transglutaminases and neurodegeneration

13
Transglutaminases and neurodegeneration Thomas M. Jeitner * , John T. Pinto , Boris F. Krasnikov , Mark Horswill , and Arthur J. L. Cooper * Red Anvil, LLC, Milwaukee, Wisconsin, USA Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, New York, USA Developmental Vascular Biology Program, Medical College of Wisconsin, Milwaukee, Wisconsin, USA Abstract Transglutaminases (TGs) are Ca 2+ -dependent enzymes that catalyze a variety of modifications of glutaminyl (Q) residues. In the brain, these modifications include the covalent attachment of a number of amine-bearing compounds, including lysyl (K) residues and polyamines, which serve to either regulate enzyme activity or attach the TG substrates to biological matrices. Aberrant TG activity is thought to contribute to Alzheimer disease, Parkinson disease, Huntington disease, and supranuclear palsy. Strategies designed to interfere with TG activity have some benefit in animal models of Huntington and Parkinson diseases. The following review summarizes the involvement of TGs in neurodegenerative diseases and discusses the possible use of selective inhibitors as therapeutic agents in these diseases. Keywords cystamine; neurodegeneration; polyamines; transglutaminase; γ-glutamylpolyamines; γ-glutamyl-ε- lysine Cerebral transglutaminases Eight active transglutaminases (TGs) (TGs 1–7 and factor XIIIa) are expressed in mammals, of which TGs 1–3 (Kim et al. 1999) 1 and 6 (Hadjivassilou et al. 2008) are present in human brain. The major reaction thus far attributed to the cerebral TGs is transamidation. In this reaction the carboxamide moiety of a Q residue [-C(O)NH 2 ] is converted to a substituted carboxamide [-C(O)NHR] by nucleophilic attack of an amine [RNH 2 ] such as various mono-, di-, and polyamines or the ε amino group of a K residue (Lorand and Graham 2003). Of the possible transamidation linkages, the γ-glutamyl-ε-lysine [N ε -(γ-L-glutamyl)-L-lysine] (GGEL) isopeptide linkage formed between Q and K resides, is the most commonly studied. GGEL bonds occur both within and between polypeptide chains, and thereby contribute to the formation of stable soluble and insoluble polymers. TGs also cross-link proteins via bis-γ- glutamylpolyamine bridges between Q residues (Piacentini et al. 1988). These linkages are formed by two successive transamidations: the first utilizes a free polyamine to generate a γ- glutamylpolyamine residue, which becomes the amine-bearing substrate for a second Address correspondence and reprint requests to Dr Arthur J. L. Cooper, Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, NY 10595, USA. [email protected]. Conflicts of interest: All authors declare no conflicts of interests. 1 Given the need for brevity, the number of citations has been restricted/limited. NIH Public Access Author Manuscript J Neurochem. Author manuscript; available in PMC 2009 September 28. Published in final edited form as: J Neurochem. 2009 May ; 109(Suppl 1): 160–166. doi:10.1111/j.1471-4159.2009.05843.x. NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript

Upload: independent

Post on 27-Nov-2023

0 views

Category:

Documents


0 download

TRANSCRIPT

Transglutaminases and neurodegeneration

Thomas M. Jeitner*, John T. Pinto†, Boris F. Krasnikov†, Mark Horswill‡, and Arthur J. L.Cooper†*Red Anvil, LLC, Milwaukee, Wisconsin, USA†Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, NewYork, USA‡Developmental Vascular Biology Program, Medical College of Wisconsin, Milwaukee, Wisconsin,USA

AbstractTransglutaminases (TGs) are Ca2+-dependent enzymes that catalyze a variety of modifications ofglutaminyl (Q) residues. In the brain, these modifications include the covalent attachment of a numberof amine-bearing compounds, including lysyl (K) residues and polyamines, which serve to eitherregulate enzyme activity or attach the TG substrates to biological matrices. Aberrant TG activity isthought to contribute to Alzheimer disease, Parkinson disease, Huntington disease, and supranuclearpalsy. Strategies designed to interfere with TG activity have some benefit in animal models ofHuntington and Parkinson diseases. The following review summarizes the involvement of TGs inneurodegenerative diseases and discusses the possible use of selective inhibitors as therapeutic agentsin these diseases.

