NP

[87,88]

R-lycosin-I

Ac-RGWFRAMRSIARFIARERLREHL-amide

24

Lys-->Arg analog

ED

FD

[89]

 

 

 

 

 

 

 

Scorpion

 

 

 

 

 

 

Chlorotoxin (CTX)

MCMPCFTTDHQMARKCDDCCGGKGRGKCYGPQCLCR

36

ICKfold/knottin

ED

FD, RD

[91–93]

[K15R/K23R]CTX

MCMPCFTTDHQMARRCDDCCGGRGRGKCYGPQCLCR

36

Mutant analog

ED

FD

[95]

[K15R/K23R/Y29W]CTX

MCMPCFTTDHQMARRCDDCCGGRGRGKCWGPQCLCR

36

Mutant analog

ED

FD

[95]

Maurocalcine (MCa)

GDCLPHLKLCKENKDCCSKKCKRRGTNIEKRCR

33

ICK fold/knottin

DT, ND

CI, NP

[99–101]

MCaUF1–9-C

GDAbuLPHLKLC

10

Truncated, unfold

DT

FD

[105]

Imperatoxin A (IpTxa)

GDCLPHLKRCKADNDCCGKKCKRRGTNAEKRCR

33

ICK fold/knottin

n

FD

[109]

Hadrucalcin (HdCa)

SEKDCIKHLQRCRENKDCCSKKCSRRGTNPEKRCR

35

ICK fold/knottin

DT, ND

-

[110]

HadUF1−11 (H11)

SEKDAbuIKHLQR-C

12

N-terminal fragment

DT, ND, ED (?)

NP

[111]

WaTx

ASPQQAKYCYEQCNVNKVPFDQCYQMCSPLERS

33

CS-helical hairpin

DT, ND

FD

[112]

Notes: Ac, acetylated; -NH2, amidated; Abu, Lys®Arg replacement; C-S, cysteine stabilized α-helix; DT, direct translocation; PF, pore-forming; ED, endocytosis; ND, non-disruptive penetration; RM, receptor-mediated endocytosis; HI, hydrophobic insertion; multiple, more than a single mechanism of cell entry, that can involve direct translocation and endocytosis; “n“, not informed; FD, fluorescent dyes; EZ, enzymes; NP, nanoparticles; RD, radionuclides; CI, diverse types of cell impermeant cargoes; “-“, not applicable.

Table 4. Examples of peptides and derivatives from fish and amphibian secretion and snake venom, with cell-penetrating and cargo delivery capacity.

Peptide

Sequence a

Size

Major Structural Features b

Mechanism(s) of Cell Penetration c

Cargo Delivery d

Ref.

Fish

 

 

 

 

 

 

Ac-Pardaxin P5

Ac-GFFALIPKIISSPLFKTLLSAVGSALSSSGDQE 

33

Hydrophobic helix

DT, PF

FD

[117]

 

 

 

 

 

 

 

Amphibian

 

 

 

 

 

 

bombesin

EQKLGNQWAVGHLM-NH2

14

helical hairpin

RM

-

[118–120]

Tat-K3-bombesin1-14

RKKRRQRRRGGCGEQKLGNQWAVGHLM-NH2

27

Chimera

RM

RD

[124]

 

 

 

 

 

 

 

Snake

 

 

 

 

 

 

Crotamine

YKQCHKKGGHCFPKEKICLPPSSDFGKMDCRWRWKCCKKGSG

42

β-defensin fold

multiple

FD, CI

[138]

NrTP1

YKQCHKKGGKKGSG

14

N«C splice variant

multiple

FD

[146,147]

NrTP6

YKQSHKKGGKKGSG

14

NrTP1 CysDSer

multiple

FD, EZ

[148]

DY676-NrTP6

C-YKQSHKKGGKKGSG

15

Extra Cys

 

IR

[152]

CyLop-1

CRWRWKCCKK

10

Encrypted sequence

DT, ED

FD

[145]

 

 

 

 

 

 

 

Crotalicidin

KRFKKFFKKVKKSVKKRLKKIFKKPMVIGVTIPF

34

Amphipathic α-helix

DT, PF

FD

[176]

Ctn[15–34]

