CA2338885A1 - Antimicrobial peptides isolated from the skin of american frogs - Google Patents

Antimicrobial peptides isolated from the skin of american frogs Download PDF

Info

Publication number
CA2338885A1
CA2338885A1 CA002338885A CA2338885A CA2338885A1 CA 2338885 A1 CA2338885 A1 CA 2338885A1 CA 002338885 A CA002338885 A CA 002338885A CA 2338885 A CA2338885 A CA 2338885A CA 2338885 A1 CA2338885 A1 CA 2338885A1
Authority
CA
Canada
Prior art keywords
seq
ranatuerin
rana
peptide
temporin
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
CA002338885A
Other languages
French (fr)
Inventor
J. Michael Conlon
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Individual
Original Assignee
BioNebraska Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by BioNebraska Inc filed Critical BioNebraska Inc
Publication of CA2338885A1 publication Critical patent/CA2338885A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/04Linear peptides containing only normal peptide links
    • C07K7/08Linear peptides containing only normal peptide links having 12 to 20 amino acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/463Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from amphibians
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Abstract

The invention is directed to 7 families of Rana peptides isolated from the skin of 6 species of American frogs, as well as modified and truncated versions thereof. These peptides are termed Rana peptides and have antibacterial activity.

Description

ANTIMICROBIAL PEPTIDES ISOLATED FROM THE
SHIN OF AMERICAN FROGS
This application is based on U.S. Provisional Application Serial No. 60/096,607, filed August 14, 1998.
Background of the Invention The indiscriminate use of antibiotics in hospitals has created drug-resistant pathogens that cause difficult=to-cure infections. This problem is particularly serious in the case of AIDS, tuberculosis and other to immunocompromised p<itients. For example, data collected by the Synercid Microbiology Assessment of Resistance Trends surveillance project have shown that more than 31 % of 17,000 bacterial isolates of Streptococcus pneumoniae obtained from patients at U.S. hospitals were intermediately or completely resistant to penicillin. The incidence of methicillin-resistant strains of 15 Staphylococcus aureus was shown by this project to be 29%. Tuberculosis, which was allegedly eradicated in the United States, has made a horrific reappearance in forms that are resistant to traditional isoniazid therapy.
Bacterial infection in animals has also become drug-resistant in some cases. The economic toll is staggering as well. It has been estimated that drug-resistant 2o infections acquired in hospitals nearly triple the cost and duration of an average hospital stay. Hoffert, S.P., "Companies seeking solutions to emerging drug resistance," The Scientist, 12(8), 1-6 (1998).
As a result of earlier dramatic successes in the development of antimicrobial agents, most large pharmaceutical companies during the 1980s and 25 early 1990s cut back or eliminated programs of research and development of new compounds on the grounds that the battle against infectious diseases had been won. However, the emergence of increasing numbers of pathogenic microorganisms with resistance to the commonly used antibiotics has greatly stimulated searches for novel antimicrobial agents to fight drug-resistant 30 infections.

Among those searches is the investigation of novel antibiotic peptides from Anuran (frog and toad) skin. Amphibians of necessity live in a warm, moist environment that is particularly conducive to the growth of microorganisms. As a result, Anurans have evolutionarily developed effective 5 strategies for their own protection.
In particular, Anurans synthesize polypeptides with a broad spectxum of antimicrobial activity through the ~'~ular glands present in their skin. The bioactive peptides are released into skin secretions in a holocrine fashion upon stress or injury and protect against invasion by pathogenic microorganisms.
The to amphibian antimicrobial peptides are generally synthesized as members of structurally-related families and examples include magainins from Xenopus laevis, bombinins from Bombina variegata and Bombina orientalis, dermaseptins from P):yllomedusa sauvagii and Phyllomedusa bicolor, buforins from Bufo bufo gargurizans, and caerins from Litoria chloris and Litoria 15 splendida. Despite the sequence similarities, the members of a particular family have distinct spectra of antimicrobial activity and it has been speculated that this molecular diversity is import ~ protecting the animal from invasion by a wide array of different microorganisms.
Frogs from the genus Rana constitute an extremely diverse and widely 2o distributed group with an estimated 250 species worldwide and at least 36 species having been identified in North America 2 et . Analysis of skin secretions and/or skin extracts of different species of Rapid frogs has led to the following characterizations: 1) gaegurins and rugosins from R. rugosa, 2) brevinins from R. brevipoda porsa, R. esculenta and R. sphenocephala, 3) 25 esculentins from R. es~lenta, 4) ranalexin and ranatuerms from R.
catesbeiana and S) temporins axed ranatuerin 1T from R. temporaria. Peptides of the brevinin family have also been isolated from an extract of gastric tissue from R.
esculenta.
Further examples of such peptides are discussed in:
1 ) L,azarus, L.H., and Attila, M., "The toad, ugly and venomous, wears yet a precious jewel in his skin," Prog. Neurobiol., 41, 473-507 (1993);
2) ~:asloff, M., "Magainins, a class of antimicrobial peptides 5 from Xenopus skin: isolation, characterization of two active forms and partial cDNA sequence of a precursor," Proc. Natl. Acad. Sci. USA 84, 5449-5453, (1987);
3) Buforins from Bufo bufo gargarizans- Park, C.B., Kim, M.S., and Kim, S.C., Biochem. Biophys. Res. Commun., 218, 408-413, (1996);
to 4) Bombinins from Bombina variegata- Simmaco, M., Barra, D., Chiarini, F., Noviello, L., Melchiorri, P., Kreil, G., and Richter, K., "A
family of bombinin-related peptides from the skin of Bombina variegata, " Eur.
J. Biochem.199, 217 :!22, (199I);
5) Bombina orientalis- Gibson, B.W., Tang, D., Mandrell, ~5 R., Kelly, M., and Spindel, E.R., "Bombinin-like peptides with antimicrobial activity from skin secretions of the Asian toad, Bombina orientalis.," J.
Biol.
Chem. 266, 23103-23111, (1991);
6) Dermaseptins from Phyllomedusa sauvagii- Mor, A., Hani, K., and Nicolas" P., "The vertebrate peptide antibiotics dermaseptins have 20 overlapping structural features but target specific microorganisms," J.
Biol.
Chem. 269, 31635-31641, (1994);
7) Caerins from Litoria chloris- Steinborner, S.T., Currie, G.J., Bowie, J.H., Wallace, J.C., and Tyler, M.J., "New antibiotic caerin 1 peptides from the skin secretion of the Australian tree frog Litoria chloris, 25 comparison of the activities of the caerin 1 peptides from the genus Litoria," J.
Peptide Res. 51, 121-126, (1998);
8) Gaegurins from the Korean frog, Rana rugosa- Park, J.M., Jung, J.E., and Lee, B.J., "Antimicrobial peptides from the skin of a Korean frog, Rana rugosa," Biochem. Biaphys. Res. Commun. 205, 948-954, (1994);

9) Brevinins from the 3apanese frog, Rana brevipoda porsa-Morikawa, N., Hagiwara, K., and Nakajima, T., "Brevinin-1 and -2, unique antimicrobial peptides from the skin of the frog, Rana brevipoda porsa,"
Biochem. Biophys. Res. Commun. 189, 184-190 (1992); and 5 10) ~yculentins and brevinins from the European frog, Rana esculenta- Simmaco, M., Mignogna, G., Barra, D., and Bossa, F., "Antimicrobial peptides from skin secretions of Rana esculenta," J. Biol.
Chem.
269, 11956-11961, (1994).
Some of these peptides discussed in the art have the following sequences.
to Gaegurin 5 FLGALFKVASKVLPSVKCAITKKC (SEQ m NO:
1) Gaegurin 6 FI.PLLAGLAANFLPTIICKISYKC (SEQ D7 NO:
is 2) Brevinin 1 FLPVLAGIAAKVVPALFCKITKKC (SEQ >D NO:
3) 20 Brevinin lE FI:,PLLAGLAANFLPKIFCKITRKC (SEQ m NO:
4) Peptide A1 FLPAIAGILSQLF (SEQ m NO:
5) Peptide B9 FI:PLIAGLLGKLF (SEQ m NO:
6) To date, no antibacterial peptides have been isolated from the adult American bullfrog and studies of immature bullfrogs have indicated the presence of only one antibacterial peptide, ranalexin having the sequence:
FLGGLIKIVPAMICAVTKKC {SEQ ID NO: 7).
The present invention has as its goal the isolation and characterization of amphibian antimicrobial peptides to assess their value as taxonomic and phylogenetic markers. A further goal of the present invention is the study of seven different classes o;f Anuran peptides with antimicrobial activity which are obtained from extracts of the skins of six closely related species of North American frogs of the genus Rana: the spotted frog R. luteiventris, the Rio Grande leopard frog R. berlandieri,~reen frog R. clamitans, the pig frog R.
grylio. and the northern leopard frog R. pipiens and the North American bullfrog, R catesbeiana.
Summary of the Invention These and other objects are achieved by the present invention which is directed to seven families of peptides obtained from North American frogs (families of Rana peptides), to purified Rana compositions containing these peptides, to purified antibacterial extracts from Rana skin, to modified Rana 1o peptides, to truncated Rana peptides, to pharmaceutical compositions containing Rana peptides or modified forms thereof, and to methods for treatment of bacterial infections using Rang peptides or modified forms thereof.
In particular, the invention is directed to seven families of Rana peptides shown below in Table 1. These Rana peptides have antibacterial activity and are 15 selected from any of the following seven peptide families. The Rang peptides may be in the form of their C-terminus carboxylic acids, or modified as C-terminus amides or C-terminus esters. The amides may be a simple anode (CONH2), which is the isolated form for the Temporin-A and B Fanmilies, or amides derived from Cl to C10 primary, secondary or tertiary aliphatic or 2o aromatic amines. The esters (COOR wherein the R group is the alcohol residue) are derived from C 1 to (:10 aliphatic or aromatic alcohols. The preferred C-terminus forms for the Temporin-A and Temporin-B families are the simple amides.
In these formulas, the single letters designate amino acid residues 25 according to accepted ccmvention except that in the formulas for the peptides designated with a Family name followed by the term "mod", the letters X, B, Z, U, O, J' and J, which are; not conventional single letter designations of amino acid residues, are used as symbols to indicate a substitution of either any of the amino acid residues occurring at the same position within the other peptides of the same family or a conservative substitution of an amino acid residue for the amino acid substitution of an amino acid residue at the same position for any of the peptides within the same family of peptides. These symbols for the Family-mod formulas have two identifiers: first is the identity of the symbol itself (X, B, Z, U, O, J' or J); second is the locationlposition of the symbol within the formula. For example, X of the Ranatuerin-1 mod appears at the 3, 16 and 21 positions of the formula. At each of these positions, X has a different meaning depending upon the corresponding amino acid residues at the same position of the other members of the family. In the case of the Ranatuerin-1 mod formula, io the X means that at the 3 position, the amino acid residue can be L, I or a conservative substitution therefor, at the 16 position the amino acid residue can be L, F or a conservative substitution therefor, and at the 21 position the amino acid residue can be I, V or a conservative substitution therefor.
The term conservative substitution means substitution of an amino acid 15 residue by another that has the same side chain ionicity, basicity, acidity, lipophilicity or hydrogen bonding character as the residue being replaced.
Examples include isoleucine (I), leucine (L), alanine (A), valine (V), phenyl analine (F), proline (P) and glycine (G) as an interchangeable group; lysine (K), histidine (I~ and arginine (R) as an interchangeable group, serine (S), tyrosine 20 (~ and threonine (T} as an interchangeable group; cysteine (C) and methionine (IVi)as an interchangeable group asparagine (I~, glutamine (~ and tryptophan (V~ as an interchangeable group, and aspartic acid (D) and glutamic acid (E) as an interchangeable group. The letters after these amino acids are their single letter symbols.
25 The dashes in the ranatuerin-2 family peptides provide alignment of homologous peptide residues. The single and multiple dashes indicate a bond between the two amino acid residues on either side of the space occupied by the dashes. For the purpose; of determining amino acid substitutions for this family, each dashed line constitutes a spacer instead of an amino acid residue. For 3o example, the 5 dashed lines of R.anatuerin-2 stand in the place of 5 amino acid residues so that the A residue following the dashed lines is counted in the 19'"
position for the purpose of placing the amino acid residues of the individual peptides of the family into homologous alignment. Likewise, the 4 and 8 dashed lines of the Ranatuerin-~Ca and Ranatuerin-2Cb peptides respectively place the A residue following those lines into the 19'~ position.

