16
Nonerythropoietic, tissue-protective peptides derived from the tertiary structure of erythropoietin Michael Brines* †‡ , Nimesh S. A. Patel § , Pia Villa , Courtenay Brines*, Tiziana Mennini , Massimiliano De Paola , Zubeyde Erbayraktar**, Serhat Erbayraktar**, Bruno Sepodes †† , Christoph Thiemermann § , Pietro Ghezzi †¶ , Michael Yamin*, Carla C. Hand , Qiao-wen Xie* , Thomas Coleman* †‡‡ , and Anthony Cerami* †‡ *Warren Pharmaceuticals, Ossining, NY 10562; The Kenneth S. Warren Institute, Ossining, NY 10562; § Centre for Translational Medicine and Therapeutics, William Harvey Research Institute, Barts and London School of Medicine and Dentistry, Queen Mary, University of London, London EC1M 6BQ, England; Mario Negri Institute for Pharmacological Research, 20-20156 Milan, Italy; **Dokuz Eylu ¨ l University, Izmir 35340, Turkey; †† Faculty of Pharmacy, University of Lisbon, 1600 Lisbon, Portugal; and National Research Council, Institute of Neuroscience, 20129 Milan, Italy Contributed by Anthony Cerami, June 10, 2008 (sent for review May 22, 2008) Erythropoietin (EPO), a member of the type 1 cytokine superfamily, plays a critical hormonal role regulating erythrocyte production as well as a paracrine/autocrine role in which locally produced EPO protects a wide variety of tissues from diverse injuries. Signifi- cantly, these functions are mediated by distinct receptors: hema- topoiesis via the EPO receptor homodimer and tissue protection via a heterocomplex composed of the EPO receptor and CD131, the common receptor. In the present work, we have delimited tissue- protective domains within EPO to short peptide sequences. We demonstrate that helix B (amino acid residues 58 – 82) of EPO, which faces the aqueous medium when EPO is bound to the receptor homodimer, is both neuroprotective in vitro and tissue protective in vivo in a variety of models, including ischemic stroke, diabetes-induced retinal edema, and peripheral nerve trauma. Remarkably, an 11-aa peptide composed of adjacent amino acids forming the aqueous face of helix B is also tissue protective, as confirmed by its therapeutic benefit in models of ischemic stroke and renal ischemia–reperfusion. Further, this peptide simulating the aqueous surface of helix B also exhibits EPO’s trophic effects by accelerating wound healing and augmenting cognitive function in rodents. As anticipated, neither helix B nor the 11-aa peptide is erythropoietic in vitro or in vivo. Thus, the tissue-protective activ- ities of EPO are mimicked by small, nonerythropoietic peptides that simulate a portion of EPO’s three-dimensional structure. cognition cytoprotection excitotoxicity ischemia–reperfusion injury wound healing I n its hormonal role, the cytokine erythropoietin (EPO) is released by the kidney into the circulation in response to hypoxia and binds to a preformed receptor homodimer (EPOR) 2 present on the cell membrane of erythrocytic progenitors. Subsequently, a molecular cascade begins with the phosphory- lation of Janus tyrosine kinase 2 and ultimately results in inhibition of programmed cell death, fostering the survival and maturation of erythroid precursors to erythrocytes [reviewed by Fisher (1)]. However, over the last 15 years, it has been discov- ered that EPO is also synthesized locally by many tissues, especially in response to metabolic stress. This pool of EPO acts as a multifunctional protective molecule [reviewed by Brines and Cerami (2)]. In this paracrine/autocrine role, EPO inhibits apoptosis in a wide variety of cell types and activates multiple mechanisms to protect stressed tissues, e.g., reducing inf lamma- tion and local edema. EPO also plays crucial roles during development (3). Therefore, it is not surprising that in the adult organism, EPO mediates multiple trophic effects, leading to accelerated healing and tissue regeneration. Finally, EPO has been shown to enhance cognition in normal (4) as well as diseased (5) human subjects. The molecular interaction of EPO with the erythropoietic receptor (EPOR) 2 has been studied intensively, such that the regions of EPO that interact with (EPOR) 2 have been identified (Fig. 1A). These include portions of helices A and C (site 2), as well as helix D and the loop connecting helices A and B (site 1) (6–9). Chemical or mutational modifications of amino acid residues within these two regions of EPO abolish its binding to (EPOR) 2 and, therefore, these modified EPOs are not erythro- poietic in vivo or in vitro. Remarkably, a number of these modified EPOs retain potent tissue-protective properties (10). Clearly, sites 1 and 2, which are essential for erythropoiesis, are not required for tissue protection. These observations suggest that an additional receptor for EPO mediates tissue protection. This receptor is pharmacolog- ically distinct from that of erythropoiesis, because it exhibits a lower affinity for EPO and forms distinct molecular species in cross-linking experiments (11). In prior studies, we have pro- vided evidence that the receptor that promotes tissue protection is a heteromer composed of EPOR and CD131, the common receptor (cR) (12). CD131 also forms receptor complexes with the receptor subunits specific for GM-CSF, IL-3, and IL-5 and has been termed the ‘‘common’’ receptor [reviewed by Murphy and Young (13)]. Results from experiments showing that chemical modification of lysine residues or amino acid substitutions made within sites 1 and/or 2 do not affect tissue protection suggest that other regions of EPO contain the recognition site for the tissue- protective receptor. Notably, in aqueous media, EPO’s tertiary structure is relatively well defined because of the interaction of the hydrophobic content of its four -helices, constraining the molecule into a compact, relatively rigid, globular structure. When EPO is bound to the hematopoietic receptor (14), helix B and parts of the AB and CD loops face the aqueous medium, away from the homodimer binding sites [Protein Data Bank (PDB) ID code 1EER; Fig. 1]. These regions do not contain lysine and therefore are not modified by carbamylation of EPO, a procedure that produces a selectively tissue-protective com- pound (10). In view of these observations, we hypothesized that tissue protection, as distinct from erythropoiesis, depends on a region within helix B and/or loop AB within the EPO molecule. Author contributions: M.B., N.S.A.P., C.T., P.G., T.C., and A.C. designed research; M.B., N.S.A.P., P.V., C.B., T.M., M.D.P., Z.E., S.E., C.C.H., Q.-w.X., and T.C. performed research; M.B. and C.C.H. contributed new reagents/analytic tools; M.B., N.S.A.P., P.V., C.B., T.M., M.D.P., B.S., Q.-w.X., and T.C. analyzed data; and M.B., N.S.A.P., C.T., M.Y., T.C., and A.C. wrote the paper. Conflict of interest statement: M.B., C.B., M.Y., Q.-w.X., T.C., and A.C. were employees of Warren Pharmaceuticals when this work was performed. Warren Pharmaceuticals is developing erythropoietin analogues and tissue-protective compounds for potential clin- ical uses. To whom correspondence may be addressed: [email protected] or acerami@ warrenpharma.com. ‡‡ Present address: Feinstein Institute for Medical Research, Manhasset, NY 11030. This article contains supporting information online at www.pnas.org/cgi/content/full/ 0805594105/DCSupplemental. © 2008 by The National Academy of Sciences of the USA www.pnas.orgcgidoi10.1073pnas.0805594105 PNAS August 5, 2008 vol. 105 no. 31 10925–10930 MEDICAL SCIENCES