KKRLKKIFKKPMVIGVTIPF-NH2

20

C-terminal fragment

DT, PF

FD

[170,176]

cathelicidin- BF30

KFFRKLKKSVKKRAKEFFKKPRVIGVSIPF

30

Shorter analog

DT, PF

-

 

Cardiotoxin

LKCNKLIPLAYKTCPAGKNLCYKMFMVSNKTVPVKRGCID ACPKNSLLVKYVCCNTDRCN

60

Three-finger fold

DT, HI

FD

[179]

PLA2 C-terminal

KKYKAYFKFKCKK-NH2

13

C-terminal fragment

DT, MD

-

[185]

PLA2 fragment K®R

RRYRAYFRFRCRR-NH2

13

Mutant K-->R

DT, MD

-

[185]

BPP-13a

<EGGWPRPGEIPP

12

Native

DT (?)

-

[187]

desPyr-BPP13a

GGWPRPGPEIPP

12

Mutant analog

DT (?)

FD

[187]

Notes: aAc, acetylated; -NH2, amidated; <E, pyroglutamic; DT, direct translocation; bN«C, a spliced derivative of crotamine, comprising the N-terminal residues 1 to 9 and C-terminal 38 to 42 ([1–9] ~ [38–42]); NrTP6, an NrTP1 with a Cys-Ser replacement; DY676, near-infrared dye; cDT, direct translocation; PF, pore-forming; RM, receptor-mediated endocytosis; multiple, more than a single mechanism of cell entry, that can involve direct translocation and endocytosis; ED, endocytosis; HI, hydrophobic insertion; MD, membrane-disruptive cell uptake; dFD, fluorescent dyes; RD, radionuclides; EZ, enzymes; IR, infrared dye; “-“ not applicable;

These designed CPPs from venom peptides have found applications in biomedicine and biopharmaceutical biotechnology, in diagnostics to detect populations of diseased cells or tissues, in therapy to induce selective cell death through molecular delivery of cell-impermeant drugs, organic compounds, radionuclides and crystal, and metallic nanoparticles. The discoveries of cell-penetrating venom peptides and animal toxins will continue to contribute to the basic and applied research in this expanding field of CPPs derived from components of poisonous and venomous animals. The cell-penetrating venom peptides and toxin derivatives have expanded the library of animal toxins that act intracellularly and have presently served and will be useful in the future, for a variety of biomedical and biotechnological applications.

 