Families of Rana Peptides to Ranatuerin-1 SMLSVLKNLGKVGLGLVACKINKQC (SEQ ID NO:
8) Ranatuerin-1G SMISVLKNLGKVGLGFVACKVNKQC (SEQ ID NO:
9) Ranatuerin-1C SMLSVLKNLGKVGLGLVACKINKQC (SEQ ID NO:

10) 15 Ranatuerin-1 SMXSVLKNLGKVGLGXVACIC3~IKQC {SEQ 117 NO:
mod 11) Eanatuerin-2 fami~
20 Ranatuerin-2 GL,FLDTLKGAAKD-----AGK-LEGLKCKITGCKLp(SEQ ID NO:
12) Ranatuerin-3 GFL-DBICNLGKTF----AGHIvII,DKIKCTIGTCPPSP(SEQ lD

NO: 13) Ranatuerin-2P GLM-DTVKNVAKNL-___AG~~,K~.~ (SEQ ID

NO: 14) 25 Ranatuerin-2BGLL-DTIKGVAKTV---AASMLDKLKCKISGC {SEQ ID

NO: 15) Ranatuerin-2Ca GLFLDTLKGAAKDV---AGKLLEGLKCKIAGCKP(SEQ ID NO:
16) Ranatuerin-2Cb GLFLDTLKGL------AGKLLQGLKCIKAGCKP (SEQ ID NO:
17) Ranatuerin-2La GI-LDSFKGVAKGVAKDLAGKLLDKLKCKTTGC (SEQ ID NO:
18) 30 Ranatuerin-2LbGILSSI-KGVAKGVAKNVAAQLLDTLKCRTTGC {SEQ ID NO:
19) Ranatuerin-2G GLLLDTLKGAAKDI---AGIALEKLKCKTTGCKP(SEQ ID N0:
20) Ranatuerin-2 GBBBBBBKBBBBBBBBBBABBBBBBBKCBBBBCBBB
mod (SEQ ID NO: 21 ) Esculentin-2 familv Esculentin-2L GII,SLFTGGIKALGKTLFKMAGKAGAEFiLACKATNQC(SEQ

ID NO: 22) Esculentin-2B GLFSILRGAAKFASKGLGKDLTKLGVDLVACKISKQC(SEQ

ID NO: 23) 40 Esculentin-2PGFSSIFRGVAKFASKGLGKDLARLGVNLVACKISKQC(SEQ

)D NO: 24) Esculentin-2 GZZSZZZGZZKTrZZKZLZKZZZZZGZZZZ,ACICZ,ZZQC(SEQ
mod )D NO: 25) 45 Brevinin-I famllv Ranatuerin-4 FLP;FIARLAAKVFPSIICSVTKKC (SEQ >D NO:
26) Brevinin-1La FLPMLAGLAASMVPKLVCLTTKKC (SEQ
ID NO:

27) Brevinin-1Lb FLPMLAGLAASMVPKFVCLITKKC (SEQ
LD NO:

28) S Brevinin-1Ba FLPAIAGMAAKFLPKIFCAISKKC (SEQ
ID NO:

29) Brevinin-1Bb FLl?ALAGMAAKFLPKIFCAISKKC(SEQ
ID NO:

30) Brevinin-1Bc FLl?FLAGVAAKFLPKIFCAISKKC(SEQ
ID NO:

1031) Brevinin-1Bd FLPAIAGVAAKFLPKIFCAISKKC (SEQ
ID NO:

32) Brevinin-1Be FLPAIVGAAAKFLPKIFCVISKKC (SEQ
ID NO:

33) 15Brevinin-1Bf FLPFIAGMAANFLPKIFCAISKKC (SEQ
ID NO:

34) Brevinin-IPa FLPIIAGVAAKVFPKIFCAISKKC (SEQ
ID NO:

35) Brevinin-1Pb FLFIIAGLAAKVFPKIFCAISKKC (SEQ
ID NO:

2036) Brevinin-1Pc FLF'LLASVAAKVFSKIFCAISKKC(SEQ
ID NO:

37) Brevinin-1Pd FLF'IIASVAANVFSKIFCAISKKC(SEQ
ID NO:

38) 25Brevinin-1Pe FLF'IIASVAAKVFPKIFCAISKKC(SEQ
ID NO:

39) Brevinin-1 FLPUIUGUAAUUUPUUUCUIUKKC (SEQ
mod ID NO:

40) 30 Ranalexin familv Ranalexin-1 GFI,GGLMKAFPALICAVTKKC
(SEQ ID

NO: 41) Ranalexin-1 CFL,GGLMKIIPAAFCAVTKKC
(SEQ ID

35 N0: 42) F:analexin-1 FLCiGLMKJ'J'PAJ'J'CAVTKKC
mod (SEQ m NO: 43) T~porin A fa~~vs the Gt~us carboxamide a Ranatuerin 5 FLPIASLLGKYL
(SEQ ID NO: 44) Ranatuerin 6 FISAIASMLGKFL (SEQ ID NO: 45) Ranatuerin 7 FLSALASMLGKFL (SEQ 1D NO: 4~

F:anatuerin FISALASFI,GKFL (SEQ ID NO: 47) 45 Ranatuerin FLFPLTTSFL,SKVL (SEQ 1D NO: 48) Temporin-1Ca FLPFI,ATLLSKVL ) Temporin-1Cb FLPFI,AKILTGVL (SEQ 1D NO: 50 Temporin-1Cc FLPLFASI,IGKKL (SEQ ID NO: 51) Temporin-IPa FLPIVGKLLSGLL (SEQ 1D NO: 52) 50 Temporin-1LcFLPILINLIF1KGL (SEQ ID NO: 53) Temporin-A mod FOOOOOOOOOOOLO (SEQ 1D NO: 54) T'~nporin B Familv as the Gterminus carboxa ide Temporin-1LaVLPLISMALGKLL (SEQ DJ NO:
55) Temporin-1LbNFI,GTLINLAKKIM (SEQ ID NO:
56) Temporin-IGaSILPTIVSFLSKFV (SEQ ID NO:
57) Temporin-1GbSILI'TIVSFLSKFL (SEQ ID NO:
58) Temporin-IGcSILPTIVSFLTKFIL (SEQ ID NO:
59) Temporin-1GdFIIFLIASFLSKFIL
(SEQ ID NO:
60) Temporin-B JJJJJIJSFLJKJJL (SEQ ID NO:
mod 61) l0 The purified compositions of the invention includes any composition containing one or more of the foregoing peptides obtained from a natural source such as Rana skin or obtained from a synthetic source.
The modified Rana peptides of the invention include any of the foregoing Rana peptides designated as Ranatuerin-1 mod, Ranatuerin-2 mod, Esculentin-2 15 mod, Brevinin-1 mod, Brevinin-2 mod, Temporin-A mod and Temporin-B mod wherein the symbols X, ;B, Z, U, O, J' and J indicate substitutions of amino acid residues as discussed above. The modified Rana peptides of the invention also include the C-terminus amides and esters described above, which are derived from C 1 to C 10 aliphatic. or aromatic amines or alcohols. Further modified Rana 2o peptides include those wherein one or more of the amino acid residues valine (~, alanine (A), lysine (K), and glutamic acid (E) are substituted for the glycine (G) residues. Further modified Rang peptides of the invention include any of the foregoing Rana peptides wherein arginine. (R) is substituted for one or more of the lysine (K) residues.
25 Additional modified Rana peptides contain modified side chains so that any of the Rana peptides with amino acid residues having functionalized side chains can be modified with amidation or esterification groups. Examples include those in which the epsilon amino group of a lysine (K) residue has been coupled to a C,o to CZO favy acid such a palinitate or glycosylated sugar, those in 3o which the epsilon carboxy group of aspartic (D) or glutamic (E) acid residues or hydrolyzed glutamine (Q) residue have been coupled to fatty alcohols or glycosylated sugars, or those in which the epsilon hydroxyl group of threonine (T), tyrosine ('I~, or serine (S) residues have been coupled to fatty acids or glycosylated sugars. In particular, ranatuerin 1 having any of these side chain modifications, where the appropriate amino acid residue is present, is preferred.
In addition, the modified Rana peptides include those in which a truncated fragment is synthetically condensed with a non-natural peptide or peptide sequence optionally containing heterocyclic organic moieties such as a beta or gamma amino acid, an aliphatic diamine, an aliphatic or aromatic dicarboxylic acid, pyridine carboxylic acid, aromatic diols and the like.
The truncated versions of Rana peptides include the fragments formed by cleaving at any of the glycine residues or between any repeat residues, and fi-agments formed by N-terminal deletion of pairs of amino acid residues.
10 Further truncated versions include those that are cleaved at the single or multiple dash symbols. Far example, truncated versions of ranatuerin 1 include those containing amino acid residues 1-9, the fragment containing amino acid residues 16 - 25, N-terminally deleted fragments such as ranatuerin 1 {3-25), (5-25), (7-25), (9-25), (11-25), (13-25), (15-25), (17-25) and {19-25).
15 The pharmaceutical composition of the invention includes the combination of one or more Rana peptides with a pharmaceutical carrier rendering the composition suitable for an appropriate route of administration to a patient.
The method of treatment of the invention involves administration of an 2o effective amount of the pharmaceutical composition by an appropriate route to a patient suffering from a target bacterial infection.
Brief Description of the Drawinøs FIG. 1 presents a graph of a gel permeation chromatography of an extract 25 of the skin of Rang luteiventris.
FIGS. 2A, B present graphs of a reverse-phase HPLC of the antimicrobial activity of R. luteiventris skin.
FIGS. 3A, B present graphs of the chromatographic results of a partial purification of the fi~actians from R. luteiventris skin having antimicrobial 30 activity.

FIGS. 4A, B and C present graphs of the chromatographic results of a purification of ranatuerin 2La, ranatuerin 2Lb and esculentin 2L.
FIG. 5 presents gel permeation chromatography on Sephadex G-25 of an extract of the skin of Rana catesbeiana after partial purification on Sep-Pak cartridges. The fractions in zone A contained ranatuerins 1-5 and the fractions in zone B contained ranatiierins 6-9.
FIGS. 6A and 6B present reversed-phase HI'LC on a semipreparative Vydac C-18 column. FIG. 6A presents the pooled fractions from zone A in Fig.
and Fig. 6B presents the pooled fractions from zone B in FIG. 5. The dashed to line shows the concentration of acetronitrile in the eluting solvent and the bars show the fractions containing the ranatuerins indicated.
T~etailed Description of the Invention The present invention is directed to a systematic investigation of extracts of the skin of adult Ranid frogs and characterization of all the peptides with 15 inhibitory activity towards bacteria. The skins of frogs of the genus Rana produce a diverse array of peptides with antimicrobial activity. A comparison of the amino acid sequences of these peptides demonstrates that they may be arranged in families on the basis of structural similarity. Attempts to catalogue the antimicrobial peptides are hampered by the absence of a systematic 2o terminology and by the fact that peptides clearly belonging to the same structurally family have been given different names. As shown in Table 1, Summary of the Invention, at least 7 different peptide families produced in the skins of Ranid frogs have been identified on the basis of amino acid sequence similarity. For the present invention, these families are named from the first 25 member to have been discovered as follows: {1) ranatuerin-1, (2) ranatuerin-which includes ranatuerin-3, (3) brevinin-1 which includes gaegurin-5 and -6, and ranatuerin-4, (4) brevinin-2 which includes rugosin A, and B and ranatuerin-IT, {5) esculentin-2 which includes gaegurin-1, -2, -3 and -4 and rugosin C, (6) ranalexin and (7) temporin which includes peptides A1 and B9 and ranatuerins -30 5, -6, -7, -8 and -9.
il The C-terminal region of peptides of the brevinin-1, ranalexin, brevinin-2, esculentin-1, esculentin-2 and ranatuerin-1 families contains a cystine-bridged cyclic lieptapeptide whereas the corresponding region of peptides of the ranatuerin-2 family contains a cystine-bridged cyclic hexapeptide. The temporin s family peptides lack the: cyclic region and their primary structures have been poorly conserved. All members of the family contain between 10-14 amino acid residues and terminate in a C-terminally (a-amidated amino acid residue.
The present invention establishes that extracts of the skins of six closely-related North American frogs of the genus Rana contain antimicrobial peptides to belonging to 5 of the known families: brevinin-1, ranatuerin-2, esculentin-2, ranalexin and temporin. These peptides classified by using the initial letter of the species name to indicate their origin (L for luteiventris, B for berlandieri and P
for pipiens, C for clamitans and G for grylio. The catesbeiana peptides are regarded as the "parent" and so are not given a letter.) Where two or more is members of the same family are produced by the one species, the peptides are differentiated by the letters a, b, c, etc. as in the examples of ranatuerin-2La and ranatuerin-2Lb.
The distribution of the molecular forms of the biological active peptides and the primary structures of the individual peptides are highly species-specific.
2o Each frog in this study produced multiple peptides of the brevinin-1 family and at least one peptide of the esculetin-2 and ranatuerin-2 families but a peptide of the temporin family was not detected in R. berlandieri skin despite a systematic search for such a component. Similarly, peptides that are structurally related to ranalexin and ranatuerin-1, previously isolated from the North American 2s bullfrog, R. catesbeiana, were not identified in the skin extracts of the other species. At this time, peptides belonging to the esculentin-1 and brevinin-2 families have not been identified in the skins of North American Rapid frogs.
None of the antimicrobia:l peptides isolated in this study have been described previously.