Keywordscystamine; neurodegeneration; polyamines; transglutaminase; γ-glutamylpolyamines; γ-glutamyl-ε-lysine

Cerebral transglutaminasesEight active transglutaminases (TGs) (TGs 1–7 and factor XIIIa) are expressed in mammals,of which TGs 1–3 (Kim et al. 1999)1 and 6 (Hadjivassilou et al. 2008) are present in humanbrain. The major reaction thus far attributed to the cerebral TGs is transamidation. In thisreaction the carboxamide moiety of a Q residue [-C(O)NH2] is converted to a substitutedcarboxamide [-C(O)NHR] by nucleophilic attack of an amine [RNH2] such as various mono-,di-, and polyamines or the ε amino group of a K residue (Lorand and Graham 2003). Of thepossible transamidation linkages, the γ-glutamyl-ε-lysine [Nε-(γ-L-glutamyl)-L-lysine] (GGEL)isopeptide linkage formed between Q and K resides, is the most commonly studied. GGELbonds occur both within and between polypeptide chains, and thereby contribute to theformation of stable soluble and insoluble polymers. TGs also cross-link proteins via bis-γ-glutamylpolyamine bridges between Q residues (Piacentini et al. 1988). These linkages areformed by two successive transamidations: the first utilizes a free polyamine to generate a γ-glutamylpolyamine residue, which becomes the amine-bearing substrate for a second

Address correspondence and reprint requests to Dr Arthur J. L. Cooper, Department of Biochemistry and Molecular Biology, New YorkMedical College, Valhalla, NY 10595, USA. [email protected] of interest: All authors declare no conflicts of interests.1Given the need for brevity, the number of citations has been restricted/limited.

NIH Public AccessAuthor ManuscriptJ Neurochem. Author manuscript; available in PMC 2009 September 28.

Published in final edited form as:J Neurochem. 2009 May ; 109(Suppl 1): 160–166. doi:10.1111/j.1471-4159.2009.05843.x.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

transamidation. bis-γ-Glutamylpolyamine cross-links are formed at least as frequently as thoseinvolving GGEL (Piacentini et al. 1988). Under physiological conditions, the majority of cross-linking bonds are generated outside of cells where the concentrations of Ca2+ are sufficientlyhigh enough to stimulate the catalysis of these bonds by TGs. TG 2 and 3 have three Ca2+

binding sites and the current indications are that the catalysis of GGEL bonds requires theoccupation of all three sites (Datta et al. 2006).

Intracellular Ca2+ concentrations rarely match the extracellular concentrations. Moreover, GTPacts in cells as an endogenous inhibitor of TGs (Bergamini et al. 1987). Nevertheless,intracellular TG-catalyzed reaction products can be detected in normal cells, especially thoseproducts related to polyamination (Piacentini et al. 1988). The consequences of polyaminationin the brain, however, are poorly understood as only a limited number of polyaminated proteinshave been identified (Tucholski et al. 1999). Of these, phospholipase A2 (PLA2) is especiallyinteresting since the polyamination of this enzyme may contribute to the inflammationassociated with neurodegeneration. PLA2 produces two groups of pro-inflammatorymediators: leukotrienes and prostaglandins. Polyamination of PLA2 results in a 3-fold increasein activity (Cordella-Miele et al. 1993) that may persist for the life of the protein given theinability of most peptidases to hydrolyze γ-glutamylamine (GGEL, γ-glutamylpolyamine andbis-γ-glutamylpolyamine) linkages (Fink and Folk 1981). Thus, polyamination may representa unique post-translational modification of enzymes that permanently affects activity. Thissituation contrasts with other types of covalent post-translational modifications, such asphosphorylation that typically are transient.