Brines et al PNAS 2008 + suppl materials–note-pHBS is ARA 290

  • Upload
    vuongtu

  • View
    221

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Brines et al PNAS 2008 + suppl materials–note-pHBS is ARA 290

Nonerythropoietic, tissue-protective peptides derivedfrom the tertiary structure of erythropoietinMichael Brines*†‡, Nimesh S. A. Patel§, Pia Villa¶�, Courtenay Brines*, Tiziana Mennini¶, Massimiliano De Paola¶,Zubeyde Erbayraktar**, Serhat Erbayraktar**, Bruno Sepodes††, Christoph Thiemermann§, Pietro Ghezzi†¶,Michael Yamin*, Carla C. Hand†, Qiao-wen Xie*†, Thomas Coleman*†‡‡, and Anthony Cerami*†‡

*Warren Pharmaceuticals, Ossining, NY 10562; †The Kenneth S. Warren Institute, Ossining, NY 10562; §Centre for Translational Medicine and Therapeutics,William Harvey Research Institute, Barts and London School of Medicine and Dentistry, Queen Mary, University of London, London EC1M 6BQ, England;¶Mario Negri Institute for Pharmacological Research, 20-20156 Milan, Italy; **Dokuz Eylul University, Izmir 35340, Turkey; ††Faculty of Pharmacy, Universityof Lisbon, 1600 Lisbon, Portugal; and �National Research Council, Institute of Neuroscience, 20129 Milan, Italy

Contributed by Anthony Cerami, June 10, 2008 (sent for review May 22, 2008)

Erythropoietin (EPO), a member of the type 1 cytokine superfamily,plays a critical hormonal role regulating erythrocyte production aswell as a paracrine/autocrine role in which locally produced EPOprotects a wide variety of tissues from diverse injuries. Signifi-cantly, these functions are mediated by distinct receptors: hema-topoiesis via the EPO receptor homodimer and tissue protection viaa heterocomplex composed of the EPO receptor and CD131, the �common receptor. In the present work, we have delimited tissue-protective domains within EPO to short peptide sequences. Wedemonstrate that helix B (amino acid residues 58–82) of EPO,which faces the aqueous medium when EPO is bound to thereceptor homodimer, is both neuroprotective in vitro and tissueprotective in vivo in a variety of models, including ischemic stroke,diabetes-induced retinal edema, and peripheral nerve trauma.Remarkably, an 11-aa peptide composed of adjacent amino acidsforming the aqueous face of helix B is also tissue protective, asconfirmed by its therapeutic benefit in models of ischemic strokeand renal ischemia–reperfusion. Further, this peptide simulatingthe aqueous surface of helix B also exhibits EPO’s trophic effects byaccelerating wound healing and augmenting cognitive function inrodents. As anticipated, neither helix B nor the 11-aa peptide iserythropoietic in vitro or in vivo. Thus, the tissue-protective activ-ities of EPO are mimicked by small, nonerythropoietic peptides thatsimulate a portion of EPO’s three-dimensional structure.

cognition � cytoprotection � excitotoxicity � ischemia–reperfusion injury �wound healing

In its hormonal role, the cytokine erythropoietin (EPO) isreleased by the kidney into the circulation in response to

hypoxia and binds to a preformed receptor homodimer (EPOR)2present on the cell membrane of erythrocytic progenitors.Subsequently, a molecular cascade begins with the phosphory-lation of Janus tyrosine kinase 2 and ultimately results ininhibition of programmed cell death, fostering the survival andmaturation of erythroid precursors to erythrocytes [reviewed byFisher (1)]. However, over the last 15 years, it has been discov-ered that EPO is also synthesized locally by many tissues,especially in response to metabolic stress. This pool of EPO actsas a multifunctional protective molecule [reviewed by Brines andCerami (2)]. In this paracrine/autocrine role, EPO inhibitsapoptosis in a wide variety of cell types and activates multiplemechanisms to protect stressed tissues, e.g., reducing inflamma-tion and local edema. EPO also plays crucial roles duringdevelopment (3). Therefore, it is not surprising that in the adultorganism, EPO mediates multiple trophic effects, leading toaccelerated healing and tissue regeneration. Finally, EPO hasbeen shown to enhance cognition in normal (4) as well asdiseased (5) human subjects.

The molecular interaction of EPO with the erythropoieticreceptor (EPOR)2 has been studied intensively, such that theregions of EPO that interact with (EPOR)2 have been identified

(Fig. 1A). These include portions of helices A and C (site 2), aswell as helix D and the loop connecting helices A and B (site 1)(6–9). Chemical or mutational modifications of amino acidresidues within these two regions of EPO abolish its binding to(EPOR)2 and, therefore, these modified EPOs are not erythro-poietic in vivo or in vitro. Remarkably, a number of thesemodified EPOs retain potent tissue-protective properties (10).Clearly, sites 1 and 2, which are essential for erythropoiesis, arenot required for tissue protection.

These observations suggest that an additional receptor forEPO mediates tissue protection. This receptor is pharmacolog-ically distinct from that of erythropoiesis, because it exhibits alower affinity for EPO and forms distinct molecular species incross-linking experiments (11). In prior studies, we have pro-vided evidence that the receptor that promotes tissue protectionis a heteromer composed of EPOR and CD131, the � commonreceptor (�cR) (12). CD131 also forms receptor complexes withthe � receptor subunits specific for GM-CSF, IL-3, and IL-5 andhas been termed the ‘‘common’’ receptor [reviewed by Murphyand Young (13)].

Results from experiments showing that chemical modificationof lysine residues or amino acid substitutions made within sites1 and/or 2 do not affect tissue protection suggest that otherregions of EPO contain the recognition site for the tissue-protective receptor. Notably, in aqueous media, EPO’s tertiarystructure is relatively well defined because of the interaction ofthe hydrophobic content of its four �-helices, constraining themolecule into a compact, relatively rigid, globular structure.When EPO is bound to the hematopoietic receptor (14), helix Band parts of the AB and CD loops face the aqueous medium,away from the homodimer binding sites [Protein Data Bank(PDB) ID code 1EER; Fig. 1]. These regions do not containlysine and therefore are not modified by carbamylation of EPO,a procedure that produces a selectively tissue-protective com-pound (10). In view of these observations, we hypothesized thattissue protection, as distinct from erythropoiesis, depends on aregion within helix B and/or loop AB within the EPO molecule.

Author contributions: M.B., N.S.A.P., C.T., P.G., T.C., and A.C. designed research; M.B.,N.S.A.P., P.V., C.B., T.M., M.D.P., Z.E., S.E., C.C.H., Q.-w.X., and T.C. performed research; M.B.and C.C.H. contributed new reagents/analytic tools; M.B., N.S.A.P., P.V., C.B., T.M., M.D.P.,B.S., Q.-w.X., and T.C. analyzed data; and M.B., N.S.A.P., C.T., M.Y., T.C., and A.C. wrote thepaper.

Conflict of interest statement: M.B., C.B., M.Y., Q.-w.X., T.C., and A.C. were employees ofWarren Pharmaceuticals when this work was performed. Warren Pharmaceuticals isdeveloping erythropoietin analogues and tissue-protective compounds for potential clin-ical uses.