It has been proposed that the synthesis of multiple structurally related peptides by the skin of an amphibian species serves to protect the organism against invasion by a broad array of microorganisms because each peptide has a discrete spectrum of antimicrobial activity and may target specific bacteria.
The peptides of the brevinin-1, esculentin-2 and ranatuerin-2 families isolated in the present study show a broad spectrum of antimicrobial activity inhibiting the growth of a gram positive bacterium (Staphylococcus aureus), a gram negative bacterium (Escherichia coli) and a yeast (Candida albicans). However, of the microorganisms tested, the peptides of the temporin family were active only against S. aureus.
As shown in Table 1, Summary of the Invention, ranatuerin 4 contains limited structural similarity, including the presence of a C-terminal heptapeptide ring, to the previously characterized gaegurins (Park, J.M., Jung, J.E., and Lee, B.J., Biochem. Biophys. Res. Commun. 205, 948-954 (1994)); and brevinins (Morikawa, N., Hagiwara, K., and Nakajima, T., Biochem. Biophys. Res.
Commun. 189, 184-I90 (1992); Simmaco, M., Mignogna, G., Barra, D., and Bossa, F., J. Biol. Chem,. 269, 11956-11961 (1994)) also isolated from the skin of other Rapid frogs. Ranatuenins 5 - 9 show some sequence identity to the hemolytic peptides A1 and B9 previously isolated from the skin of Rana 2o esculenta (Simmaco, M., De Biase, D., Severinin, C., Aita, M., Falconieri Erspamer, G., Barra, D., and Bossa, F., Biochim. Biophys. Acta 1033, 318-323 (1990)). Ranatuerins 1, :2, and 3, however, do not resemble closely any other anti-microbial peptides isolated from amphibian skin.
The amino acid sequence motif C-K-(V/I/I,]-(A/S/T]_K-[K/T/Q]-C, asserted for all antimicrobial peptides from species of the genus Rana without exception (Park, J.M., Jung, J.E., and Lee, B.J., Biochem. Biophys. Rer.
Commun. 205, 948-954 (1994)), is absent from the many of the Rana peptides of the present invention. A :further unexpected feature of the present invention is that raualexin, previously isolated from an extract of whole bullfrog tadpoles (Clark, D.P., Durell, S., Maloy, W.L., and Zasloff, M., J. Biol. Chem. 269, 10849-10855 (1994)), is not present among the ranatuerins. Since ranalexin shows potent cytolytic activity towards S, aureus {minimal inhibitory concentration of 4 ~g/ml), it would not have been missed during the investigation of the bullfrog skin extracts. It is probable, therefore, that the expression of the antimicrobial peptides in bullfrog skin is developmentally regulated.
Many of the Rana peptides show high potency and a broad spectrum of antimicrobial properties against the gram positive, gram negative bacteria and against the fungi, see the tables and figures. Thus, the amino acid sequence of the potent Rang peptides can also form the basis for modified versions exhibiting higher potency and with specificity towards pathogenic microorganisms that have developed resistance to commonly used antibiotics.
In particular, one modification focuses upon the presence of a cystine bridge at the C-terminal ~end of the brevinin-1, ranalexin, brevinin-2, esculentin-1, esculentin-2 and ranatuerin-1 families and the ranatuerin-2 family.
Synthesis of modified Rang peptides containing the substitutions Cys -> Ala and Cys -~
Ser provides peptides that will have no cysteine bridges. These modified Rana peptides will show inhibitor activity against Escherichia coli, Staphylococcus aureus and Candida albicans.
2o In general, other modified ranatuerins can be formulated through an understanding of the mechanism by which antimicrobial peptides are cytotoxic.
Antimicrobial peptides such as the Rana peptides have a propensity to form amphipathic a-helical structures and their mechanism of action is believed to involve a direct interaction with the fatty aryl chains in the cell membrane 25 leading to disruption of normal membrane function responsible for osmotic balance. The presence of cationic residues (particularly lysine) in antimicrobial peptides is believed to destroy the ionic gradient across cell membranes by forming ion channels. Cruciani, R.A., Barker, J.L., Durell, S.R, Raghunathan, G., Guy, H., Zasloff, M., and Stanley, F., Eur. J. Pharmacol. 226, 287-296 30 (1992). Analysis of the secondary structure of a Rana peptide, either by the method of Gamier, Osguthorpe and Robson or by the method of Qian and Sejnowski, predicts that most of the peptides contain two a-helical regions joined by a glycine-containing "hinge" region. Gamier, J., Osguthorpe, D., and Robson, B., J.MoI. Biol'. 120, 97-121 (1978); Qian, N., and Sejnowski, T., J.
5 Mol. Biol. 202, 865-88~ (1988)). This conformation is, for example, found in the cecropin family of antimicrobial peptides isolated from insect hemolymph and from mammalian intestine.
Gamier, Osguthorpe and Robson to SMLSVLKNLGKVGLGFVACKINKQC (sEQ m rro: 62) Qian and Sejnowski SMLSVLKNLGKVGLGFVACKINKQC (sEQ m No: 63) __~_____-__~ ______ A a-helix; B (i-sheet; T p-turn; - random coil Based upon this understanding, one generation of modified Rana peptides is constructed by substituting one or more of the a-helix-extending residues (hydrogen bonding residues) for the lipophilic amino acid residues within the helix regions of an individual Rana peptide, especially those within the C-terminus region of the peptides. Other amino acid residues can be substituted as follows:
(a) Ala is substituted for Gly in order to replace the p-sheet region in the central portion of the peptide with an extended a-helix. The increased a-helical character of the peptide promotes interaction with the cell 3o membrane of the targeted microorganism;
(b) Val is substituted for Gly in order to stabilize the p-sheet region in the central portion of the peptide;
(c) Ala or another lipophilic amino acid residue is substituted for Asn in order to promote a-helical character;
1s (d) Cilu is substituted for Gly in order to promote a-helical character.
Modified Rana peptides also include those Rana peptide Family modified peptides with the formulas containing the symbols X, B, Z, U, O, J' and J as discussed in the Summary of the Invention.
Still fizrther modified Rana peptides can also prepared by substituting one or more of the lysine residues by arginine. This substitution will increase the cationic character of the peptide so as to promote its ability to affect the cellular membrane ionic gradient.
1 o Another generation of modified Rana peptides includes truncated Rana peptides lacking redundant segments of the original molecules, and protease-resistant modified Rana peptides containing D-amino acid residues and/or unnatural amino acids and/or side chain modified amino acids. The Rang peptides can also be modified by substitution of one or more of the amino acid 15 residues valine (V), alanine (A), lysine (K), and glutamic acid (E) for the glycine (G) residues. Further modified Rana peptides include those wherein arginine (R) is substituted for one or more of the lysine (K) residues. The truncated versions of Rana peptides include the fi~agments cleaved on the N side of the any of the glycine residues or between repeat residues as well as N-terminally deleted 2o fragments. Any of the Rana peptides can be modified with amino acid residues having modified side chains such as those in which the epsilon amino group of a lysine (K) residue has been coupled to a fatty acid such a pahnitate or glycosylated sugar, those in which the epsilon carboxy group of aspartic (D) or glutamic (E) acid residues or a hydrolyzed glutamine (~ residue have been 25 coupled to fatty alcohols or glycosylated sugars, or those in which the epsilon hydroxyl group of threon.ine (T), tyrosine (~, or serine (S) residues have been coupled to fatty acids ar glycosylated sugars. In addition, the modified Rang peptides include those in which a truncated version is synthetically condensed with a non-natural peptide or peptide sequence optionally containing 3o heterocyclic organic moieties such as a beta or gamma amino acid, an aliphatic diamine, an aliphatic or aromatic dicarboxylic acid, pyridine carboxylic acid, aromatic diols and the like.
The Rang. peptides and modified versions can be obtained by automated peptide synthesis using such techniques as the "Men-ifeld" technique, the 5 recombinant DNA techniques involving insertion, expression and isolation, and by extraction techniques from any source in nature (for naturally occurring Rana peptides). In addition, semi-synthetic Rana peptides can be obtained by preparation of the peptide sequence using any of the foregoing techniques and coupling the peptide sequence to the chemical moiety by esterification, to amidation, Schif~base fbnmation under appropriate side chain protection if needed. These techniques are well known in the art. For example, a Merrifeld technique is described v1 US Patent No. 5,049,656, the disclosure of which is incorporated herein by reference, and a recombinant technique is described at U.S. Patent Nos. 4,237, 224 and 5,595,887, the disclosures of which are 15 incorporated herein by reference. Semisynthetic coupling techniques as well as amidation and esterification of carboxylic acids (eg., for the formation of the C-terminus amides and esters) are described in "Advanced Organic Chemistry", J.
March, Wiley Interscience 4th Ed., 1992. An example of an extraction technique is set forth in the following experimental example.
2o In regard to the recombinant techniques, the Rana peptides are present in Rapid frog skin in very high abundance and so it is not envisaged that cloning of the cDNAs encoding thE; peptides will present a particularly difficult challenge.
Standard cloning techniques can be used. Poly(A)-rich RNA from bullfrog skin can be prepared by affinity chromatography and a constructed cDNA library. A
25 pool of mixed nucleotides encoding appropriate regions of the Rang peptide DNA sequence such as the 19-25 region of ranatuerin 1 or analogous regions of the other Rang peptide DNA sequences can be synthesized for use as a primer in the RACE (rapid amplification of cDNA 3' end) protocol using the polymerase chain reaction. Amplification products can be cloned into the XhoUEcoRV
30 restriction site of the BlueScript vector (Stratagene). The clone can be labeled by random priming and then used to screen the cDNA to obtain a full-length clone. Positive clones can be selected and analyzed by nucleotide sequencing.
This approach has been used successfully to clone the cDNAs encoding esculentin and brevinin lE from the frog Rana esculenta (J. Biol. Chem.
5 269:11956-11961, 1994). The cDNA clone containing the genome for ranatuerin or a modified version thereof can be expressed by known recombinant techniques such as by insertion and, expression in E Coli. Since the Rana peptides are antimicrobial, a preferred technique will involve triggering the culture to express after ~.t has reached maturity.
1o For effective prophylactic and anti-infectious use, the Rana peptides and their modified and truncated forms as well as the compositions of the present invention may be administered either alone or in combination with a pharmaceutically-acceptable carrier, by topical, oral, anal, ocular, buccal, nasal, intramuscular, subcutaneous, intravenous, or parenteral routes. The ultimate 15 choice of route, formulation and dose is made by the attending physician or veterinarian and is based upon the patient's or animal's unique condition.
However, the usual dosage for administration to humans lies in the range of approximately 50-2000 mg. P.O. per day, and preferably in about one to four doses where the dose is based upon the activity of pure Rang peptide. The usual 2o dosage for administration to small animals lies in the same approximate range as that for humans. For large animals, the usual dosage is higher per unit of body weight so that the dose given lies in the range of about 20 to 20,000 mg. P.O.
per day. This dosage may vary somewhat with the weight of the subject (human or animal) being treated; in general, about 1-40 mg:/kg. of body weight per day can 25 be employed for humans and small animals while about 1 to 400 mg./kg. of body weight per day can be employed for large animals.
The Rana peptides of this invention can be combined with inert phamia~ceutical excipients such as lactose, oil, mannitol and starch, and formulated into dosage forms such as elixirs, liquids, ointments, lotions, IV
3o fluids, alcohol, tablets, capsules, and the like. For parenteral, intramuscular, subcutaneous and intravenous administration, these peptides can be formulated with an inert, parenterally acceptable vehicle such as water, saline, sesame oil, ethanol buffered aqueous medium, propylene glycol and the like. For topical and oral administration, these peptides can be formulated with waxes, oils, buffered aqueous medium, and the like. These various pharmaceutical dosage forms are compounded by methods well known to the pharmacist's art.
Following the foregoing regimen, the antibacterial activity of the Rana peptides as well as the modified and truncated forms thereof likely will show inhibition action against the following strains of bacteria and fungi.
1. Escherichia coli Drug-resistant strains of this "harmless" organism are responsible for twice as many cases of :nosocomial infections as any other microbe, particularly hospital-acquired urinary tract infections.
2. Enterococcus sp.
These organisms are responsible for infection of wounds especially in elderly patients undergoing surgery. E, faecalis is also responsible for urinary tract infections and frequently invades heart valves and prosthetic devices of 2o hospitalized patients.
3. Bactereoides fragilis Although this Gram-Negative anaerobe is a component of the normal flora of the gut, resistant strains are responsible for wound and abdominal infections in hospitalized patients.
4. Pseudomonas aeruginosa Infections that may lead to septicemia and pneumonia, are especially common in burn patients.. The organism is frequently introduced into body orifices by contaminated catheters and other invasive instruments and proliferates in respirators and renal dialysis units S. Klebsiella pneumoniae The organism readily grows in glucose solutions used for intravenous therapy leading to deaths from bacteremia. Pneumonia caused by Klebsiella has a 50% mortality rate in spite of treatment with existing antibiotics.
6. Serratia marcescens Like Klebsiella, this Gram-Negative organism grows in glucose solutions and infection leads to colonization of the oropharynx of chronically ill persons leading to pneumonia.
7. Mycobacterium tuberculosis Since 1985, the incidence of tuberculosis has dramatically increased in the U.S.A, due to the emergence of strains resistant to Isoniazid and ltifampin, the major drugs used in treatment. Hospital epidemics in AIDS and other immunosuppressed patients are common.
8. Streptococcus pneumoniae The organism is responsible for 80% of all bacterial pneumonias and is particularly prevalent in institutions housing old people such as nursing homes and geriatric wards. In young people infection of the middle ear leads to meningitis and otitis media.
9. Streptococcus pyrogenes The organism responsible for "strep throat" which, if improperly treated, can lead to complications of rheumatic heart disease, pneumonia and glomerulonephritis.
10. Xaemophilus influenzae This pathogen is responsible for the increasing incidence of acute bacterial meningitis in children and is often transmitted in day-care settings.
Even with prompt diagnosis and aggressive treatment with existing antibiotics, 33% of children sustain residual disability.
11. Staphylococcus saprophyticus Staphylococcus sp. in hospitals is generally antibiotic resistant and poses a particular problem in nurseries, delivery rooms and burn units. S.
1o saprophyticus causes urinary-tract infections particularly in sexually active young women and in immunosuppressed patients.
12. Candida albicans Antibiotic therapy or severe immunosuppression often precedes 15 extensive invasion by C: albicans and is particularly common in burn patients and in recipients of renal, heart and bone marrow transplants.
The following experimental examples further illustrate the invention.
They are not meant to provide limitations of the invention that has been fully 2o characterized by the foregoing discussion. The foregoing specification, examples and data provide a complete description of the preparation and use of the composition of the invention. Since many embodiments of the invention can be made without departing from the spirit and scope of the invention, the invention resides in the claims hereinafter appended.
1~ZAT'ERIALS AND METHODS
Taxonomic identification of individual specimens of frogs of the genus Rana is often difficult especially in regions where several species coexist and produce hybrids. Similarly, the phylogenetic interrelationships between the 3o different species are incompletely understood. It is suggested, therefore, that study of the distribution and amino acid sequences of antimicrobial peptides in skin may prove to be a valuable tool for the identification and taxonomic classification of Rapid frogs. Once a larger data set of primary structures has been compiled, the amino acid sequences may form a basis for cladistic analysis 5 to investigate phylogenetic interrelationships between species. Skin secretions, containing very high concentrations of antimicrobial peptides, can easily be collected from Rapid frogs by non-invasive procedures (mild electrical stimulation or subcutaneous injection of epinephrine) so that identification of the species does not necessitate sacrifice of the animal. Molecular techniques have i0 been used previously to facilitate identification and to study phylogenetic relationships among Rapid frogs. For example, analysis of the nucleotide sequences of mitochondt~ial and genomic DNA has been used to investigate the evolutionary history of specimens of the closely related species of European water frogs, R. ridibunda, R. esculenta and R. lessonae.
Animals Adult specimens of both sexes of the spotted frog, R. luteiventris (n =16;
wt range 12 - 40 g), the Rio Grande leopard frog, R. berlandieri (n = 8; wt range 51 - 115 g}, the North American bull frog R. catesbeiana, R. clamitans (n=21;
wt 20 range 44-75 g), R. grylio (n=6; wt range 106-204g) and the northern leopard frog, R. pipiens (n =12; wt range 41 - 54 g) were purchased from Charles D.
Sullivan Company (Nashville, TN, U.S.A.). The animals were anaesthetized by immersion in crushed ice and were sacrificed by pithing. Skin was immediately removed, frozen on dry ice and stored at -SS°C until time of extraction.
Tissue Extraction The frozen skins (R. luteiventris 45 g; R. berlandieri 66 g; R. clamitans 125g; R. grylio; 96 g; R. pipiens 58 g) were separately extracted by homogenization in ethanol/0.7 M HCl (3:1 v/v; 10 ml/g ) at 0°C using a blaring 3o blender. The homogenates were stirred for 2 h at 0°C and centrifuged (4000 x g for 30 min at 4°C). Ethanol was removed from the supernatants under reduced pressure and, after further centrifugation (4000 x g for 30 min at 4°C), the extracts were separately pumped onto 8 Sep-Pak C-18 cartridges (Waters Associates, Milford, MA, U.S.A.) connected in series at a flow rate of 2 mUmin.
Bound material was eluted with acetonitrile/water/trifluoroacetic acid (70.0:29.9:0.1, v/v/v) and freeze-dried.
For the R. catesbeiana the tissue extraction was as follows. Skin (176 g) was removed from pithed adult specimens of R. catesbeiana of both sexes (n =
6) and immediately frozen on dry-ice. The tissue was stored at -55°C
until time to of extraction. The tissuc; was extracted by homogenization in ethanol/0.7 M
HCl (3:1 v/v; 1800 ml) at 0 "C: using a blaring blender. The homogenate was stirred for 2 h at 0 °C and cenhifuged (4000 x g for 30 min at 4 °C).
Ethanol was removed from the supernatant under reduced pressure and, after further centrifugation (4000 x ~; for 30 min at 4 °C), the extract was pumped onto 8 Sep-is Pak C-18 cartridges (Waters Associates) connected in series at a flow rate of 2 ml/min. Bound material was eluted with acetonitrile/ water/ trifluoroacetic acid ((70.0:29.9:0.1, v/v/v) and freeze-dried.
Peptide Purification.
2o The same chromatographic procedures were used to purify antimicmbial peptides from each of the species R. luteiventris, R. berlandieri, R.
clamitans, R.
grylio and R. pipiens so only the methods used to isolate the R. luteiventris peptides will be described in detail. The skin extract, after partial purification on Sep-Pak cartridges, was redissolved in 1 % (v/v) trifluoroacetic acid/water (5 mI) 25 and chromatographed on a (100 x 2.6 cm) column of Sephadex G-25 (Pharmacia Biotech, Uppsala, Sweden) equilibrated with 1 M acetic acid. The column was eluted at a flow rate of 48 ml/h and fractions (8 ml) were collected.
Absorbance was measured at 280 nm. The ability of aliquots (50 ~1) of the fi~actions to inhibit the growth of S. aureus was determined as described in the previous 30 section: Fractions containing maximum activity (denoted by the bars in Fig.
1) were pooled and injected onto a (25 x 1 cm) Vydac 218TP510 C-18 reverse-phase HPLC column (Separations Group, Hesperia, CA, USA) equilibrated with 0.1 % (v/v) trifluoroacetic acid/water at a flow rate of 2 mUmin. The concentration of acetonitrile in the eluting solvent was raised to 21 % (v/v) over 10 rnin and to 49% (v/v) over 60 min using linear gradients. Absorbance was monitored at 214 nm and 280 nm and fractions (1 min) were collected. The fractions containing antimicrobial activity were successively rechromatographed on (250 x 4.6 mm) Vydac 214TP54 (C-4) and Vydac 219TP54 (phenyl) reverse-phase HPLC columns. ~~Che concentration of acetonitrile in the eluting solvent to was raised from 21% to 55% over 40 min and the flow rate was 1.5 ml/min.
The purification of the R. catesbeiana frog skin extract was accomplished in similar fashion. After partial purification on Sep-Pak cartridges, the extract was redissolved in 1% (v/v) trifluoroacetic acidlwater (5 mI) and chromatographed on a (100 x 2.6 cm) column of Sephadex G-25 (Pharmacia Biotech) equilibrated with 1 M acetic acid. The column was eluted at a flow rate of 48 ml/h and fractions (8 ml) were collected. Absorbance was measured at 280 nm. The antimicrobial activity of aliquots of the fractions was determined. Fractions containing maximum activity (denoted by the bars in Fig.
1) were pooled and injected onto a (25 x 1 cm) Vydac 218TP510 C-18 reversed-2o phase HPLC column (Separations Group) equilibrated with 0.1% (v/v) trifluoroacetic acid/wate~- at a flow rate of 2 ml/min. The concentration of acetonitrile in the eluting solvent was raised to 21% (v/v) over 10 min and to 49% (v/v) over 60 min using linear gradients. Absorbance was monitored at 214 nm and 280 nm and fractions (1 min) were collected. The fractions containing 25 antimicrobial activity were successively rechromatographed on (250 x 4.6 mm) Vydac 219TP54 (phenyl;> and Vydac 208TP54 (C-8) columns. The concentration of acetonitrile in the eluting solvent was raised from 14% to 35% over 40 min and the flow rate was 1.5 mUmin.