Increased TG(s) in neurodegenerative diseasesTransglutaminase activity is widespread in brain (Kim et al. 1999) and is present in primarycultures of neurons and astrocytes (Perry et al. 1995; Caccamo et al. 2004). TG 2 is associatedwith the extracellular matrix, cell membranes and cytosol of neurons, and TG activity has beenidentified in synaptosomes (Pastuszko et al. 1986), mitochondria (Krasnikov et al. 2005), andnuclei (Lesort et al. 1998). The activity, expression and amounts of individual TG enzymesare increased in a variety of neurodegenerative diseases. TG activity is significantly elevatedin the affected cerebral regions in Alzheimer disease (AD) (Johnson et al. 1997; Kim et al.1999), Huntington disease (HD) (Karpuj et al. 1999; Lesort et al. 1999), and supranuclear palsy(Zemaitaitis et al. 2003). These increases in activity are accompanied by gains in the amountof TG 1 and TG 2 proteins in AD brain (Kim et al. 1999; Bonelli et al. 2002), and also of TG2 protein in the brains of HD (Lesort et al. 1999) and supranuclear palsy (Zemaitaitis et al.2003) patients. Increased TG 2 protein is also found in the CSF of AD (Bonelli et al. 2002)and Parkinson disease (PD) (Vermes et al. 2004) patients.

Not only are the amounts of TG increased in AD, HD and supranuclear palsy, but the conditionsfavoring the activation of these enzymes are also enhanced in these diseases. These conditionsinclude elevations in intracellular Ca2+ due to glutamate-mediated excitotoxity (Caccamo etal. 2004) and other perturbations in Ca2+ homeostasis (Mattson 2007) as well as decreases inGTP concentrations following from losses in energy production (Lin and Beal 2006).

The number of TG 2 transcripts is also increased in HD (Lesort et al. 1999) and supranuclearpalsy (Zemaitaitis et al. 2003), and a shortened alternate transcript of TG 2 encoding a formof enzyme missing the GTP binding domain is expressed in AD brain (Festoff et al. 2002). Anumber of mechanisms may account for the increased transcription and translation of TGs inneurodegenerative disorders. The TG 1 promoter has Ca2+ (Kawabe et al. 1998), retinoid(Polakowska et al. 1999), cAMP, Sp1, and AP1 responsive elements (Medvedev et al. 1999),while the TG 2 promoter contains elements that respond to retinoids (Nagy et al. 1996; Yanet al. 1996), interleukin 6, transforming growth factor β1 (Ritter and Davies 1998), and tumor

Jeitner et al. Page 2

J Neurochem. Author manuscript; available in PMC 2009 September 28.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

necrosis factor-α (Kuncio et al. 1998). The inflammatory mediators are likely to act via theNF-κB (Nuclear factor κB) binding region in the TG 2 promoter (Kuncio et al. 1998; Kim etal. 2008). NF-κB translocation and DNA binding are stimulated by tumor necrosis factor-αand glutamate, both of which have been shown to increase TG 2 expression in microglia andastrocytes (Campisi et al. 2004; Park et al. 2004). As noted earlier, inflammation accompaniesneurodegeneration and TGs may contribute to this response via the sustained activation ofpolyaminated PLA2. The possibility that TGs may contribute to their continued activationhighlights the necessity of limiting the activity of these enzymes in neurodegenerativedisorders.

Increased TG-catalyzed products in neurodegenerative diseasesIncreased TG activity in neurodegenerative disorders is accompanied by an increase in TG-catalyzed products. Selkoe et al. (1982a,b) demonstrated that cerebral TGs catalyze the invitro polymerization of cytoskeletal elements, and hypothesized that TGs might facilitatepaired helical formation in AD tangles. TG 2 and GGEL cross-links were subsequently shownto co-localize with the tangles (Miller and Anderton 1986; Johnson et al. 1997) and TG2 wasshown to co-localize with plaques (Zhang et al. 1998) in AD brain. Components of the plaquesor tangles, including β-amyloid (Aβ) (Ikura et al. 1993; Dudek and Johnson 1994; Ho et al.1994; Rasmussen et al. 1994), the Dutch mutation of Aβ (Q22 → E22) (Dudek and Johnson1994), tau (Miller and Anderton 1986; Dudek and Johnson 1993; Miller and Johnson 1995;Appelt and Balin 1997; Murthy et al. 1998; Tucholski et al. 1999), and the non-Aβ componentderived from α-synuclein (Jensen et al. 1995) are TG substrates. The in vitro products of thereaction of these substrates with TG bear a striking resemblance to the insoluble polymersfound in AD brain (Jensen et al. 1995; Appelt and Balin 1997; Hartley et al. 2008).