‡To whom correspondence may be addressed: [email protected] or [email protected].

‡‡Present address: Feinstein Institute for Medical Research, Manhasset, NY 11030.

This article contains supporting information online at www.pnas.org/cgi/content/full/0805594105/DCSupplemental.

© 2008 by The National Academy of Sciences of the USA

www.pnas.org�cgi�doi�10.1073�pnas.0805594105 PNAS � August 5, 2008 � vol. 105 � no. 31 � 10925–10930

MED

ICA

LSC

IEN

CES

Page 2: Brines et al PNAS 2008 + suppl materials–note-pHBS is ARA 290

Results and DiscussionBased on data available from PDB ID code 1EER, we began thestudy by synthesizing helix B peptide (HBP; residues 58–82). Asexpected, this molecule, like carbamylated EPO [CEPO (10)],was not erythropoietic in the UT-7 EPO cell assay or in vivo (seesupporting information (SI) Materials and Methods and Fig. S1).

However, HBP possessed potent neuroprotective activity com-parable to EPO (Fig. 2A) and to CEPO (15) in a rat motoneuronmodel in vitro. In this model, the neurotoxic effects of theglutamate receptor agonist kainic acid were blocked by eitherHBP (1.8 nM) or EPO (3.3 nM). Because HBP is small and doesnot contain features designed to resist proteolysis or decreaseclearance, its plasma half-life is presumably very short. It wastherefore of great interest to determine whether HBP exhibitedprotective properties in vivo.

HBP was protective in a rat model of middle cerebral arteryocclusion that has previously shown large protective effects forEPO (16), asialo-EPO (17), and CEPO (10) (Fig. 2B). In thisexperiment, HBP administered as a single i.v. dose [1.5 nmol/kgof body weight (bw)] reduced infarct volume as determined bytetrazolium salt staining 24 h after reperfusion. HBP was alsoassociated with an improved behavioral outcome (foot faults ofthe saline group 24.6 � 2 versus 16.4 � 2 in the single dose HBPgroup and 14.5 � 0.9 in the four-dose HBP group; P � 0.05between saline and treated groups). Notably, additional doses ofHBP administered i.p. at 2-h intervals for three additional dosesdid not further improve the extent of neuroprotection.

Confirming that HBP was neuroprotective in vitro and in vivo,we then assessed whether HBP possessed other propertiesconsistent with EPO’s nonerythropoietic activities. For example,EPO reduces injury-related local edema in a number of tissues(18–21), including the retina (22). Specifically, in models ofdiabetic retinopathy, hyperglycemia produces endothelial injury,leading to vascular leakage and retinal edema. Further, it isnotable that in a small, retrospective study of diabetic patientswith macular edema, EPO treatment was associated with anincrease in visual acuity and a decrease in retinal exudates (23).To determine whether HBP could inhibit diabetes-related ret-inal edema, rats were administered streptozotocin. After theconfirmation of the diabetic state, HBP (1.5 nmol/kg of bw) orsaline was administered i.p. 5 days each week. After 3 weeks ofhyperglycemia, retinal leakage in the HBP group (as assessed byextravasation of Evans blue dye) was not different from animalswithout diabetes (Fig. 2C). In contrast, retinas from animals thatreceived only saline exhibited significant edema.

The results of these experiments showed that a peptidefragment of EPO comprising the amino acid sequence corre-sponding to helix B exhibited tissue-protective effects similar toEPO and its nonerythropoietic derivatives in a variety of in vitroand in vivo models. Previous study has shown that peptides can

AD

B

C

12

EPOR EPOR

Q Q A V E V W Q G L A L L S E A V L R G Q A L L V N S S

58 85

Q-E-Q-L-E-R-A-L-N-S-S

U-E-Q-L-E-R-A-L-N-S-S

A

B

Fig. 1. Structure of EPO indicating tissue protective domains and sequences.(A) Schematized drawing of EPO bound to the hematopoietic receptor dimer(EPOR)2, shown inserted into the plasma membrane. Helices A–D associate viahydrophobic interactions to form a compact, globular structure. Sites 1 and 2(indicated by dashed boxes) within the topography of the EPO molecule bindwith high affinity to each EPOR monomer. The aqueous face of helix B facesaway from the interior of the receptor, as indicated by the dashed ellipse. (B)Helix B peptide (HBP) was synthesized as the linear sequence of helix B (aminoacids 58–82: boxed region; single-letter code). Circled residues show thoseamino acid residues on the aqueous face of the 4-3 �-helix B. Leucine inposition 80 is also on the aqueous face. A linear peptide comprising only theseresidues was synthesized as HBSP. Residues 83–85 are relatively rigidly ori-ented because of the associated helices. Glutamine in the N-terminal positioncan spontaneously undergo cyclization into pyroglutamate (U), forming pH-BSP (bottom peptide).

A CB

control

HBP1.8 n

M MµµKA

5 Mµµ

HBP1.8 n

M +KA

5 Mµµ

EPO3.3 n

M +KA

5

0

20

40

60

80

100

120

140 ***

***

RelativeSurvival

PBS

HPB1.5 n

mole/kg

HBP1.5 n

mole/kg: 4

doses

0

50

100

150

200

250

300

** **

InfarctVolume[mm3 ]

diabetes +

PBS

diabetes +

HBP1.5 n

mole/kg

0.0

0.5

1.0

1.5

2.0

evansblueleakage

[comparedtonormal]

**

Fig. 2. Helix B peptide (HBP) is tissue protective in vitro and in vivo. (A) HBP protects against kainic acid (KA)-induced motoneuron excitotoxic death in vitro.Mixed anterior horn cultures obtained from the ventral horn of the spinal cord of 14-day rat embryos were treated on the sixth day in vitro by incubation for48 h with kainic acid (5 �M) alone or in cotreatment with EPO or HBP. Data are means and SEM; ***, P � 0.001 compared with kainic acid alone. (B) HBP isneuroprotective in a stroke model. A single dose of HBP administered i.v. immediately after a 1-h arterial occlusion significantly reduces infarct volumedetermined after 24 h. Additional doses of HBP do not improve protection. Data are means and SEM; **, P � 0.01 compared with saline. (C) HBP (n � 16) preventsthe development of retinal edema in a rat model of diabetes (PBS; n � 14). Results of three experiments are shown. Evans blue extravasation in normal retinas(n � 12) was 6 � 0.2 ng/mg of dry retina. Data are means and SEM; **, P � 0.01.

10926 � www.pnas.org�cgi�doi�10.1073�pnas.0805594105 Brines et al.

Page 3: Brines et al PNAS 2008 + suppl materials–note-pHBS is ARA 290

be synthesized to mimic the helical structure of a protein thatinteracts with its receptor to reproduce the biological activitiesof the full molecule (24). Upon further consideration, however,we reasoned that because helix B is amphipathic and of the 4-3�-helix type, specific amino acid residues within the hydrophilicportion face the external, aqueous face (i.e., every fourth andthird residue in the b and f position, respectively). Spatially (butnot linearly) adjacent residues therein could constitute a recog-nition site for the tissue-protective receptor.