Separation of R. luteiventris peptides The peptide fractions in the extract of R. luteiventris skin showing growth-inhibiting activity against S. aureus, after partial purification on Sep-Pak cartridges, were eluted fram a Sephadex G-25 gel permeation column as two 5 discrete zones, designated by the bars in Fig. 1. Subsequent characterization studies demonstrated that zone A contained esculentin-2L, ranatuerin-2La, and ranatuerin-2Lb and zone B contained brevinin-lLa, brevinin-lLb, temporin-lLa, temporin-1Lb and temporin-lLc. After chromatography of pooled fi~actions from zone A on a semipreparative Vydac C-18 reverse-phase HPLC column (Fig. 2A), the antimicrobial activity was eluted in a single, but very broad region, of the chromatogram with retention time between 45 and 70 min.
Chromatography of pooled fractions from zone B (Fig. 1) on the semipreparative C-18 column resulted in the elution of the antimicrobial activity in overlapping fractions with retention times between 48 an 75 min (Fig. 2B).
15 The same chromatographic procedures were used to purify all the biologically active peptides and so only the purification of those finm zone A
(Fig. 1) will be described in detail. Fractions with retention times between and 55 min from the semipreparative C-18 column (Fig. 2A) were pooled and chromatographed on a Vydac C-4 column (Fig. 3A). Although resolution was 2o poor, antimicrobial activity (associated with ranatuerin 2La) was eluted in the fractions denoted by the bar. Fractions with retention times between 56 and 70 min from the C-18 column (Fig. 2A) were pooled and chromatographed on a Vydac C-4 column (Fig. 3B). Antimicrobial activity was eluted in two discrete zones denoted by the bars. The earlier eluting fi~actions contained ranatuerin--25 2Lb and the later eluting fractions contained esculetin-2La. The antimicrobial peptides were purified to near homogeneity, as assessed by symmetrical peak shape, by a final chromatography on a Vydac phenyl column as shown in Figs. 4 (A- C). The final yields of pure peptides were: ranatuerin 2La, 4050 nmol;
ranatuerin 2Lb, 320 nmol; and esculentin-2L, 740 nmol.