Huntington disease is caused by a CAG expansion in the huntingtin (htt) gene that encodes alength of contiguous Q residues [polyglutamine (Qn)] in the N-terminus of the expressedprotein. Green (1993) hypothesized that the expanded Qn region would favor the formation ofTG-catalyzed GGEL linkages and lead to the formation of htt-containing aggregates. In supportof this hypothesis, expanded Qn domains are excellent TG substrates (Kahlem et al. 1996;Cooper et al. 1997a; Gentile et al. 1998; Lesort et al. 1999; Zainelli et al. 2005) and mutanthtt is present in HD aggregates (DiFiglia et al. 1995) as are GGEL crosslinks (Zainelli et al.2003).

The increased cerebral aggregation seen in HD, PD, and supranuclear palsy is also associatedwith a comparable increase in GGEL immunoreactivity within the polymers (Zemaitaitis etal. 2000; Zainelli et al. 2003; Andringa et al. 2004). Although some concerns have been raisedabout the specificity of GGEL antibodies in immunoblots (Johnson and LeShoure 2004), theincrease in protein-associated GGEL in AD brain has been unequivocally confirmed usingmass spectrometric techniques (Kim et al. 1999; Nemes et al. 2004).

As noted earlier, the isopeptide bonds in γ-glutamylamine linkages are resistant to proteolysis(Fink and Folk 1981). Moreover, the ability to metabolize free γ-glutamylamines in brain islimited. Consequently, γ-glutamylamines are excised intact during proteolysis and are presentin brain and CSF (Jeitner et al. 2001, 2008; Dedeoglu et al. 2002). A several-fold increase infree GGEL has been measured in the brains of HD patients (Dedeoglu et al. 2002), and theamount of GGEL in the CSF of patients with AD, PD (Sárvari et al. 2002), or HD (Jeitner etal. 2001, 2008; Dedeoglu et al. 2002) is also increased relative to control CSF. The increasein CSF GGEL reported in HD is also matched by comparable increases in the amounts of CSFγ-glutamylspermidine, γ-glutamylputrescine and bis-γ-glutamylputrescine (Jeitner et al.2008). CSF contains higher (μM) quantities of γ-glutamylspermidine than GGEL (< μM) in

Jeitner et al. Page 3

J Neurochem. Author manuscript; available in PMC 2009 September 28.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

accord with the suggestion noted above that TGs predominantly catalyze polyamination overQ → K cross-linking within the brain.

Possible mechanisms for TG-mediated neurotoxicityAlthough the formation of insoluble protein aggregates has been proposed to account for thetoxic actions of TGs, the role of such aggregates in the etiology of diseases such as HD iscontroversial (Kuemmerle et al. 1999; Sieradzan and Mann 2001). Indeed, mice that lackedTG 2 and over-express mutant htt had 30% more brain aggregates and still lived longer thantheir TG 2- and mutant htt-expressing littermates (Mastroberardino et al. 2002). The increasedaggregation is unlikely to have been due to compensatory cross-linking by TG 1 and 3, sincethe TG 2-deficient mice also exhibited a 10-fold decrease in the number of GGEL linkages. Itwas subsequently shown that GGEL cross-links serve to produce soluble Qn aggregates,whereas polyaminated or unmodified Qn domains spontaneously aggregate to form insolublepolymers (Lai et al. 2004; Konno et al. 2005). These observations suggest that TG 2-catalyzedGGEL bond formation generates soluble aggregates that may be neurotoxic.

Another possibility is that rather than being toxic per se, the soluble and insoluble polymerscause neuronal death by sequestering critical proteins within the aggregates. These proteinscould include, for example, glyceraldehyde 3-phosphate dehydrogenase, α-ketoglutaratedehydrogenase, and histones (Cooper et al. 1997b, 2000; Gentile et al. 1998) in HD, andubiquitin, HSP27, parkin, and α-synuclein in AD (Nemes et al. 2004). It has also been suggestedthat congestion of proteasomes may contribute to CAG-expansion and other neurodegenerativediseases (Cooper et al. 2002; Wang et al. 2008). In support of this hypothesis, components ofthe ubiquitin proteasome system are found in HD aggregates (Bennett et al. 2007).