Data obtained from crystallographic studies of EPO bound to(EPOR)2 show that the aqueous face of helix B consists of aminoacids QEQLERAL (PDB ID code 1EER; Fig. 1B). Thus, apeptide derived from surface-simulation analysis of EPO shouldpossess the biological activities of helix B. To test this hypothesis,a peptide was synthesized to include these surface amino acidsas well as the three residues within the proximal portion of theBC loop that are relatively constrained by the rigid structure ofthe associated helices. The resulting 11-mer helix B surfacepeptide (HBSP: QEQLERALNSS), unrelated in primary se-quence to EPO, was thus intended to mimic a particular featureof EPO’s three-dimensional structure, notwithstanding the pos-sible steric constraints of spatially but not linearly adjacentresidues bonded directly together. We subsequently assessedwhether this peptide was a nonerythropoietic, tissue-protectivemolecule.

HBSP, which does not contain site 1 or 2, was, as predicted,not erythropoietic in vitro (UT7-EPO cells) or in vivo in the rat(data not shown). However, HBSP was highly active in reducingthe degree of injury observed in a sciatic nerve crush injurymodel to a degree identical on a molar basis to EPO andchemical derivatives of EPO that are not erythropoietic (10, 17).In this model, the sciatic nerve was reversibly compressed by

using a ligature for a duration of 1 min and single doses of HBPor HBSP (0.3 nmol/kg of bw) administered i.v. immediately afterremoval of the constriction. The tissue-protective potency ofthese molecules was assessed by the static sciatic index as we havepreviously reported for asialo-EPO (17) and CEPO (10) andwere found to be equivalent (Fig. 3). In contrast, an equimolaramount of a 20-mer fragment of pigment epithelium-derivedfactor (amino acids 102–121), derived from a biologically activeregion of this molecule (25), was inactive.

Thus, a peptide designed to mimic the external, aqueous faceof helix B resembled EPO sufficiently to activate tissue-protective pathways. In the past, successful surface-simulationsynthesis has been reported for antigenic determinants in pro-teins (26), as well as the surface of the �-helix of HIV-1 virus(27). Although there have also been claims that peptides exhib-iting enzymatic activity can be synthesized from surface simu-lation analysis of the catalytic site of an enzyme (28), these havenot been substantiated (29). In retrospect, it seems very unlikelythat a small peptide could effectively reproduce the complexthree-dimensional structure required for enzymatic activity,because binding sites for proper orientation of the substrate tothe catalytic site require a rigid, three-dimensional scaffold notstructurally attainable by using a small peptide. In the case ofreceptor-mediated biological activity, however, binding and ac-tivation of a receptor can topologically be much simpler. Hence,a number of examples exist of small peptides that reproduce thebiological activity of a larger protein. For example, the clinicallyuseful parathyroid hormone fragment (1–34) possesses the samebiological activity as the full 84-aa protein (30). Further, it shouldbe noted that a 17-mer peptide derived from a portion of EPO’sAB loop (residues 30–47) has been reported to possess neuro-trophic activity (31). It is currently unclear whether this peptideinteracts with the tissue-protective receptor subtype or mediatesit biological effects by a different mechanism.

With respect to the primary structure of HBSP, however, it iswell known that N-terminal glutamine residues can undergo aspontaneous, irreversible cyclization (particularly at room tem-perature under acidic conditions) into pyroglutamate (32). Inconfirmation of this fact, amino acid analysis of productionbatches of HBSP revealed that �90% of the product possesseda free N-terminal glutamine, whereas the remainder was cy-clized. Thus, HBSP was actually a mixture of two peptides. Todetermine whether pyroglutamate HBSP (pHBSP) was biolog-ically active, it was synthesized de novo (Fig. 1B).

pHBSP (which was nonerythropoietic; see Figs. S2–S5) wasevaluated in a rodent model of renal ischemia–reperfusioninjury. Specifically, mice were randomized into five experimentalgroups and administered either vehicle or various amounts ofpHBSP as an i.p. bolus at 1 min, 6 h, and again at 12 h afterreperfusion. Twenty-four hours later, plasma creatinine and

PBS

PEDF

EPO

HBP

HBSP

0.50

0.55

0.60

0.65

0.70

****** ***-S

SI

Fig. 3. HBSP is equipotent to HBP and EPO in a sciatic nerve injury model.PEDF, pigment epithelium-derived factor. Compounds were administered ata dose of 0.3 nmol/kg of bw i.v. immediately after a 1-min compression of thesciatic nerve at the level of the mid-thigh. Data are means and SEM plotted asthe negative of the static sciatic index (SSI). n � 6–8 for each group; ***, P �0.001 compared with PBS.

C

sham PBS

pHBSP

0.08nmo

le/kg

pHBSP

0.8nmo

le/kg

pHBSP

8 nmole/

kg

0.0

0.5

1.0

1.5

PlasmaCreatinine

(mg/dl)

* *

sham PBS

pHBSP

0.08nmo

le/kg

pHBSP

0.8nmo

le/kg

pHBSP

8 nmole/

kg

0

100

200

300

PlasmaUrea

(mg/dl)

* *

sham PBS

pHBSP

0.08nmo

le/kg

pHBSP

0.8nmo

le/kg

pHBSP

8 nmole/

kg

0

100

200

300

400

PlasmaAST

(IU/L)

* *

A B

Fig. 4. Pyroglutamate Helix B surface peptide improves renal ischemia-reperfusion injury. Plasma creatinine (A), urea (B), and aspartate aminotransferase (AST)(C) were measured from mice (n � 12 each group) as biochemical markers of renal dysfunction and injury subsequent to sham-operation or renal ischemia–reperfusion injury (bilateral renal pedicle occlusion for 30 min). PBS or pHBSP (8.0 nmol/kg of bw) was administered i.p. 1 min, 6 h, and 12 h into reperfusion.Data represent mean and SEM; ***, P � 0.001 versus PBS.

Brines et al. PNAS � August 5, 2008 � vol. 105 � no. 31 � 10927

MED

ICA

LSC

IEN

CES

Page 4: Brines et al PNAS 2008 + suppl materials–note-pHBS is ARA 290

urea were obtained to estimate renal function and aspartateaminotransferase to assess injury. The results show a dose-dependent renoprotective effect, with the lowest dose adminis-tered (0.08 nmol/kg of bw) ineffective (Fig. 4). The degree ofprotection observed was similar to previous observations of EPOin this model (33).

Results of administering pHBSP in a stroke model as a singlei.v. dose (1.5 nmol/kg of bw) upon reperfusion after 1 h ofocclusion, followed by three additional injections at 2-h intervals,demonstrated a significant reduction in infarct volume at 24 h[225 � 20 mm3 for pHBSP (n � 8) compared with 291 � 23 mm3

for saline (n � 7); P � 0.05] and an improvement in neurologicalfunction (saline group foots faults 20.2 � 0.8 versus 11.2 � 1.1in the pHBSP group; P � 0.001). In contrast, a scrambled versionof HBSP (LSEQARNQSEL; n � 6) was biologically inactive(20.1 � 2.1 foot faults; P � 0.05 versus the pHBSP group). Thisobservation provides additional support that the surface struc-ture of helix B is specific for tissue-protective activities of EPO.