WO 00/09553 PCT/US99/185'75 Purification of antimicrobial peptide from zone B from gel permeation chromatography (Fig. 1 ) on Vydac C-4 and Vydac phenyl columns resulted in the isolation in pure form of brevinin-1La (880 nmol), brevinin-llb (45 nmol), temporin-1La (120 nmol), temporin-1Lb (200 nmol) and temporin-1Lc (60 mnol). The final yields of the pure peptides are shown in parentheses.
Separation of R berlandieri, R clamitans, R grylio and R pipiens peptides Using the same chromatographic procedures for separation of antimicrobial peptides fi~om R. luteiventris led to the isolation in pure form of the to following peptides from R. berlandieri skin in the yields indicated:
brevinin-1Ba (4 nmol), brevinin-1Bb (15 nmol), brevinin-1Bc (8 nmol), brevinin-1Bd (28 nmol), brevinin-1Be (12 nmol), brevinin-1Bf (9 nmol), esculentin-2B (31 nmol), and ranatuerin-2B (75 nmol). The following peptides with antimicrobial activity were isolated from R. pipiens skin in the yields indicated: brevinin-1Pa (1050 15 nmol), brevinin-1Pb (1110 nmol), brevinin-1Pc (64 nmol), brevinin-1Pd (315 nmol), brevinin-IPe (8 mnol), esculentin-2P (12 nmol), ranatuerin-2P (690 nmol), and temporin-1P (205 nmol). The following peptides were isolated from R. clamitans skin in the yields indicated: ranatuerin-1C (570nmo1), ranalexin-(715 nmol), ranatuerin-2Ca (30 nmol), ranatuerin-2Cb (110 nmol), Temporin-20 1Ca (240 nmol), temporin-1Cb (qnm), temporin-1Cc (qnm). The following peptides were isolated from R. grylio skin in the yields indicated: ranalexin-(50 nmol), ranatuerin-1G (70 nmol), ranatuerin-2G (55 nmol), and temporin-1Ga (180 nmol). (qnm mean,; quantity not measured.) 25 Separation of R catesbeiana peptides The peptide fractions in the extract of R, catesbeiana skin, after partial purification on Sep-Pak cartridges, were eluted from a Sephadex G-25 gel permeation column as two zones ( Fig. 5). After chromatography of pooled fractions from the earlier-eluting zone A on a semipreparative Vydac C-18 3o reversed-phase HPLC co'.lumn, the activity was eluted in three non-overlapping 2b fractions that were subsequently shown to contain ranatuerins 1+ 5, ranatuerins 2+3 and ranatuerin 4 {F'ig. 6A). Similarly, chromatography of pooled fraction from zone B led to the separation of activity into three fractions subsequently shown to contain ranatuerins 6+ 7, ranatuerin 8 and rantuerin 9 (Fig. 6B). The peptides were purified to near homogeneity, as assessed by a symmetrical peak shape, by chromatography on analytical Vydac phenyl and C-8 columns. The approximate yields of pure peptides were: ranatuerin 1, 120 nmol; ranatuerin 2, 80 nmol; ranatuerin 3, 50 nmol; ranatuerin 4, 160 nmol; ranatuerin 5, 140 nmol;
ranatuerin 6, 170 nmol; ranatuerin 7, 250 nmol; ranatuerin 8, 120 nmol and 1o ranatuerin 9, 400 nmol.
Structural analysis.
Amino acid compositions were determined by precolumn derivatization with 6-aminoquinolyl-N-hydroxysuccinimidyl carbamate using a Waters AccQ
15 Tag system with fluorescence detection and separation of the amino acid derivatives by reverse-phase HPLC. Hydrolysis in 5.7 M HCl (24 h at 110°C) of approximately 1 nmol of peptide was carried out. The primary structures of the peptides were determined by automated Edman degradation using an Applied Biosystems model 471 A sequenator modified for on-line detection of 2o phenylthiohydantoin amino acids under gradient elution conditions.
Electrospray mass spectrometry was carried out using a Perkin Elmer Sciex API
150EX single quadrupole instrument. The accuracy of mass determinations was f 0.02%.
The primary structures of the peptides their amino acid sequences are 25 shown in Table 1, Summary of the Invention. In all cases, the proposed amino acid sequences, including the presence of a cystine bridge in the brevinin-1, esculentin-2 and ranatuerin-2 peptides were confirmed by amino acid composition analysis (data not shown) and mass spectrometry (Fig. 5). Mass spectrometry also demonstrated that the peptides of the temporin family 3o contained a C-terminally «-amidated residue.

WO 00/09553 PCT/US99/i8575 Antimicrobial assays Minimal inhibitory concentrations (MICs) of the peptides were determined by a standard microdilution method using 96-well microliter cell s culture plates as previously described. Serial dilutions of the peptides in Mueller-Hinton broth (50 ~L) were incubated with an inoculum (50 wL of 103 CFU/mL) from an overnight culture of Escherichia coli (ATCC 25922) and Staphylococcus aureus (NCTC 8325), for 18 h at 37°C in an humidified atmosphere of 5% C02 in air. Incubations with Candida albicans (ATCC 90028) io were earned out in RPMI 1640 medium for 48 h at 35°C. After incubation, the absorbance at 550 nm of each well was determined using a M.A. Bioproducts model MA308 microliter plate reader. The MIC of each peptide was taken as the lowest concentration where no visible growth was observed. In order to monitor the validity of the assay, incubations with E. coli and S. aureus were carried out 15 in parallel with increasing concentrations of the broad-spectrum antibiotic, bacitracin and incubations with C albicans in parallel with amphotercin B.
The abilities of the isolated peptides to inhibit the growth of the gram-positive bacterium Staphylococcus aureus, the gram-negative bacterium Escherichia coli and the: yeast, Candida albicans are compared in Table 2 (NA
2o in Table 2 means no activity; ND means not determined.) Ranatuerins 1-9, tested at a concentration of 20 ~,g/ml, showed no detectable hemolytic activity towards human erythrocytes.

2s MIC (N,~
S. aureus E. coli C. Albicans Itana catesbeiana Ranatuerin 1 SO 2 70 Ranatuerin 2 60 60 NA

WO 00/09553 PCT/tJS99/18575 Ranatuerin 60 70 NA

Ranatuerin 55 ND NA

Ranatuerin >200 ND NA

Ranatuerin 100 ND NA

Ranatuerin 200 ND NA

Ranatuerin 130 ND NA

Ranatuerin 130 ND NA

Rana luteiventris Brevinin-1Lb $ 16 ND

Esculetin-2L 3 6 53 Ranatuerin-2La I1 4 >150 Ranatuerin-2Lb 4 4 62 Temporin-1La 60 ND

Temporin-1Lb 48 >150 >150 Temporin-1Lc 125 >150 >I50 Rana berlandieri Brevinin-1Ba 2 ND I~

Brevinin-1Bb 1 3 10 Brevinin-1Bc 13 ND N~

Brevinin-1Bd 3 7 7 Brevinin-1Be 3 15 ND

Brevinin-1Bf 8 19 ND

Esculetin-2B 1 1 29 Ranatuerin-2B 1 1 35 Rana pipiens Brevinin-Ipa 7 14 Brevinin-1Pb S 14 7 Brevinin-1Pc 7 $

Brevinin-1Pd 27 78 29 Esculetin-2P ND 10 ND

Ranatuerin-2P 50 13 67 Temporin-1P 110 ND ND

Rana clamitans Ranatuerin-1C 55 1.5 58 Ranalexin-1C 17 4 14 Ranatuerin-2Cb 40 2 4b Temporin-1Ca 100 NA NA

Temporin-1Cb 160 NA NA

Temporin-1Cc 100 NA NA

Rana grylio Ranatuerin-1G 60 3.5 110 Ranatuerin-2G 150 19 >150 Ranalexin-1G 18 9 Temporin-1 Ga 125 NA NA

Temporin-1Gb 55 NA NA

Structure-Activity Studv of ranatuerin-1 5 Substitutions in the C-terminal domain of the molecule that increase the alpha-helical character are studied. Many increase potency towards grarn-negative bacteria.
Ranatuerin-1 (SMLSVLKNLG'°KVGLGLVACKz°INKQC) (SEQ ID
NO: 8), isolated from the skin of the American bullfrog Rana catesbeiana, shows to antimicrobial activity against the gram-negative bacterium, Escherichia coli, the gram-positive bacterium Staphylococcus aureus, and the yeast, Candida albicans.
The peptide is predicted to comprise three sizuctural domains: alpha-helix (residues 1- 8), beta-sheet (residues 11- 16) and beta-turn (residues 20-25).
Substitution of Cysl9 and Cys25 by Ser residues had only a minor effect on 15 potency demonstrating that the disulfide bridge is not necessary for activity. but deletion of the cyclic heptapeptide region produced an inactive analog.
Substitution of G1y10, G1y13 and G1y15 by either Lys or Glu residues produced analogs with increased a-helical content but with decreased or absent antimicrobial activity peptide thus demonsixating the importance of the central 2o beta-sheet region of the 1>eptide.
Substitution of Asn22 in the beta-turn region by Ala resulted in an increase in predicted alpha-helical character and an 8-fold increase in potency against E. coli.

In contrast, substitution of Ser4 in the N-terminal a-helical region by either Ala or Gly resulted in relatively minor effects on potency but deletion of residues (1-8) produced an inactive analog.
Substitution of'Lys7 and Lysl 1 by Arg produced analogs with increased positive charge but with decreased potency. The data demonstrate that the full sequence of the peptide is necessary for biological activity and that an increase in the alpha-helical content of the C-terminal region results in an increase in activity towards a gram-negative bacterium.
io Ranatuerin Analog Synthesis All peptides were synthesized by solid-phase methodology on a 0.025 mmol scale on an Applied Biosystems model 432A peptide synthesizer using a 4-(2,4-dimethoxy-phenyl-Fmoc-aminomethyl)phenoxyacetamido-ethyl resin (Perkin Eliner, Foster Cil:y, CA). Fmoc amino acid derivatives were activated with O-benzotriazol-1-yl-N,N,N,N-tetramethyluronium hexafluorophosphate (1-equivalent), 1-hydroxybenzotriazole hydrate (lequivalent) and diisopropylethylamine (2 equivalents) . Deprotection of the N-terminus by piperidine was monitored by on-line measurement of the conductance of the carbamate salt of the Fmoc group and optimum coupling times were determined 2o by the instrument in response to the deprotection times. The peptide was cleaved from the resin with trifluaroacetic acid/water/thioanisole/1,2-ethanedithiol (90.0/5.0/2.5/2.5) at 250 for 3 hr.
The crude synthetic peptides were purified to near homogeneity by chromatography on a 1 x. 25-cm Vydac 218TP510 C-18 reversed-phase HPLC
column (Separations Group, Hesperia, CA) equilibrated with 20 acetonitrile/water/ trifluoroacetic acid at a flow rate of 2 ml/min. The concentration of acetonitrile in the eluting solvent was raised to 49% over 60 min using linear gradients. Absorbance was measured at 214 and 280 nm and the major peak in the chromatogram was collected by hand.