The above observations suggest the following model for the contribution of TGs toneurodegenerative diseases. Early in these diseases, TGs predominantly catalyzepolyamination reactions. As these diseases progress, TGs begin to form more GGEL cross-links, which stabilize soluble toxic protein aggregates, eventually leading to removal of keyproteins and to a fatal congestion of proteasomes.

TGs as potential therapeutic targets in neurodegenerative diseasesSeveral authors have raised the possibility that TG inhibitors may be of therapeutic benefit inneurodegenerative diseases (e.g. Cooper et al. 2002; Gentile and Cooper 2004), and one suchin vitro inhibitor – cystamine – is beneficial in murine models of HD and PD (e.g. Dedeogluet al. 2002; Van Raamsdonk et al. 2005; Stack et al. 2008). We have shown that cysteamine,the reduced form of cystamine, is a competitive inhibitor/alternative substrate of TG 2 (Jeitneret al. 2005). Cysteamine attenuates polyamination by acting as an alternative TG 2 substrate,presumably forming Nβ-(γ-L-glutamyl)-cysteamine linkages (Jeitner et al. 2005). In additionto cysteamine, cystamine is metabolized to hypotaurine and taurine, and cystamine treatmentin mice leads to increased brain cysteine levels (Fox et al. 2004; Pinto et al. 2005). We testedthe ability of hypotaurine, taurine, cysteine, and cysteamine to inhibit TG 2. Of the testedcompounds, only cysteamine was able to inhibit TG 2-catalyzed polyamination (Fig. 1). Asneither cystamine nor cysteamine can be detected (detection limit ≤ 20 μM) in the brains ofcystamine-treated YAC128 (HD) mice (Pinto et al. 2005), the conversion of cystamine tocysteamine, and then to Nβ-(γ-L-glutamyl)-cysteamine, is likely to be rapid.

Prolonged cystamine treatment results in decreased TG activity (Dedeoglu et al. 2002; VanRaamsdonk et al. 2005), even though cysteamine is not an irreversible TG inhibitor (Jeitneret al. 2005). Thus, another mechanism must account for the diminished TG activity. Wehypothesize that cystamine-derived cysteamine inhibits the binding of transcription factors toTG promoters and thereby limits the transcription of TGs. In support of this hypothesis,

Jeitner et al. Page 4

J Neurochem. Author manuscript; available in PMC 2009 September 28.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

cysteamine attenuates the DNA binding of AP1 and NF-κB (Goldstone et al. 1995), and bindingsites for these factors are present in the TG 1 and TG 2 promoters (Kuncio et al. 1998;Medvedev et al. 1999; Kim et al. 2008).

As indicated above, cystamine has multiple biological actions in the brain, including raisingthe levels of cysteine (Fox et al. 2004; Pinto et al. 2005). Cystamine also causes elevation ofbrain-derived neurotropic factor (Borrell-Pages et al. 2006) and possibly attenuates apoptosisthrough inhibition of caspase 3 activity (Lesort et al. 2003). Recently, we discovered anotherpotentially beneficial property of cystamine. Cystamine, at concentrations as low as 15 μM,significantly attenuated dopamine-induced macroautophagy in SH SY5Y cells, whereascysteamine had no effect (Fig. 2). Dopamine reduced the viability of SH SY5Y cells by 48 ±3% (mean ± SEM, n = 11) after 24 h, while the combination of dopamine and cystamine (15μM) only reduced viability by 24 ± 2% (p < 0.05, paired t-test). In these experiments, the cellswere pre-treated with cystamine for 2 h, and then treated for a further 24 h (i.e. 26 h).Importantly, the treatment of cells with cystamine for only 4 h prior to the application ofdopamine was as effective as the 26-h cystamine treatment (Fig. 2). This observation suggeststhat cystamine primes the cells against the induction of macroautophagy.

Given the multiplicity of biological activities attributed to cyst(e)amine, it is difficult to assignthe therapeutic benefit of this agent to the inhibition of TGs per se. Moreover, excesscysteamine has been reported to be harmful in at least one setting. Thus, Frankel and Schipper(1999) noted that cysteamine induces the appearance of iron-rich (peroxidase-positive)cytoplasmic inclusions in cultured rat astroglia, which are identical to glial inclusions thatprogressively accumulate in substantia nigra and other subcortical brain regions with advancingage.