As noted above, EPO has also been observed to mediate otherbiological activities in addition to purely tissue-protective effects(reviewed in refs. 2 and 34). Among these pleiotropic effects,EPO accelerates wound healing and modulates cognitive func-tion. For example, EPO has been observed to promote incisionalwound closure in rodent models by reducing ischemic andreperfusion injury, mobilizing endothelial progenitor cells, aug-menting angiogenesis, and decreasing inflammation (35). Todetermine whether pHBSP could also provide benefits in woundhealing, we examined its effect in the healing of punch biopsywounds.

In this experiment, 3.5-mm-diameter full-thickness skinwounds were placed at the corners of a 3-cm-wide square on theshaved and depilated scapular region of the rat. pHBSP (24nmol/kg of bw) or PBS was administered s.c. daily for 10 days.The area of open wound, measured in a blinded fashion fromserial digital photographs, exhibited faster healing in animalsthat received peptide compared with saline controls (Fig. 5).Previous studies using systemically administered EPO haveshown acceleration of wound healing in association with in-creased expression of inducible NO synthase (iNOS) and in-creased angiogenesis, vascular endothelial growth factor, andwound collagen content (reviewed in ref. 34).

Historically, one of the first nonerythropoietic activities notedfor EPO was a strong neurotrophic effect (36). EPO and itsnonerythropoietic tissue-protective derivatives have been shownto possess these properties in vitro, as well as in vivo for injured(37–39) and normal (40) animals. Recently, by using functionalMRI, EPO has also been shown to enhance cognition in normalhuman volunteers (4, 41, 42). To determine whether pHBSPmodulates cognitive function, we used the novel object recog-nition paradigm in rats, in which memory recall for previously

experienced objects is evaluated. Specifically, rats were exposedto novel test objects and then reexposed to them 24 h later.Animals having received galantamine (3 mg/kg i.p. 1 h beforetesting; a positive control) displayed enhanced memory reten-tion of previously experienced objects (Fig. 6). Similarly, animalsreceiving pHBSP (24 nmol/kg of bw i.p.) 3 h after the firstexposure to the objects to be learned, or receiving twice dailydoses 5 days before training and continued through the dayimmediately after training, showed enhanced memory for theobjects. In contrast, animals that received pHBSP 1 h before thefirst object exposure did not show enhancement. Because pH-BSP was effective only when administered after training, thismolecule likely acts by intensifying the consolidation phase ofmemory acquisition.

In summary, using a variety of in vitro and in vivo models, wehave shown that helix B of EPO has tissue-protective activitiesrepresentative of the full molecule. Further, a peptide con-structed to mimic the external, aqueous surface of EPO withoutprimary sequence similarity recapitulates EPO’s tissue-protective, neurotrophic, and reparative properties. Peptidedoses that exhibited tissue protection were similar on a molarbasis to those observed for EPO and are higher than thoserequired for EPO-mediated erythropoiesis. For example, in therenal ischemia model, 0.08 nmol/kg of bw (equivalent to �300units/kg of bw of EPO) was ineffective, whereas a 10-fold higherdose elicited strong tissue protection.

Finally, pharmacokinetic studies confirm that pHBSP pos-sesses a plasma half-life of �2 min in the rat and rabbit (see SIMaterials and Methods, Figs. S6 and S7, and Tables S1 and S2).It is especially notable that, similar to asialo-EPO (17), an agentpresent within the circulation for only a short time after i.v.dosing elicits protective effects equivalent to EPO or CEPO withplasma half-lives of 4–6 h. Tissue-protective peptides maytherefore be of use as pharmacological reagents to delineateaspects of timing in tissue protection and trophic effects, inaddition to potentially being of therapeutic benefit in a widevariety of clinical scenarios.

Materials and MethodsThe animal protocols followed in this study were approved by the respectiveAnimal Use and Care Committees of each institution in accordance with thedirectives of the Guide for the Care and Use of Laboratory Animals of theNational Research Council or the Home Office Guidance on the operation ofanimals (Scientific Procedures) Act 1986 published by Her Majesty’s StationeryOffice or in compliance with national (D.L. n. 116, G.U., suppl. 40, Feb. 18,1992) and international laws and policies (EU Council Directive 86/609, OJ L358, 1, Dec. 12, 1987).

Materials. Peptides were obtained from commercial manufacturers. The UT-7EPO hematopoietic assay (10, 17), motoneuron excitotoxicity study (15, 43),

0 2 4 6 8 10

0

3

6

9

12 PBSpHSBP

days

WoundArea[mm2]

Fig. 5. Full-thickness punch biopsy wounds placed over the scapular regionof a rat heal more rapidly after pHBSP (24 nmol/kg of bw) administered dailyvia the s.c. route. n � 9 animals each group. Curves differ at the P � 0.05 levelby repeated-measures analysis.

vehicle

galantam

ine

pHBSP

multiple

pHBSP

post

pHBSP

pre

0

20

40

60

80

100

* * *

RecognitionIndex

Fig. 6. Effect of pHBSP (24 nmol/kg of bw) and the positive control galan-tamine (3 mg/kg of bw) on novel object recognition memory test performedwith adult Wistar rats. Data represent mean and SEM recognition index at the24-h retention test. n � 8 each group; *, P � 0.05 versus saline.

10928 � www.pnas.org�cgi�doi�10.1073�pnas.0805594105 Brines et al.

Page 5: Brines et al PNAS 2008 + suppl materials–note-pHBS is ARA 290

middle cerebral artery occlusion model (44), and sciatic nerve compressioninjury (10) were performed as previously reported.

Diabetic Retinal Edema. Fasting male Sprague–Dawley rats weighing �250 gwere administered streptozotocin (60 mg/kg of bw) i.p., and diabetes wasconfirmed by a fasting blood glucose of �250 mg/dl 2 days later. Diabeticanimals were administered HBP (1.5 nmol/kg of bw) or saline i.p. 5 days aweek, while a third group of normal animals received saline. After 3 weeks,animals were anesthetized by using isoflurane, and Evans blue solution (45mg/kg of bw as a 7.5 mg/ml solution) was injected via the tail vein. Three hourslater, the animals were reanesthetized, a small blood sample was obtained todetermine plasma concentration of Evans blue, and each rat was perfusedwith pH 7.4 citrate buffer at 120 mmHg for 2 min and, thereafter, both eyeswere immediately removed. Under an operating microscope, the eyes werebisected along the equator and the retinas were removed. The retinas weredesiccated at 60°C overnight in a vacuum, weighed, crushed in 120 �l offormamide, and incubated at 70°C for 18 additional hours. The retinal form-amide solution was filter-centrifuged at 15,000 � g for 30 min to removeretinal debris. Evans blue concentration was determined by a background-subtracted absorbance at wavelengths of 620 nm (maximum) and 740 nm(minimum). Fluid extravasation was calculated as Evans blue (�g)/retina dryweight (g). Data were analyzed by analysis of variance (ANOVA) followed byDunnett’s post hoc test comparison.