The synthetic peptides were characterized by automated Edman degradation using an Applied Biosystems model 471A sequenator modified for on-line detection of phenylthiohydantoin amino acids under gradient elution conditions and by electrospray mass spectrometry using a Perkin Elmer Sciex API 1 SOEX single quadrupole instrument. The accuracy of mass determinations was B 1 0.02%.
Andmicrobial assays Minimal inhibitory concentrations (NIICs) of the peptides were 1o determined by a standard microdilution method using 96-well microtiter cell-culture plates as previously described. Serial dilutions of the peptides in Mueller-Hinton broth (50 mL) were incubated with an inoculum (SO mL of 103 CFU/mL) from an overnight culture of Escherichia coli (ATCC 25922) and Staphylococcus aureus (NCTC 8325), for 18 h at 37 OC in a humidified atmosphere of 5% C02 in air. Incubations with Candida albicans (ATCC 90028) were carried out in RPMI 1640 medium for 48 h at 35 OC. After incubation, the absorbance at 550 nm of each well was determined using a M.A. Bioproducts model MA308 microtiter plate reader. The MIC of each peptide was taken as the lowest concentration where no visible growth was observed. In order to monitor the 2o validity of the assay, incubations with E. coli and S. aureus were corned out in parallel with increasing wncentrations of the broad-spectrum antibiotic, bacitracin and incubations with C. albicans in parallel with amphotercin B. The MIC's are presented in Table 3.

MIC values (p,m) Analog ~ E. coli ~ S. aureus ~ C. albicans Analog E. coli S. aureusC. albicans ~

itanatueru''1 3 50 70 [Serl9,Ser25]Ranatuerin-1 14 28 110 [Ala4, Serl9,Ser25]Ranatuerin-110 190 110 [Gly4, Serl9,Ser25]Ranatuerin-128 NA 140 [Arg7, Serl9,Ser25]Ranatuerin 14 190 190 [Argl l, Serl9,Ser25]Ranatuerin28 110 110 I

[Lysl3,Ser19,Ser25]Ranatuerein-1190 NA NA

[A1a22, Serl9,Ser25]Ranatuerin-11.5 200 110 [LyslO,Lysl3Lys15- 120 NA NA
Serl9,Ser25]Ranatuerin-1 [Glul0,G1u13,G1u15- NA NA NA
Serl9,SeflS]Ranatuerin-1 Ranatuerin-1(1-19)fragmdent NA NA NA

Ranatuerin-1(9-25)fragrraent NA NA NA

NA: no activity up to 200 mIVi SEQUENCE LISTING
<110> BioNebraska, Inc., et al.
<120> ANTIMICROBIAL PEPTIDES ISOLATED FROM THE
SKIN OF AMERICAtd FROGS
<130> 11B5.090W01 <140:> Unknown <141> 1999-08-13 <150> 60/096,607 <151> 1998-08-14 <160> 63 <170> FastSEQ for Windows Version 3.0 <210> 1 <211> 24 <212> PRT
<213> Rana rugosa <400> 1 Phe Leu Gly Ala Leu Phe Lys Val Ala Ser Lys Val Leu Pro Ser Val Lys Cys Ala Ile Thr Lys Lys Cys <210> 2 <211> 24 <212> PRT
<213> Rana rugosa <400> 2 Phe Leu Pro Leu Leu Ala Gly Leu Ala Ala Asn Phe Leu Pro Thr Ile Ile Cys Lys Ile Ser Tyr Lys Cys <210> 3 <211> 24 <212> PRT
<213> Rana brevipoda <400> 3 Phe Leu Pro Val Leu Ala Gly Ile Ala Ala Lys Val Val Pro Ala Leu Phe Cys Lys Ile Thr Lys Lys Cys <210> 4 <211> 24 <212> PRT
<213> Rana esculen.ta <400> 4 Phe Leu Pro Leu Leu Ala Gly Leu Ala Ala Asn Phe Leu Pro Lys Ile Phe Cys Lys Ile Thr Arg Lys Cys .20 <210> 5 <211> 13 <212> PRT
<213> Rana esculenta <400> 5 Phe Leu Pro Ala Ile Ala Gly Ile Leu Ser Gln Leu Phe <210> 6 <211> 13 <212> PRT
<213> Rana esculenta <400> 6 Phe Leu Pro Leu Ile Ala Gly Leu Leu Gly Lys Leu Phe <210> 7 <211> 20 <212> PRT
<213> Rana catesbe:iana <400> 7 Phe Leu Gly GIy Leu Ile Lys Ile Val Pro Ala Met Ile Cys Ala Val Thr Lys Lys Cys <210> 8 <211> 25 <212> PRT
<213> Rana catesbeiana <400> 8 Ser Met Leu Ser Val Leu Lys Asn Leu Gly Lys Val Gly Leu Gly Leu Val Ala Cys Lys Ile Asn Lys Gln Cys <210> 9 <211> 25 <212> PRT
<213> Rana grylio <400> 9 Ser Met Ile Ser Val Leu Lys Asn Leu Gly Lys Val Gly Leu Gly Phe Val Ala Cys Lys Val Asn Lye Gln Cys <210> 10 <211> 25 <212> PRT
<213s Rana clamitans <400> 10 Ser Met Leu Ser Val Leu Lys Asn Leu Gly Lys Val Gly Leu Gly Leu Val Ala Cys Lys Ile Asn Lys Gln Cys <210> 11 <211> 25 <212> PRT
<213> Rana catesbeiana <400> 11 Ser Met Xaa Ser Val Leu Lys Asn Leu Gly Lys Val Gly Leu Gly Xaa Val Ala Cys Lys Xaa Asn Lys Gln Cys <210> 12 <211> 30 <212> PRT
<213> Rana catesbeiana <400> 12 Gly Leu Phe Leu Asp Thr Leu Lys Gly Ala Ala Lys Asp Ala Gly Lys Leu Glu Gly Leu Lys Cys Lys Ile Thr Gly Cys Lys Leu Pro <210> 13 <211> 32 <212> PRT
<213> Rana catesbeiana <400> 13 Gly Phe Leu Asp Ile Ile Lys Asn Leu Gly Lys Thr Phe Ala Gly His Met Leu Asp Lys Ile Lys Cys Thr Ile Gly Thr Cys Pro Pro Ser Pro <210> 14 <211> 28 <212> PRT
<213> Rana pipiens~
<400> 14 Gly Leu Met Asp Thr Val Lys Asn Val Ala Lys Asn Leu Ala Gly His Met Leu Asp Lys Leu Lys Cys Lys Ile Thr Gly Cys <210> 15 <211> 28 <212> PRT
<213> Rana berlandieri <400> 15 Gly Leu Leu Asp Thr Ile Lys Gly Val Ala Lys Thr Val Ala Ala Ser Met Leu Asp Lys Leu Lye C'ys Lys Ile Ser Gly Cys <210> 16 <211> 31 <212> PRT
<213> Rana clamitans <400> 16 Gly Leu Phe Leu Asp Thr Leu Lys Gly Ala Ala Lys Asp Val Ala Gly Lys Leu Leu Glu Gly Leu Lys Cys Lys Ile Ala Gly Cys Lys Pro <210> 17 <211> 27 <212> PRT
<213> Rana clamitans <400> 17 Gly Leu Phe Leu Asp Thr Leu Lys Gly Leu Ala Gly Lys Leu Leu Gln Gly Leu Lys Cys Ile Lys Ala Gly Cys Lys Pro <210> 18 <211> 32 <212> PRT
<213> Rana luteiventris <400> 18 Gly Ile Leu Asp Ser Phe Lys Gly Val Ala Lys Gly Val Ala Lys Asp Leu Ala Gly Lys Leu Leu Asp Lys Leu Lys Cys Lys Ile Thr Gly Cys <210> 19 <211> 32 <212> PRT
<213> Rana luteiventris <400> 19 Gly Ile Leu Ser Ser Ile Lys Gly Val Ala Lys Gly Val Ala Lys Asn Val Ala Ala Gln Leu Leu Asp Thr Leu Lys Cys Lys Ile Thr Gly Cys <210> 20 <211> 31 <212> PRT
<213a Rana grylio <400> 20 Gly Leu Leu Leu Asp Thr Leu Lys Gly Ala Ala Lys Asp Ile Ala Gly Ile Ala Leu Glu Lys Leu Lys Cys Lys Ile Thr Gly Cys Lys Pro <210> 21 <211> 36 <212> PRT
<213 > Rana catesbe:iana <220>
<221> MUTAGEN

<222> 1....36 <223> Xaa = H = Any amino acid residue in same residue pasition of other family members arranged homologously <400> 21 Gly Xaa Xaa Xaa Xaa Xaa Xaa Lys Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Ala Xaa Xaa Xaa Xaa Xaa Xaa Xaa Lys Cys Xaa Xaa Xaa Xaa Cys Xaa Xaa Xaa <210> 22 <211> 37 <212> PRT
<213> Rana luteiventris <400> 22 Gly Ile Leu Ser Leu Phe Thr Gly Gly Ile Lys Ala Leu Gly Lys Thr Leu Phe Lys Met Ala Gly Lys Ala Gly Ala Glu His Leu Ala Cys Lys Ala Thr Asn Gln Cys <210> 23 <211> 37 <212> PRT
<213> Rana berlandieri <400> 23 Gly Leu Phe Ser Ile Leu Arg Gly Ala Ala Lys Phe Ala Ser Lys Gly Leu Gly Lys Asp Leu Thr Lys Leu Gly Val Asp Leu Val Ala Cys Lys Ile Ser Lys Gln Cys <210> 24 <211> 37 <212> PRT
<213> Rana pipiene>
<400> 24 Gly Phe Ser Ser Ile Phe Arg Gly Val Ala Lys Phe Ala Ser Lys Gly Leu Gly Lys Asp Leu Ala Arg Leu Gly Val Asn Leu Val Ala Cys Lys Ile Ser Lys Gln Cys <210> 25 <211> 37 <212> PRT
<213> Rana catesbeaana <220>
<221> MUTAGEN
<222> 1....37 <223> Xaa = Z = Any amino acid residue in same residue position of other family members arranged homologously <400> 25 Gly Xaa Xaa Ser Xaa Xaa Xaa Gly Xaa Xaa Lys Xaa Xaa Xaa Lys Xaa Leu Xaa Lys Xaa Xaa Xaa Xaa Xaa Gly Xaa Xaa Xaa Xaa Ala Cys Lys Xaa Xaa Xaa Gln Cys <210> 26 <211> 24 <212> PRT
<213> Rana catesbe;iana <400> 26 Phe Leu Pro Phe Ile Ala Arg Leu Ala Ala Lys Val Phe Pro Ser Ile Ile Cys Ser Val Thr Lys Lys Cys <210> 27 <211> 24 <212> PRT
<213a Rana luteive:ntris <400> 27 Phe Leu Pro Met Leu Ala Gly Leu Ala Ala Ser Met Val Pro Lys Leu Val Cys Leu Ile Thr Lys Lys Cys <210> 28 <211> 24 <212> PRT
<213> Rana luteive:ntris <400> 28 Phe Leu Pro Met Leu Ala Gly Leu Ala Ala Ser Met Val Pro Lys Phe Val Cys Leu Ile Thr Lys Lys Cys <210> 29 <211> 24 <212> PRT
<213> Rana berland.ieri <400> 29 Phe Leu Pro Ala Ile Ala Gly Met Ala Ala Lys Phe Leu Pro Lys Ile Phe Cys Ala Ile Ser Lys Lys Cys <210> 30 <211> 24 <212> PRT
<213> Rana berland.ieri <400> 30 Phe Leu Pro Ala Ile Ala Gly Met Ala Ala Lys Phe Leu Pro Lys Ile Phe Cys Ala Ile Ser Lys :Gys Cys <210> 31 <211> 24 <212> PRT
<213> Rana berlandieri <400> 31 Phe Leu Pro Phe Ile Ala G1y Val Ala Ala Lys Phe Leu Pro Lys Ile Phe Cys Ala Ile Ser Lys :Lys Cys <210> 32 <211> 24 <212> PRT
<213> Rana berlandieri <400> 32 Phe Leu Pro Ala Ile Ala c3ly Val Ala Ala Lys Phe Leu Pro Lys Ile Phe Cys Ala Ile Ser Lys Lys Cys <210> 33 <211> 24 <212> PRT
<213> Rana berland:ieri <400> 33 Phe Leu Pro Ala Ile Val c3ly Ala Ala Ala Lys Phe Leu Pro Lys Ile 1 5 l0 15 Phe Cys Val Ile Ser Lys Lys Cys <210> 34 <211> 24 <212> PRT
<213> Rana berland:ieri <400> 34 Phe Leu Pro Phe Ile Ala c3ly Met Ala Ala Asn Phe Leu Pro Lys Ile Phe Cys Ala Ile Ser Lys Lys Cys <210> 35 <211> 24 <212> PRT
<213> Rana pipiens <400> 35 Phe Leu Pro Ile Ile Ala c3ly Val Ala Ala Lys Val Phe Pro Lys Ile Phe Cys Ala Ile Ser Lys Lys Cys <210> 36 <211> 24 <212> PRT