The positive results obtained with a mouse model of HD in which the animals lacked TG 2 area more compelling argument for the involvement of TGs in neurodegeneration(Mastroberardino et al. 2002) than the results obtained with cystamine. In this regard, severalgroups are actively synthesizing more selective TG inhibitors than cystamine as possibletherapeutic agents. These inhibitors include dihydroisoxazole derivatives, peptide-bound1,2,4-thiadiazoles, peptides containing diazo-5-oxo-L-norleucine in place of glutamine, α,β-unsaturated amides and epoxides (Pardin et al. 2008a,b). Pardin et al. have recently focusedtheir attention on trans-cinnamoyl benzotriazole amides and 3-(substituted cinnamoyl)pyridines [azachalcones], which are potent reversible TG inhibitors (Pardin et al. 2008a,b),and have discovered a triazole compound that inhibits guinea pig TG 2 with a Ki value of∼170 nM (Pardin et al. 2008b). Stein and colleagues have discovered another series ofreversible TG inhibitors that are thieno[2,3-d]pyrimidin-4-one acylhyd-razide derivatives(Duval et al. 2005; Case and Stein 2007).

Finally, Sohn et al. (2003) have developed a novel strategy that blocks the polyamination ofPLA2. This group noted that uteroglobin and lipocortin-1 contain common sequences thatantagonize the interaction of TG 2 with PLA2. Synthesized variants of these sequencesprevented both the polyamination of PLA2 and experimentally-induced allergic conjunctivitisin experimental animals. These results suggest that rather than inhibiting TG 2 directly, somebenefit may be derived from targeting the interaction of TGs and specific pathogenic TGsubstrates. Polyaminated PLA2 may be one such target in neurodegenerative disorders.

Conclusion and future prospectsAlthough TGs do not cause neurodegenerative diseases directly, the current evidence suggeststhat this family of enzymes contributes to the neuropathology once the disease process hasbegun. It is anticipated that potent TG inhibitors will soon be evaluated for their therapeutic

Jeitner et al. Page 5

J Neurochem. Author manuscript; available in PMC 2009 September 28.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

potential in cellular and animal models of HD and other neurodegenerative diseases. Care willbe required to ensure that these inhibitors are sufficiently selective so as not to affect crucialTG reactions critical to normal metabolic processes or to inhibit blood clot formation.

AcknowledgmentsPart of the authors' work cited herein was supported by the National Institutes of Health grant PO1 AG14930.

AbbreviationsAD

Alzheimer disease

Aβ β-amyloid

GGEL γ-glutamyl-ε-lysine [Nε-(γ-L-glutamyl)-L-lysine]

HD Huntington disease

htt huntingtin

PD Parkinson disease

PLA2 phospholipase A2

Qn polyglutamine

TG transglutaminase

ReferencesAndringa G, Lam KY, Chegary M, Wang X, Chase TN, Bennett MC. Tissue transglutaminase catalyzes

the formation of α-synuclein crosslinks in Parkinson's disease. FASEB J 2004;18:932–934. [PubMed:15001552]

Appelt DM, Balin BJ. The association of tissue transglutaminase with human recombinant tau results inthe formation of insoluble filamentous structures. Brain Res 1997;745:21–31. [PubMed: 9037390]

Bennett EJ, Shaler TA, Woodman B, Ryu KY, Zaitseva TS, Becker CH, Bates GP, Schulman H, KopitoRR. Global changes to the ubiquitin system in Huntington's disease. Nature 2007;448:704–708.[PubMed: 17687326]

Bergamini CM, Signorini M, Poltronieri L. Inhibition of erythrocyte transglutaminase by GTP. BiochimBiophys Acta 1987;916:149–151. [PubMed: 2889472]

Bonelli RM, Aschoff A, Niederwieser G, Heuberger C, Jirikowski G. Cerebrospinal fluid tissuetransglutaminase as a biochemical marker for Alzheimer's disease. Neurobiol Dis 2002;11:106–110.[PubMed: 12460550]