Renal Ischemia–Reperfusion Model. Sixty male C57/BL6 mice (�25 g; CharlesRiver Laboratories) were anesthetized with ketamine (150 mg/kg) and xyla-zine (15 mg/kg) i.p. Each animal was placed on a homeothermic blanket set at37°C, and after a mid-line laparotomy, the renal pedicles were clamped for 30min by using nontraumatic microvascular clamps. pHBSP was administered atthe indicated dose via i.p. injection at 1 min, 6 h, and 12 h after reperfusion.Twenty-four hours later, mice were reanesthetized and blood was obtainedby cardiac puncture. Plasma urea and creatinine were used as indicators ofrenal dysfunction and aspartate aminotransferase was used as an indicator ofrenal injury. Data were analyzed by ANOVA followed by Dunnett’s post hoctest comparison.

Wound Healing. Methods were adapted from the protocol of Padgett et al.(45). Male Sprague–Dawley rats (�200 g) were fasted from the evening before

the procedure. Under isoflurane anesthesia, a 5 � 5 cm region of skin wasshaved on the dorsum in the subscapular region and washed with povidoneiodine solution. Four full-thickness wounds (3.5-mm diameter) were placed atthe corners of a square of 3-cm sides by using a biopsy punch. The wound edgewas then infiltrated with 1% lidocaine solution and lidocaine-saturated gelfoam was attached with adhesive tape. Wound assessment was obtained byserial digital photographs that included a 3.5-mm diameter standard. Areawas determined by using digital planimetry and the four measurements wereaveraged. Data were analyzed by using a repeated-measures analysis.

Novel Object Recognition in Rats. This model is based on the greater sponta-neous exploration of a novel object, compared with a familiar object, shownby rodents (46). Male Wistar rats were assessed for cognitive ability in a testapparatus comprising an open-field arena placed in a sound-attenuated roomunder dim lighting.

After a 5-min habituation period, each rat was placed into the test arena inthe presence of two identical plastic shapes, and the time spent activelyexploring the objects during a 5-min test period (T1) was recorded. The rat wasreturned to its home cage between tests. After 24 h, each rat was again placedin the test arena for 5 min (T2) in the presence of one of the familiar objectsand a novel object, and the time spent exploring each object was againrecorded. A recognition index for each object, the ratio of the time spentexploring either the familiar object or the novel object over the total timespent exploring both objects (during retention session T2), was used tomeasure cognitive (memory) function.

Rats (n � 8 each group) were treated with the test compounds before thetest period (T1), after T1, or chronically for 5 days before T1, via the i.p. route.Groups consisted of those that received vehicle, galantamine (3 mg/kg of bw)administered 1 h before the first 5-min exposure to the two identical objectsto be learned, pHBSP (24 nmol/kg of bw) administered 1 h before the first5-min exposure to the two identical objects to be learned, pHBSP (24 nmol/kgof bw) administered 3 h after the first 5-min exposure to the two identicalobjects to be learned, or pHBSP (24 nmol/kg of bw) administered every 12 h for5 days before training and then 12 and 24 h after training (the last dose wasadministered 1 h before the novel object exposure). Data were analyzed byANOVA followed by Dunnett’s post hoc test comparison.

ACKNOWLEDGMENTS. We thank Annie Zhu and Deborah Gomez for experttechnical assistance. This work was funded in part by the William HarveyResearch Foundation (N.S.A.P. and C.T.).

1. Fisher JW (2003) Erythropoietin: Physiology and pharmacology update. Exp Biol Med(Maywood) 228:1–14.

2. Brines M, Cerami A (2006) Discovering erythropoietin’s extra-hematopoietic functions:Biology and clinical promise. Kidney Int 70:246–250.

3. Juul SE (2000) Nonerythropoietic roles of erythropoietin in the fetus and neonate. ClinPerinatol 27:527–541.

4. Miskowiak K, O’Sullivan U, Harmer CJ (2007) Erythropoietin enhances hippocampalresponse during memory retrieval in humans. J Neurosci 27:2788–2792.

5. Ehrenreich H, et al. (2007) Improvement of cognitive functions in chronic schizophrenicpatients by recombinant human erythropoietin. Mol Psychiatry 12:206–220.

6. Boissel JP, Lee WR, Presnell SR, Cohen FE, Bunn HF (1993) Erythropoietin structure-function relationships. Mutant proteins that test a model of tertiary structure. J BiolChem 268:15983–15993.

7. Cheetham JC, et al. (1998) NMR structure of human erythropoietin and a comparisonwith its receptor bound conformation. Nat Struct Biol 5:861–866.

8. Elliott S, Lorenzini T, Chang D, Barzilay J, Delorme E (1997) Mapping of the active siteof recombinant human erythropoietin. Blood 89:493–502.

9. Wen D, Boissel JP, Showers M, Ruch BC, Bunn HF (1994) Erythropoietin structure-function relationships. Identification of functionally important domains. J Biol Chem269:22839–22846.

10. Leist M, et al. (2004) Derivatives of erythropoietin that are tissue protective but noterythropoietic. Science 305:239–242.

11. Masuda S, et al. (1993) Functional erythropoietin receptor of the cells with neuralcharacteristics. Comparison with receptor properties of erythroid cells. J Biol Chem268:11208–11216.

12. Brines M, et al. (2004) Erythropoietin mediates tissue protection through an erythro-poietin and common beta-subunit heteroreceptor. Proc Natl Acad Sci USA 101:14907–14912.

13. Murphy JM, Young IG (2006) IL-3, IL-5, and GM-CSF signaling: Crystal structure of thehuman beta-common receptor. Vitam Horm 74:1–30.

14. Syed RS, et al. (1998) Efficiency of signalling through cytokine receptors dependscritically on receptor orientation. Nature 395:511–516.

15. Mennini T, et al. (2006) Nonhematopoietic erythropoietin derivatives prevent mo-toneuron degeneration in vitro and in vivo. Mol Med 12:153–160.

16. Brines ML, et al. (2000) Erythropoietin crosses the blood-brain barrier to protectagainst experimental brain injury. Proc Natl Acad Sci USA 97:10526–10531.

17. Erbayraktar S, et al. (2003) Asialoerythropoietin is a nonerythropoietic cytokinewith broad neuroprotective activity in vivo. Proc Natl Acad Sci USA 100:6741–6746.

18. Cuzzocrea S, et al. (2005) Erythropoietin reduces the degree of arthritis caused by typeII collagen in the mouse. Arthritis Rheum 52:940–950.

19. Okutan O, Turkoglu OF, Gok HB, Beskonakli E (2008) Neuroprotective effect oferythropoietin after experimental cold injury-induced vasogenic brain edema in rats.Surg Neurol, in press.

20. Verdonck O, et al. (2007) Erythropoietin protects from post-traumatic edema in the ratbrain. J Cereb Blood Flow Metab 27:1369–1376.

21. Wu H, et al. (2006) Pretreatment with recombined human erythropoietin attenu-ates ischemia-reperfusion-induced lung injury in rats. Eur J Cardiothorac Surg29:902–907.

22. Zhang J, et al. (2008) Intravitreal injection of erythropoietin protects both retinalvascular and neuronal cells in early diabetes. Invest Ophthalmol Vis Sci 49:732–742.

23. Friedman EA, L’Esperance FA, Brown CD, Berman DH (2003) Treating azotemia-induced anemia with erythropoietin improves diabetic eye disease. Kidney Int SupplS57–S63.