References

  1. Singh, S.; Peláez, F. Biodiversity, chemical diversity and drug discovery. Prog. Drug Res. 2008, 65, 141, 143–174.
  2. Nelsen, D.R.; Nisani, Z.; Cooper, A.M.; Fox, G.A.; Gren, E.C.; Corbit, A.G.; Hayes, W.K. Poisons, toxungens and venoms: Redefining and classifying toxic biological secretions and the organisms that employ them. Biol. Rev. Camb. Philos. Soc. 2014, 89, 450–465.
  3. Modahl, C.M.; Brahma, R.K.; Koh, C.Y.; Shioi, N.; Kini, R.M. Omics Technologies for Profiling Toxin Diversity and Evolution in Snake Venom: Impacts on the Discovery of Therapeutic and Diagnostic Agents. Annu. Rev. Anim. Biosci. 2020, 8, 91–116.
  4. Kazuma, K.; Masuko, K.; Konno, K.; Inagaki, H. Combined Venom Gland Transcriptomic and Venom Peptidomic Analysis of the Predatory Ant Odontomachus monticola. Toxins 2017, 9, 323.
  5. Wiezel, G.A.; Shibao, P.Y.T.; Cologna, C.T.; Filho, M.R.; Vieira, U.C.; De Pauw, E.; Quinton, L.; Arantes, E.C. In-Depth Venome of the Brazilian Rattlesnake Crotalus durissus terrificus: An Integrative Approach Combining Its Venom Gland Transcriptome and Venom Proteome. J. Proteome Res. 2018, 17, 3941–3958.
  6. Liao, Q.; Gong, G.; Poon, T.C.W.; Ang, I.L.; Lei, K.M.K.; Siu, S.W.I.; Wong, C.T.T.; Baptista, R.G.; Lee, S.M. Combined transcriptomic and proteomic analysis reveals a diversity of venom-related and toxin-like peptides expressed in the mat anemone Zoanthus natalensis (Cnidaria, Hexacorallia). Arch. Toxicol. 2019, 93, 1745–1767.
  7. Carreto, R.S.; Estrella, V.R.; Bobadilla, P.T.; Navarro, L.A.; Sarabia, B.J.; Piñera, R.E.; Verleyen, J.J.; Rodríguez, E.; Almazán, R.C. Transcriptomic and Proteomic Analysis of the Tentacles and Mucus of Anthopleura dowii Verrill, 1869. Mar. Drugs 2019, 17, 436.
  8. Aili, S.R.; Touchard, A.; Hayward, R.; Robinson, S.D.; Pineda, S.S.; Lalagüe, H.; Vetter, I.; Undheim, E.A.B.; Kini, R.M.; Escoubas, P.; et al. An Integrated Proteomic and Transcriptomic Analysis Reveals the Venom Complexity of the Bullet Ant Paraponera clavata. Toxins 2020, 12, 324.
  9. Dalmazzone, S. Economics and Policy of Biodiversity Loss. In Sustainable Development and Environmental Management: Experiences and Case Studies; Clini, C., Musu, I., Gullino, M.L., Eds.; Springer: Dordrecht, The Netherlands, 2008; pp. 451–466.
  10. Kessler, P.; Marchot, P.; Silva, M.; Servent, D. The three-finger toxin fold: A multifunctional structural scaffold able to modulate cholinergic functions. J. Neurochem. 2017, 142, 7–18.
  11. Shafee, T.M.A.; Lay, F.T.; Phan, T.K.; Anderson, M.A.; Hulett, M.D. Convergent evolution of defensin sequence, structure and function. Cell. Mol. Life Sci. 2017, 74, 663–682.
  12. Zhang, S.K.; Song, J.W.; Gong, F.; Li, S.B.; Chang, H.Y.; Xie, H.M.; Gao, H.W.; Tan, Y.X.; Ji, S.P. Design of an α-helical antimicrobial peptide with improved cell-selective and potent anti-biofilm activity. Sci. Rep. 2016, 6, 7394.
  13. Joliot, A.; Prochiantz, A. Transduction peptides: From technology to physiology. Nat. Cell Biol. 2004, 6, 189–196.
  14. Mándity, I.M.; Fülöp, F. An overview of peptide and peptoid foldamers in medicinal chemistry. Expert Opin. Drug Discov. 2015, 10, 1163–1177.
  15. Kalafatovic, D.; Giralt, E. Cell-Penetrating Peptides: Design Strategies beyond Primary Structure and Amphipathicity. Molecules 2017, 22, 1929.
  16. Sandvig, K.; van Deurs, B. Delivery into cells: Lessons learned from plant and bacterial toxins. Gene Ther. 2005, 12, 865–872.
  17. Baptista, R.G.; Kerkis, A.; Silva, P.Á.R.; Hayashi, M.A.F.; Kerkis, I.; Tetsuo, Y. Membrane-translocating peptides and toxins: From nature to bedside. J. Braz. Chem. Soc. 2008, 19, 211–225.
  18. Sandvig, K.; Bergan, J.; Kavaliauskiene, S.; Skotland, T. Lipid requirements for entry of protein toxins into cells. Prog. Lipid Res. 2014, 54, 1–13.
  19. Bechara, C.; Sagan, S. Cell-penetrating peptides: 20years later, where do we stand? FEBS Lett. 2013, 587, 1693–1702.