Borrell-Pages M, Canals JM, Cordelieres FP, et al. Cystamine and cysteamine increase brain levels ofBDNF in Huntington disease via HSJ1b and transglutaminase. J Clin Invest 2006;116:1410–1424.[PubMed: 16604191]

Jeitner et al. Page 6

J Neurochem. Author manuscript; available in PMC 2009 September 28.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Caccamo D, Campisi A, Curro M, Li Volti G, Vanella A, Ientile R. Excitotoxic and post-ischemicneurodegeneration: involvement of transglutaminases. Amino Acids 2004;27:373–379. [PubMed:15365907]

Campisi A, Caccamo D, Li Volti G, Curro M, Parisi G, Avola R, Vanella A, Ientile R. Glutamate-evokedredox state alterations are involved in tissue transglutaminase upregulation in primary astrocytecultures. FEBS Lett 2004;578:80–84. [PubMed: 15581620]

Case A, Stein RL. Kinetic analysis of the interaction of tissue transglutaminase with a nonpeptidic slow-binding inhibitor. Biochemistry 2007;46:1106–1115. [PubMed: 17240993]

Cooper AJL, Sheu KF, Burke JR, Onodera O, Strittmatter WJ, Roses AD, Blass JP. Polyglutaminedomains are substrates of tissue transglutaminase: does transglutaminase play a role in expandedCAG/poly-Q neurodegenerative diseases? J Neurochem 1997a;69:431–434. [PubMed: 9202340]

Cooper AJL, Sheu KR, Burke JR, Onodera O, Strittmatter WJ, Roses AD, Blass JP. Transglutaminase-catalyzed inactivation of glyceraldehyde 3-phosphate dehydrogenase and α-ketoglutaratedehydrogenase complex by polyglutamine domains∼of pathological length. Proc Natl Acad Sci USA1997b;94:12604–12609. [PubMed: 9356496]

Cooper AJL, Wang J, Pasternack R, Fuchsbauer HL, Sheu RK, Blass JP. Lysine-rich histone (H1) is alysyl substrate of tissue transglutaminase: possible involvement of transglutaminase in the formationof nuclear aggregates in (CAG)n/Qn expansion diseases. Dev Neurosci 2000;22:404–417. [PubMed:11111157]

Cooper AJL, Jeitner TM, Gentile V, Blass JP. Cross linking of polyglutamine domains catalyzed by tissuetransglutaminase is greatly favored with pathological-length repeats: does transglutaminase activityplay a role in (CAG)n/Qn-expansion diseases? Neurochem Int 2002;40:53–67. [PubMed: 11738472]

Cordella-Miele E, Miele L, Beninati S, Mukherjee AB. Transglutaminase-catalyzed incorporation ofpolyamines into phospholipase A2. J Biochem 1993;113:164–173. [PubMed: 8096844]

Datta S, Antonyak MA, Cerione RA. Importance of Ca2+-dependent transamidation activity in theprotection afforded by tissue transglutaminase against doxorubicin-induced apoptosis. Biochemistry2006;45:13163–13174. [PubMed: 17073438]

Dedeoglu A, Kubilus JK, Jeitner TM, et al. Therapeutic effects of cystamine in a murine model ofHuntington's disease. J Neurosci 2002;22:8942–8950. [PubMed: 12388601]

DiFiglia M, Sapp E, Chase K, et al. Huntingtin is a cytoplasmic protein associated with vesicles in humanand rat brain neurons. Neuron 1995;14:1075–1081. [PubMed: 7748555]

Dudek SM, Johnson GVW. Transglutaminase catalyzes the formation of sodium dodecyl sulfate-insoluble, Alz-50-reactive polymers of tau. J Neurochem 1993;61:1159–1162. [PubMed: 8103081]

Dudek SM, Johnson GVW. Transglutaminase facilitates the formation of polymers of the beta-amyloidpeptide. Brain Res 1994;651:129–133. [PubMed: 7922559]

Duval E, Case A, Stein RL, Cuny GD. Structure-activity relationship study of novel tissuetransglutaminase inhibitors. Bioorg Med Chem Lett 2005;15:1885–1889. [PubMed: 15780627]