24. D’Andrea LD, et al. (2005) Targeting angiogenesis: Structural characterization andbiological properties of a de novo engineered VEGF mimicking peptide. Proc Natl AcadSci USA 102:14215–14220.

25. Liu H, et al. (2004) Identification of the antivasopermeability effect of pigmentepithelium-derived factor and its active site. Proc Natl Acad Sci USA 101:6605–6610.

26. Kazim AL, Atassi MZ (1980) Antibody combining sites can be mimicked synthetically.Surface-simulation synthesis of the phosphorylcholine-combining site of myelomaprotein M-603. Biochem J 187:661–666.

27. Dong XN, Chen Y, Chen YH (2007) Surface simulation synthesis: A new strategy to spyalpha-helix structure. Vaccine 25:6569–6571.

28. Atassi MZ, Manshouri T (1993) Design of peptide enzymes (pepzymes): Surface-simulation synthetic peptides that mimic the chymotrypsin and trypsin active sitesexhibit the activity and specificity of the respective enzyme. Proc Natl Acad Sci USA90:8282–8286.

29. Matthews BW, Craik CS, Neurath H (1994) Can small cyclic peptides have the activityand specificity of proteolytic enzymes? Proc Natl Acad Sci USA 91:4103–4105.

30. Tregear GW, et al. (1973) Bovine parathyroid hormone: Minimum chain length ofsynthetic peptide required for biological activity. Endocrinology 93:1349–1353.

31. Campana WM, Misasi R, O’Brien JS (1998) Identification of a neurotrophic sequence inerythropoietin. Int J Mol Med 1:235–241.

32. Yu L, et al. (2006) Investigation of N-terminal glutamate cyclization of recombinantmonoclonal antibody in formulation development. J Pharm Biomed Anal 42:455–463.

Brines et al. PNAS � August 5, 2008 � vol. 105 � no. 31 � 10929

MED

ICA

LSC

IEN

CES

Page 6: Brines et al PNAS 2008 + suppl materials–note-pHBS is ARA 290

33. Patel NS, et al. (2004) Pretreatment with EPO reduces the injury and dysfunction causedby ischemia/reperfusion in the mouse kidney in vivo. Kidney Int 66:983–989.

34. Arcasoy MO (2008) The non-haematopoietic biological effects of erythropoietin. Br JHaematol 141:14–31.

35. Buemi M, et al. (2002) Recombinant human erythropoietin influences revasculariza-tion and healing in a rat model of random ischaemic flaps. Acta Derm Venereol 82:411–417.

36. Konishi Y, Chui DH, Hirose H, Kunishita T, Tabira T (1993) Trophic effect of erythro-poietin and other hematopoietic factors on central cholinergic neurons in vitro and invivo. Brain Res 609:29–35.

37. Gonzalez FF, et al. (2007) Erythropoietin enhances long-term neuroprotection andneurogenesis in neonatal stroke. Dev Neurosci 29:321–330.

38. Lu D, et al. (2005) Erythropoietin enhances neurogenesis and restores spatial memoryin rats after traumatic brain injury. J Neurotrauma 22:1011–1017.

39. Wang L, Zhang Z, Wang Y, Zhang R, Chopp M (2004) Treatment of stroke witherythropoietin enhances neurogenesis and angiogenesis and improves neurologicalfunction in rats. Stroke 35:1732–1737.

40. Ransome MI, Turnley AM (2007) Systemically delivered erythropoietin transientlyenhances adult hippocampal neurogenesis. J Neurochem 102:1953–1965.

41. Miskowiak K, et al. (2008) Differential effects of erythropoietin on neural and cogni-tive measures of executive function 3 and 7 days post-administration. Exp Brain Res184:313–321.

42. Miskowiak K, Inkster B, Selvaraj S, Goodwin G, Harmer C (2007) Erythropoietin has noeffect on hippocampal response during memory retrieval 3 days post-administration.Psychopharmacology (Berlin) 195:451–453.

43. De Paola M, et al. (2008) Chemokine MIP-2/CXCL2, acting on CXCR2, induces mo-tor neuron death in primary cultures. Neuroimmunomodulation 14:310–316.

44. Villa P, et al. (2007) Reduced functional deficits, neuroinflammation, and secondarytissue damage after treatment of stroke by nonerythropoietic erythropoietin deriva-tives. J Cereb Blood Flow Metab 27:552–563.

45. Padgett DA, Marucha PT, Sheridan JF (1998) Restraint stress slows cutaneous woundhealing in mice. Brain Behav Immun 12:64–73.

46. Ennaceur A, Delacour J (1988) A new one-trial test for neurobiological studies ofmemory in rats. 1: Behavioral data. Behav Brain Res 31:47–59.

10930 � www.pnas.org�cgi�doi�10.1073�pnas.0805594105 Brines et al.

Page 7: Brines et al PNAS 2008 + suppl materials–note-pHBS is ARA 290

Supporting InformationBrines et al. 10.1073/pnas.0805594105SI Materials and MethodsHematopoietic Potency. Peptides were tested in vitro for hemato-poietic potency by use of the EPO-responsive human erythro-leukemic cell line UT-7 EPO as previously described in detail (1,2). The assay was performed over 48 h, and proliferation wasquantified by using WST-1 reduction (Roche; no. 1644807). Asshown in supporting information (SI) Fig. S1, HBP over therange of 5 pM to 50 nM did not increase cell number, in contrastto the large hematopoietic effect of EPO.

Additional experiments were performed in which the rat orrabbit received repeated injections of pHBSP. Specifically,pHBSP was administered twice daily i.v. to Sprague–Dawley ratsfor 28 days. For this study, nine male and nine female rats wereassigned to Groups 1–4 receiving 0, 60, 180, and 600 �g/kg perdose (0, 48, 143, and 477 nmol/kg, respectively) of pHBSP in PBSby bolus i.v. administration from days 1–28. Blood samples toassay for hematological variables were collected on day 29. Therewas no difference between any of the groups in hemoglobinconcentration (Fig. S2).

Further, pHBSP was administered twice daily i.v. to NewZealand White rabbits for 28 days. For this study, six males andsix females were assigned to Groups 1 and 4, and four male andfour female rabbits were assigned to Groups 2 and 3 receiving 0(Group 1), 30 (24 nmol/kg; Group 2), 90 (72 nmol/kg; Group 3),and 300 (240 nmol/kg; Group 4) �g/kg per dose pHBSP in PBSby bolus i.v. administration. Comparison of baseline versus day29 hematological parameters showed no difference in hemoglo-bin concentration, hematocrit, or platelet count (Figs. S3–S5).Thus, studies carried out in two species confirm that pHBSP isnot erythropoietic in vivo.

Pharmacokinetics of pHBSP. Rat. The pharmacokinetic behavior ofpHBSP was determined after a single i.v. dose to male Sprague–Dawley rats ported with bilateral jugular vein cannulae. Twogroups, each of three rats, received either 60 �g/kg (48 nmol/kg;Group 1) or 180 �g/kg (143 nmol/kg; Group 2) of pHBSP diluted

in PBS. The dose was administered over a period of 10–12 secinto one of two ports, and samples were withdrawn from theother port predose and at 2, 4, 6, 8, 10, 12, 14, 16, and 18 min afteradministration of the dose. Plasma concentrations of pHBSPwere determined by using a liquid chromatography–mass spec-troscopy assay.