<213> Rana pipiens <400> 36 Phe Leu Pro Ile Ile Ala Gly Ile Ala Ala Lys Val Phe Pro Lys Ile Phe Cys Ala Ile Ser Lys Lys Cys <210> 37 <211> 24 <2I2> PRT
<213> Rana pipiens <400> 37 Phe Leu Pra Ile Ile Ala Ser Val Ala Ala Lys Val Phe Ser Lys Ile Phe Cys Ala Ile Ser Lys Lys Cys <210> 38 <211> 24 <212> PRT
<213> Rana pipiens <400> 38 Phe Leu Pro Ile Ile Ala Ser Val Ala Ala Asn Val Phe Ser Lys Ile Phe Cys Ala Ile Ser Lys Lys Cys <210> 39 <211> 24 <212> PRT
<213> Rana pipiens <400> 39 Phe Leu Pro Ile Ile Ala Ser Val Ala Ala Lys Val Phe Pro Lys Ile Phe Cys Ala Ile Ser Lys Lys Cys <210> 40 <211> 24 <212> PRT
<213> Rana catesbeiana <220>
<221> MUTAGEN
<222> 1....24 <223> Xaa = U a Any amino acid residue in same residue position of ether family members arranged homologously <400> 40 Phe Leu Pro Xaa Ile Xaa Gly Xaa Ala Ala Xaa Xaa Xaa Pro Xaa Xaa Xaa Cys Xaa Ile Xaa Lys Lys Cys <210> 41 <211> 20 <212> PRT

<213> Rana grylio <400> 41 Phe Leu Gly Gly Leu Met Lys Ala Phe Pro Ala Leu Ile Cys Ala Val Thr Lys Lys Cys <210> 42 <211> 20 <212> PRT
<213> Rana clamitans <400> 42 Phe Leu Gly Gly Leu Met Lys Ala Phe Pro Ala Leu Ile Cys Ala Val Thr Lys Lys Cys <210> 43 <211> 20 <212> PRT
<213> Rana rugosa <220>
< 2 21 > MUTA(3EN
<222> 1....20 <223> Xaa = J = Any amino acid residue in same residue position of other family members arranged homologously <400> 43 Phe Leu Gly Gly Leu Met Lys Xaa Xaa Pro Ala Xaa Xaa Cys Ala Val Thr Lys Lys Cys <210> 44 <211> 12 <212> PRT
<213> Rana catesbe:iana <400> 44 Phe Leu Pro Ile Ala Ser Leu Leu Gly Lys Tyr Leu <210> 45 <211> 13 <212 > PRT
<213> Rana catesbeiana <400> 45 Phe Ile Ser Ala Ile Ala Ser Met Leu Gly Lys Phe Leu <210> 46 <211> 13 <212> PRT
<213> Rana catesbeiana <400> 46 Phe Leu Ser Ala Ile Ala Ser Met Leu Gly Lys Phe Leu <210> 47 <211> 13 <212> PRT
<213 > Rana catesbe:iana <400> 47 Phe Ile Ser Ala Ile Ala Ser Phe Leu Gly Lys Phe Leu <210> 48 <211> 14 <212> PRT
<213> Rana catesbe:iana <400> 48 Phe Leu Phe Pro Leu Ile Thr Ser Phe Leu Ser Lys Val Leu <210> 49 <211> 13 <212> PRT
<213> Rana clamitans <400> 49 Phe Leu Pro Phe Leu Ala Thr Leu Leu Ser Lys Val Leu <210> 50 <211> 13 <212> PRT
<213> Rana clamita.ns <400> 50 Phe Leu Pro Phe Leu Ala Lys Ile Leu Thr Gly Val Leu <210> 51 <211> 13 <212> PRT
<213> Rana clamita.ns <400> 51 Phe Leu Pro Leu Phe Ala Ser Leu Ile Gly Lys Lys Leu <210> 52 <211> 13 <212> PRT
<213> Rana pipiens <400> 52 Phe Leu Pro Ile Val Gly Lys Leu Leu Ser Gly Leu Leu <210> 53 <211> 14 <212> PRT
<213> Rana luteiventris <400> 53 Phe Leu Pro Ile Leu Ile Asn Leu Ile His Lys Gly Leu Leu <210> 54 <211> 14 <212> PRT
<213> Rana catesbeiana <220>
<221> MUTAGEN
<222> 1....14 <223> Xaa = 0 = Any amino acid residue in same residue position of other family members arranged homologously <400> 54 Phe Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Leu Xaa <210> 55 <211> 13 <212> PRT
<213> Rana luteiventris <400> 55 Val Leu Pro Leu Ile Ser /Met Ala Leu Gly Lys Leu Leu <210> 56 <211> 14 <212> PRT
<213> Rana luteive:ntris <400> 56 Asn Phe Leu Gly Thr Leu Ile Asn Leu Ala Lys Lys Ile Met <210> 57 <211> 14 <212> PRT
<213> Rana grylio <400> 57 Ser ile Leu Pro Thr Ile 'Val Ser Phe Leu Ser Lys Phe Val 1 S l0 <210> 58 <211> 14 <212> PRT
<213> Rana grylio <400> 58 Ser Ile Leu Pro Thr Ile 'Val Ser Phe Leu Ser Lys Phe Leu <210> 59 <211> 15 <212> PRT
<213> Rana grylio <400> 59 Ser Ile Leu Pro Thr Ile 'Val Ser Phe Leu Thr Lys Phe Ile Leu <210> 60 <211> 15 <212> PRT
<213> Rana grylio <400> 60 Phe Ile Ile Pro Leu Ile Ala Ser Phe Leu Ser Lys Phe Ile Leu <210> 61 <211> 15 <212> PRT
<213> Rana catesbeiana <220>
<221> MUTAGEN
<222> 1....15 <223> Xaa = J = P.ny amino acid residue in same residue position of other family members arranged homologously <400> 61 Xaa Xaa Xaa Xaa Xaa Ile Xaa Ser Phe Leu Xaa Lys Xaa Xaa Leu <210> 62 <211> 25 <212> PRT
<213> Unknown <220>
<221> VARIANT
<222> 1....25 <223> Model sequence for structure activity relationship determination <400> 62 Ser Met Leu Ser Val Leu Lys Asn Leu Gly Lys Val Gly Leu Gly Phe Val Ala Cys Lys Ile Asn Lys Gln Cys <210> 63 <211> 25 <212> PRT
<213> Unknown <220>
<221> VARIANT
<222> 1....25 <223> Model sequence for structure activity relationship determination <400> 63 Ser Met Leu Ser Val Leu Lys Asn Leu Gly Lys Val Gly Leu Gly Phe i 5 10 15 Val Ala Cys Lys Ile Asn Lys Gln Cys

Claims (11)

What is claimed is:
1. An antibacterial peptide selected from any of the following Rana peptide families wherein. each peptide of a family is in the form of a C-terminus carboxylic acid, a C-terminus amide or a C-terminus ester, wherein the single or multiple dashed lines of the Ranatuerin-2 family peptides signify a single bond between the two amino acid residues adjacent the dashed lines, and wherein each dashed line of the Ranatuerin-2 family peptides stands for an amino acid residue position for the purpose of aligning the amino acid residues of the peptides of that family so that the residues are homologously positioned:
Ranatuerin-1 family Ranatuerin-1 SMLSVLKNLGKVGLGFVACKINKQC (SEQ ID NO:8) Ranatuerin-1G SMISVLKNLGKVGLGFVACKVNKQC (SEQ ID NO:9) Ranatuerin-1C SMLSVLKNLGKVGLGLVACKINKQC (SEQ ID NO:
10) Ranatuerin-2 family Ranatuerin-2 GLFLDTLKGAAKD-----AGK-LEGLKCKITGCKLP (SEQ ID NO:
12) Ranatuerin-3 GFL-DIIKNLGKTF---AGHMLDKIKCTIGTCPPSP (SEQ ID NO:
13) Ranatuerin-2P GLM DTVKNVAKNL---AGHMLDKLKCKITGC (SEQ ID NO:
14) Ranatuerin-2B GLL DTIKGVAKTV---AASMLDKLKCKISGC (SEQ 1D NO:
15) Ranatuerin-2Ca GLFLDTLKGAAKDV---AGKLLEGLKCKIAGCKP (SEQ ID NO:

16) Ranatuerin-2Cb GLFLDTLKGL--------AGKLLQGLKCIKAGCKP (SEQ ID NO:
17) Ranatuerin-2La GI-LDSFKGVAKGVAKDLAGKLLDKLKCKITGC (SEQ ID NO:
18) Ranatuerin-2Lb GILSSI-KGVAKGVAKNVAAQLLDTLKCKITGC (SEQ ID NO:
19) Ranatuerin-2G GLLLDTLKGAAKDI----AGIALEKLKCKITGCKP (SEQ ID NO:
20) Esculentin-2 family Esculentin-2L GILSLFTGGIKALGKTLFKMAGKAGAEHLACKATNQC (SEQ 1D NO:
22) Esculentin-2B GLFSILRGAAKFASKGLGKDLTKLGVDLVACKISKQC (SEQ ID NO:
23) Esculentin-2P GFSSIFRGVAKFASKGLGKDLARLGVNLVACKISKQC (SEQ ID NO:
24) Brevinin-1 family Ranatuerin-4 FLPFIARLAAKVFPSIICSVTKKC (SEQ 1D NO:26) Brevinin-1La FLPMLAGLAASMVPKLVCLITKKC (SEQ ID NO:27) Brevinin-ILb FLPMLAGLAASMVPKFVCLITKKC (SEQ ID NO:28) Brevinin-1Ba FLPAIAGMAAKFLPKIFCAISKKC (SEQ ID NO:29) Brevinin-1Bb FLPAIAGMAAKFLPKIFCAISKKC (SEQ ID NO:30) Brevinin-1Bc FLPFTAGVAAKFLPKIFCAISKKC (SEQ ID NO:31) Brevinin-1Bd FLPAIAGVAAKFLPKIFCAISKKC (SEQ ID NO:32) Brevinin-1Be FLPAIVGAAAKFLPKIFCVISKKC (SEQ ID NO:33) Brevinin-1Bf FLPFIAGMAANFLPKIFCAISKKC (SEQ ID NO:34) Brevinin-1Pa FLPIIAGVAAKVFPKIFCAISKKC (SEQ ID NO:35) Brevinin-1Pb FLPIIAGIAAKVFPKIFCAISKKC (SEQ ID NO:36) Brevinin-1Pc FLPIIASVAAKVFSKIFCAISKKC (SEQ ID NO:37) Brevinin-1Pd FLPIIASVAANVFSKIFCAISKKC (SEQ ID NO:38) Brevinin-1Pe FLPIIASVAAKVFPKIFCAISKKC (SEQ ID NO:39) Ranalexin-1 family Ranalexin-1C FLGGLMKAFPALICAVTKKC (SEQ ID NO:41) Ranalexin-1G FLGGLMKIIPAAFCAVTKKC (SEQ ID NO:42) Temporin A family wherein the C-terminus is a simple carboxamide Ranatuerin 5 FLPIASLLGKYL (SEQ ID NO:
44) Ranatuerin 6 FISAIASMLGKFL (SEQ ID NO:

45) Ranatuerin 7 FISAIASFLGKFL (SEQ ID NO:

46) Ranatuerin 8 FISAIASFLGKFL (SEQ ID NO:
47) Ranatuerin 9 FLFPLITSFLSKVL (SEQ ID NO:

48) Temporin-1Ca FLPFLATLLSKVL (SEQ ID NO:

49) Temporin-1Cb FLPFLAKILTGVL (SEQ ID NO:

50) Temporin-1Cc FLPIVGKLLSGLL (SEQ ID NO:

51) Temporin-lPa FIPIVGKLLSGLL (SEQ ID NO:

52) Temporin-1Lc FLPILINLIHKGLL (SEQ ID NO:
53) Temporin-B Family wherein the C-terminus is a simple carboxamide Temporin-1La VLPLISMALGKLL (SEQ ID NO:

55) Temporin-1Lb NFLGTLINLAKKIM (SEQ ID NO:

56) Temporin-1Ga SILPTIVSFLSKFV (SEQ ID NO:

57) Temporin-1Gb SILPTIVSFLSKFL (SEQ ID NO:

58) Temporin-1Gc SILPTIVSFLTKFIL (SEQ ID NO:

59) Temporin-1Gd FIIPLIASFLSKFIL (SEQ ID NO:

60)
2. A modified Rana peptide of any of the following formulas wherein each formula corresponds to a single Rana peptide family of claim 1 and wherein the symbols X, B, Z, U, O, J' and J and their position/location within a formula designate either any of the amino acid residues occurring at the same position within the other peptides of the same family or a conservative substitution of an amino acid residue for the amino acid substitution of an amino acid residue at the same position for any of the peptides within the same family:
(i) a Ranatuerin-1 family modified Rana peptide of the formula SMXSVLKNLGKVGLGXVACKXNKQC (SEQ ID NO: 11) having the designation Ranatuerin-A mod;
(ii) a Ranatuerin-2 family modified Rang peptide of the formula GBBBBBBKBBBBBBBBBBABBBBBBBKCBBBBCBBB (SEQ ID NO:21) and having the designation Ranatuerin-2 mod;

(iii) an Esculentin-2 family modified Rana peptide of the formula GZZSZZZGZZKZZZKZLZKZZZZZGZZZZACKZZZQC (SEQ ID NO:22) and having the designation Esculentin-2 mod;
(iv) a Brevinin-1 family modified Rana peptide of the formula FLPUUUUUAAUUUUUUUCUUUKKC (SEQ ID NO:
40) and having the designation Brevinin-1mod;
(v) a Ranalexin-1 family modified Rana peptide of the formula FLGGLMKJ'J'PAJ'J'CAVTKKC (SEQ ID NO:43) and having the designation Ranalexin mod;
(vi) a Temporin-A family modified Rana peptide of the formula FOOOOOOOOOOOLO (SEQ ID NO:54) and having the designation Temporin-A mod;
(vii) a Temporin-B family modified Rana peptide of the formula JJJJJIJSFLJKFJL (SEQ ID NO:61) and having the designation Temporin-B mod.
3. A modified Rana peptide having the amino acid sequence of any of the Rana peptides of claim 1 wherein one or more of the following amino acid substitutions have been made:
a) alanine for cysteine;
b) serine for cysteine;
c) alanine for glycine;
d) valine for glycine;
e) lysine for glycine;
f) glutamine for glycine;
g) arginine for lysine;
h) alanine for asparagine i) coupling a C10 to C20 fatty acid to the lysine side chain.
4. A truncated Rana peptide of claim 1.
5. A composition comprising a purified, antibacterial extract of the skin of a mature Ranid frog of the species selected from catesbeiana, luteiventris, berlandieri, grylio, clamitans and pipiens.
6. A pharmaceutical composition comprising an effective antibacterial amount of a Rana peptide of claim 1 and a suitable pharmaceutical carrier.
7. A pharmaceutical composition comprising an effective antibacterial amount of a modified Rana peptide of claim 2 and a suitable pharmaceutical carrier.
8. A pharmaceutical composition comprising an effective antibacterial amount of a truncated Rana peptide of claim 3 and a suitable pharmaceutical carrier.
9. A method of treatment of a bacterial infection in a patient comprising administering to the patient an antibacterially effective amount of a Rana peptide of claim 1, a modified Rang peptide of claim 2 or 3, or a truncated Rana peptide of claim 4, or a composition of claim 5, or a pharmaceutical composition of claim 6, 7 or 8.
10. A Rana peptide of claim 1 or 2 wherein the peptide is the amide or ester, the amide being a simple amide or an amide of a C1 to C10 aliphatic or aromatic primary, secondary or tertiary amine, and the ester being an ester of a C1 to C10 aliphatic or aromatic alcohol.
11. A Rana peptide of claim 1 selected from the Temporin A or Temporin B family wherein the C-terminus is a carboxylic acid.
CA002338885A 1998-08-14 1999-08-13 Antimicrobial peptides isolated from the skin of american frogs Abandoned CA2338885A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US9660798P 1998-08-14 1998-08-14
US60/096,607 1998-08-14
PCT/US1999/018575 WO2000009553A2 (en) 1998-08-14 1999-08-13 Antimicrobial peptides isolated from the skin of american frogs

Publications (1)

Publication Number Publication Date
CA2338885A1 true CA2338885A1 (en) 2000-02-24

Family

ID=22258186

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002338885A Abandoned CA2338885A1 (en) 1998-08-14 1999-08-13 Antimicrobial peptides isolated from the skin of american frogs

Country Status (5)

Country Link
EP (1) EP1104437A2 (en)
JP (1) JP2002522556A (en)
AU (1) AU5564299A (en)
CA (1) CA2338885A1 (en)
WO (1) WO2000009553A2 (en)

Families Citing this family (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001058942A1 (en) * 2000-02-11 2001-08-16 Bionebraska, Inc. Antimicrobial peptides isolated from the skin of the hyperoliid frog, kassina senegalensis
CN1650005B (en) 2002-04-25 2010-12-08 东亚合成株式会社 Antimicrobial polypeptide and utizliation thereof
CN1269837C (en) 2002-09-02 2006-08-16 上海高科联合生物技术研发有限公司 Serial synthetic antibacterial peptide
EP1688486B1 (en) 2003-10-29 2016-12-14 Toagosei Co., Ltd. Antibacterial peptide and utilization of the same
WO2005109158A2 (en) 2004-05-05 2005-11-17 Massachusetts Institute Of Technology Methods and systems for generating peptides
JP4507080B2 (en) 2004-07-30 2010-07-21 東亞合成株式会社 Antibacterial peptides and their use
JP4730584B2 (en) 2004-12-06 2011-07-20 東亞合成株式会社 Antibacterial peptides and their use
WO2006088010A1 (en) 2005-02-15 2006-08-24 Toagosei Co., Ltd. Antimicrobial peptide and use thereof
US7739055B2 (en) 2005-11-17 2010-06-15 Massachusetts Institute Of Technology Methods and systems for generating and evaluating peptides
KR100794499B1 (en) * 2006-05-16 2008-01-16 주식회사 프로메디텍 Novel analogues of antimicrobial and anticancer peptide synthesized and produced from Gaegurin 5
MX351747B (en) * 2008-06-04 2017-10-17 Enrique Islas Rodriguez Alfonso Extract from the skin of rana catesbeiana, compound and method for treating mastitis in cattle.
CN102250216B (en) * 2011-06-27 2013-03-06 昆明理工大学 Rana nigromaculata antimicrobial peptide as well as gene and application thereof
GB201401673D0 (en) * 2014-01-31 2014-03-19 Univ Ulster Esculentin-2CHa peptide and analogues thereof
CN110386961B (en) * 2019-08-29 2022-05-10 昆明医科大学 Skin repair polypeptide RL-RL10 and application thereof
CN112042879A (en) * 2020-09-17 2020-12-08 湖南富高农业科技开发有限公司 Salted egg yolk fried frog skin and preparation method thereof
CN113855783B (en) * 2021-11-09 2023-02-21 中国药科大学 Application of polypeptide in preparation of antitumor drugs
CN115246877A (en) * 2021-12-27 2022-10-28 昆明理工大学 Rana nigromaculata source antibacterial peptide Brevinin-1EG and application
CN115819515B (en) * 2022-11-30 2023-10-20 四川大学华西医院 Antibacterial peptide and preparation method and application thereof
CN117338956B (en) * 2023-11-08 2024-03-05 山东第一医科大学附属眼科医院(山东省眼科医院) Cascade immune regulation nano-drug for treating pseudomonas aeruginosa keratitis and preparation method and application thereof

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5225399A (en) * 1991-04-15 1993-07-06 The Children's Hospital Of Philadelphia Antimicrobial amphiphilic peptides
SE9604593D0 (en) * 1996-12-13 1996-12-13 Sbl Vaccin Ab Antimicrobially active polypeptides

Also Published As

Publication number Publication date
WO2000009553A3 (en) 2000-05-18
WO2000009553A2 (en) 2000-02-24
JP2002522556A (en) 2002-07-23
AU5564299A (en) 2000-03-06
EP1104437A2 (en) 2001-06-06

Similar Documents

Publication Publication Date Title
CA2338885A1 (en) Antimicrobial peptides isolated from the skin of american frogs
EP1853620B1 (en) Antimicrobial hexapeptides
Basir et al. Multiple antimicrobial peptides and peptides related to bradykinin and neuromedin N isolated from skin secretions of the pickerel frog, Rana palustris
Mor et al. Structure, synthesis, and activity of dermaseptin b, a novel vertebrate defensive peptide from frog skin: relationship with adenoregulin
Conlon et al. Antimicrobial peptides from the skins of North American frogs
Conlon et al. Strategies for transformation of naturally-occurring amphibian antimicrobial peptides into therapeutically valuable anti-infective agents
EP0846128B1 (en) Antimicrobial cationic peptides and methods of screening for the same
Halverson et al. Purification and characterization of antimicrobial peptides from the skin of the North American green frog Rana clamitans☆
KR102137941B1 (en) Antimicrobial Peptide
CA2441562C (en) Short bioactive peptides and methods for their use
Batista et al. Antimicrobial peptides from the Brazilian frog Phyllomedusa distincta1
KR101046426B1 (en) Novel antifungal peptides and antifungal composition containing thereof
Kim et al. Purification and characterization of antimicrobial and vasorelaxant peptides from skin extracts and skin secretions of the North American pig frog Rana grylio
US6310176B1 (en) Antimicrobially active polypeptides
US20030219854A1 (en) Methods for producing modified anti-infective peptides
Jin et al. Characterization of antimicrobial peptides isolated from the skin of the Chinese frog, Rana dybowskii
Goraya et al. Ranatuerin 1T: an antimicrobial peptide isolated from the skin of the frog, Rana temporaria
Won et al. Structure-activity relationships of antimicrobial peptides from the skin of Rana esculenta inhabiting in Korea
JP2001186887A (en) Antimicrobial peptide originating from pandinus imperator
WO2002040536A2 (en) Acylated antimicrobial peptides
JPH06504260A (en) Amphipathic peptide compositions and analogs thereof
EP1814904A2 (en) Antibiotic drosocin derivatives
KR101720539B1 (en) Antimicrobial peptide analogues Ov1-1, Ov1-2 derived from Octopus variabilis and antimicrobial composition containing the same
KR101723949B1 (en) Antimicrobial peptide analogue Ov2-2 derived from Octopus variabilis and antimicrobial composition containing the same
WO2022162214A1 (en) Novel selective antimicrobial fusion peptides

Legal Events

Date Code Title Description
FZDE Dead