  20. Copolovici, D.M.; Langel, K.; Eriste, E.; Langel, Ü. Cell-Penetrating Peptides: Design, Synthesis and Applications. ACS Nano 2014, 8, 1972–1994.
  21. Gautam, A.; Singh, H.; Tyagi, A.; Chaudhary, K.; Kumar, R.; Kapoor, P.; Raghava, G.P. CPPsite: A curated database of cell penetrating peptides. Database J. Biol. Databases Curation 2012, 2012.
  22. Agrawal, P.; Bhalla, S.; Usmani, S.S.; Singh, S.; Chaudhary, K.; Raghava, G.P.; Gautam, A. CPPsite 2.0: A repository of experimentally validated cell-penetrating peptides. Nucleic Acids Res. 2016, 44, D1098–D1103.
  23. Kardani, K.; Bolhassani, A. Cppsite 2.0: An Available Database of Experimentally Validated Cell-Penetrating Peptides Predicting their Secondary and Tertiary Structures. J. Mol. Biol. 2020, 6703.
  24. Wei, H.H.; Yang, W.; Tang, H.; Lin, H. The Development of Machine Learning Methods in Cell-Penetrating Peptides Identification: A Brief Review. Curr. Drug Metab. 2019, 20, 217–223.
  25. Yamashita, H.; Kato, T.; Oba, M.; Misawa, T.; Hattori, T.; Ohoka, N.; Tanaka, M.; Naito, M.; Kurihara, M.; Demizu, Y. Development of a Cell-penetrating Peptide that Exhibits Responsive Changes in its Secondary Structure in the Cellular Environment. Sci. Rep. 2016, 6.
  26. Eiríksdóttir, E.; Konate, K.; Langel, Ü.; Divita, G.; Deshayes, S. Secondary structure of cell-penetrating peptides controls membrane interaction and insertion. Biochim. Biophys. Acta (BBA) Biomembr. 2010, 1798, 1119–1128.
  27. Milletti, F. Cell-penetrating peptides: Classes, origin and current landscape. Drug Discov. Today 2012, 17, 850–860.
  28. Borrelli, A.; Tornesello, A.L.; Tornesello, M.L.; Buonaguro, F.M. Cell Penetrating Peptides as Molecular Carriers for Anti-Cancer Agents. Molecules 2018, 23, 295.
  29. Baptista, R.G.; Campelo, I.S.; Morlighem, J.R.L.; Melo, L.M.; Freitas, V.J.F. Cell-penetrating peptides (CPPs): From delivery of nucleic acids and antigens to transduction of engineered nucleases for application in transgenesis. J. Biotechnol. 2017, 252, 15–26.
  30. Huang, Y.W.; Lee, H.J. 13-Cell-penetrating peptides for medical theranostics and targeted drug delivery. In Peptide Applications in Biomedicine, Biotechnology and Bioengineering; Koutsopoulos, S., Ed.; Woodhead Publishing: Cambridge, UK, 2018; pp. 359–370.
  31. Pierantoni, G.M.; Paladino, S. Cell-penetrating peptides: Two faces of the same coin. Biochem. J. 2020, 477, 1363–1366.
  32. Ruseska, I.; Zimmer, A. Internalization mechanisms of cell-penetrating peptides. Beilstein J. Nanotechnol. 2020, 11, 101–123.
  33. Tünnemann, G.; Martin, R.M.; Haupt, S.; Patsch, C.; Edenhofer, F.; Cardoso, M.C. Cargo-dependent mode of uptake and bioavailability of TAT-containing proteins and peptides in living cells. FASEB J. Off. Publ. Fed. Am. Soc. Exp. Biol. 2006, 20, 1775–1784.
  34. Andaloussi, E.S.; Järver, P.; Johansson, H.J.; Langel, U. Cargo-dependent cytotoxicity and delivery efficacy of cell-penetrating peptides: A comparative study. Biochem. J. 2007, 407, 285–292.
  35. Hedegaard, S.F.; Derbas, M.S.; Lind, T.K.; Kasimova, M.R.; Christensen, M.V.; Michaelsen, M.H.; Campbell, R.A.; Jorgensen, L.; Franzyk, H.; Cárdenas, M.; et al. Fluorophore labeling of a cell-penetrating peptide significantly alters the mode and degree of biomembrane interaction. Sci. Rep. 2018, 8, 6327.
  36. Patel, S.G.; Sayers, E.J.; He, L.; Narayan, R.; Williams, T.L.; Mills, E.M.; Allemann, R.K.; Luk, L.Y.P.; Jones, A.T.; Tsai, Y.H. Cell-penetrating peptide sequence and modification dependent uptake and subcellular distribution of green florescent protein in different cell lines. Sci. Rep. 2019, 9, 6298.
More
Information
Contributor MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register :
View Times: 649
Entry Collection: Peptides for Health Benefits
Revisions: 2 times (View History)
Update Date: 12 Mar 2021
1000/1000
Video Production Service