Plasma pHBSP concentration as a function of time after doseis shown in Fig. S6. Individual and group mean pharmacokineticparameters are shown in Table S1. The mean peak drug con-centration (Cmax) values were 254.67 (�53.59) and 1,103.67(�194.53) ng/ml for Groups 1 and 2, respectively. Both Cmax andarea under the concentrations vs. time curve (area under thecurve; AUC) increased with increasing dose in a slightly morethan dose-proportional manner, although the variability be-tween animals within each dose group was high. The mean t1�2

was 0.028 h for Group 1 and 0.047 h for Group 2.Rabbit. The pharmacokinetic behavior of pHBSP was determinedafter a single i.v. dose to male New Zealand White rabbits. Twogroups, each with three rabbits, received either 30 �g/kg (24nmol/kg; Group 1) or 90 �g/kg (72 nmol/kg; Group 2) of pHBSPdiluted in PBS. The dosing solution was administered over aperiod of 10–12 sec via ear vein. Blood samples were collectedfrom the contralateral ear by venipuncture of a central auricularartery or a marginal vein. Samples were withdrawn predose andat 2, 4, 6, 8, 10, 12, 14, 16, and 18 min postdose. Plasma pHBSPconcentration as a function of time after dose is shown in Fig. S7.Individual and group mean pharmacokinetic parameters areshown in Table S2.

The mean Cmax values were 95.10 (�44.34) and 200.67(�52.92) ng/ml for Groups 1 and 2, respectively. Both Cmax andAUC increased with increasing dose in a less than dose-proportional manner. Similar to the observations in rats, vari-ability in measured plasma drug concentrations in rabbits washigh with near overlap between Cmax and AUC values betweenanimals in the two dose groups. The mean t1�2

was 0.028 h forGroup 1 and 0.038 h for Group 2.

1. Erbayraktar S, et al. (2003) Asialoerythropoietin is a nonerythropoietic cytokine withbroad neuroprotective activity in vivo. Proc Natl Acad Sci USA 100:6741–6746.

2. Leist M, et al. (2004) Derivatives of erythropoietin that are tissue protective but noterythropoietic. Science 305:239–242.

Brines et al. www.pnas.org/cgi/content/short/0805594105 1 of 10

Page 8: Brines et al PNAS 2008 + suppl materials–note-pHBS is ARA 290

Fig. S1. Peptide HBP has no effect on cell number in the UT-7 EPO assay. In contrast, EPO promotes cell growth and is, therefore, hematopoietic.

Brines et al. www.pnas.org/cgi/content/short/0805594105 2 of 10

Page 9: Brines et al PNAS 2008 + suppl materials–note-pHBS is ARA 290

Fig. S2. pHBSP is not erythropoietic in vivo. Sprague–Dawley rats administered peptide twice daily i.v. at the indicated dosages did not exhibit changes inhemoglobin concentration over a 28-day period.

Brines et al. www.pnas.org/cgi/content/short/0805594105 3 of 10

Page 10: Brines et al PNAS 2008 + suppl materials–note-pHBS is ARA 290

Fig. S3. Rabbits administered pHBSP twice daily i.v. did not exhibit changes in hemoglobin concentration over 28 days of administration.

Brines et al. www.pnas.org/cgi/content/short/0805594105 4 of 10

Page 11: Brines et al PNAS 2008 + suppl materials–note-pHBS is ARA 290

Fig. S4. No change in the hematocrit was observed in rabbits administered pHBSP twice daily for 28 days.

Brines et al. www.pnas.org/cgi/content/short/0805594105 5 of 10

Page 12: Brines et al PNAS 2008 + suppl materials–note-pHBS is ARA 290

Fig. S5. Platelet count did not change over 28 days after the administration of pHBSP twice daily to rabbits.

Brines et al. www.pnas.org/cgi/content/short/0805594105 6 of 10

Page 13: Brines et al PNAS 2008 + suppl materials–note-pHBS is ARA 290

Fig. S6. Mean plasma levels of pHBSP in rats over time after a single dose at the indicated amount. The mean half-life in the rat was estimated to be �2 min.

Brines et al. www.pnas.org/cgi/content/short/0805594105 7 of 10

Page 14: Brines et al PNAS 2008 + suppl materials–note-pHBS is ARA 290

Fig. S7. Mean plasma levels in rabbits over time after a single dose of pHBSP at the indicated amount. The mean half-life in the rabbit was estimated to be�2 min.

Brines et al. www.pnas.org/cgi/content/short/0805594105 8 of 10

Page 15: Brines et al PNAS 2008 + suppl materials–note-pHBS is ARA 290

Table S1. Pharmacokinetic parameters after a single i.v. bolus dose of pHBSP in rats

Subject Body wt, kg Dose, �g/kg Tmax, hr Cmax, ng/ml AUC LAST, h�ng/ml AUC inf, h�ng/ml t1⁄2, h

1 0.282 60.00 0.033 281.00 26.43 26.52 0.0152 0.251 60.00 0.033 290.00 34.19 34.29 0.0173 0.288 60.00 0.033 193.00 18.70 18.93 0.050

Mean 0.27 60.00 0.033 254.67 26.44 26.58 0.028SD 0.02 53.59 7.74 7.68 0.020

4 0.283 180.00 0.033 911.00 80.10 80.99 0.0635 0.294 180.00 0.033 1,300.00 134.08 134.44 0.0456 0.318 180.00 0.033 1,100.00 96.37 96.75 0.034

Mean 0.30 180.00 0.033 1,103.67 103.52 104.06 0.047SD 0.02 194.53 27.69 27.47 0.014

Nominal time and dosage used for pharmacokinetic analysis. Median is calculated for Tmax., the time at which Cmax (maximum observed plasma concentration)occurs. AUC LAST, area under the curve from time 0 to last measured concentration; AUC inf, area under the curve from time 0 to infinity.

Brines et al. www.pnas.org/cgi/content/short/0805594105 9 of 10

Page 16: Brines et al PNAS 2008 + suppl materials–note-pHBS is ARA 290

Table S2. Pharmacokinetic parameters after a single i.v. bolus dose of pHBSP in rabbits

Subject Body wt, kg Dose, �g/kg Tmax, h Cmax, ng/ml AUC LAST, h�ng/ml AUC inf, h�ng/ml t1⁄2, h

1 3.51 30.00 0.033 126.00 16.33 16.50 0.0272 3.00 30.00 0.033 115.00 16.36 16.52 0.0293 3.07 30.00 0.033 44.30 4.72 * *

Mean 3.19 30.00 0.033 95.10 12.47 16.51 0.028SD 0.28 44.34 6.71 0.01 0.002

4 2.99 90.00 0.033 212.00 22.20 22.33 0.0395 3.08 90.00 0.033 247.00 30.64 30.78 0.0456 3.07 90.00 0.033 143.00 18.03 18.17 0.030

Mean 3.05 90.00 0.033 200.67 23.63 23.76 0.038SD 0.05 52.92 6.43 6.42 0.008

Nominal time and dosage used for pharmacokinetic analysis. Median is calculated for Tmax.*Not enough data available to calculate given parameter.

Brines et al. www.pnas.org/cgi/content/short/0805594105 10 of 10