Festoff BW, SantaCruz K, Arnold PM, Sebastian CT, Davies PJA, Citron BA. Injury-induced “switch”from GTP-regulated to novel GTP-independent isoform of tissue transglutaminase in the rat spinalcord. J Neurochem 2002;81:708–718. [PubMed: 12065630]

Fink ML, Folk JE. γ-Glutamylamine cyclotransferase. An enzyme involved in the catabolism of ε-(γ-glutamyl)lysine and other gamma-glutamylamines. Mol Cell Biochem 1981;38(Spec No):59–67.[PubMed: 6117008]

Fox JH, Barber DS, Singh B, et al. Cystamine increases L-cysteine levels in Huntington's diseasetransgenic mouse brain and in a PC12 model of polyglutamine aggregation. J Neurochem2004;91:413–422. [PubMed: 15447674]

Frankel D, Schipper HM. Cysteamine pretreatment of the astroglial substratum (mitochondrial ironsequestration) enhances PC12 cell vulnerability to oxidative injury. Exp Neurol 1999;160:376–385.[PubMed: 10619554]

Gentile V, Cooper AJL. Transglutaminases - possible drug targets in human diseases. Curr Drug TargetsCNS Neurol Disord 2004;3:99–104. [PubMed: 15078184]

Gentile V, Sepe C, Calvani M, Melone MA, Cotrufo R, Cooper AJL, Blass JP, Peluso G. Tissuetransglutaminase-catalyzed formation of high-molecular-weight aggregates in vitro is favored with

Jeitner et al. Page 7

J Neurochem. Author manuscript; available in PMC 2009 September 28.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

long polyglutamine domains: a possible mechanism contributing to CAG-triplet diseases. ArchBiochem Biophys 1998;352:314–321. [PubMed: 9587422]

Gillet SM, Chica RA, Keillor JW, Pelletier JN. Expression and rapid purification of highly activehexahistidine-tagged guinea pig liver transglutaminase. Protein Expr Purif 2004;33:256–264.[PubMed: 14711514]

Goldstone SD, Fragonas JC, Jeitner TM, Hunt NH. Transcription factors as targets for oxidative signallingduring lymphocyte activation. Biochim Biophys Acta 1995;1263:114–122. [PubMed: 7640301]

Gomez-Santos C, Ferrer I, Santidrian AF, Barrachina M, Gil J, Ambrosio S. Dopamine inducesautophagic cell death and alpha-synuclein increase in human neuroblastoma SH-SY5Y cells. JNeurosci Res 2003;73:341–350. [PubMed: 12868068]

Green H. Human genetic diseases due to codon reiteration: relationship to an evolutionary mechanism.Cell 1993;74:955–956. [PubMed: 8104707]

Hadjivassiliou M, Aeschlimann P, Strigun A, Sanders DS, Woodroofe N, Acshlimann D. Autoantibodiesin gluten ataxia recognize a novel neural transglutaminase. Ann Neurol 2008;64:332–343. [PubMed:18825674]

Hartley DM, Zhao C, Speier AC, Woodard GA, Li S, Li Z, Walz T. Transglutaminase induces protofibril-like amyloid beta-protein assemblies that are protease-resistant and inhibit long-term potentiation. JBiol Chem 2008;283:16790–16800. [PubMed: 18397883]

Ho GJ, Gregory EJ, Smirnova IV, Zoubine MN, Festoff BW. Cross-linking of beta-amyloid proteinprecursor catalyzed by tissue transglutaminase. FEBS Lett 1994;349:151–154. [PubMed: 7913896]

Ikura K, Takahata K, Sasaki R. Cross-linking of a synthetic partial-length (1–28) peptide of the Alzheimerbeta/A4 amyloid protein by transglutaminase. FEBS Lett 1993;326:109–111. [PubMed: 8100780]

Jeitner TM, Bogdanov MB, Matson WR, et al. Nε-(γ-L-Glutamyl)-L-lysine (GGEL) is increased incerebrospinal fluid of patients with Huntington's disease. J Neurochem 2001;79:1109–1112.[PubMed: 11739625]

Jeitner TM, Delikatny EJ, Ahlqvist J, Capper H, Cooper AJL. Mechanism for the inhibition oftransglutaminase 2 by cystamine. Biochem Pharmacol 2005;69:961–970. [PubMed: 15748707]

Jeitner TM, Matson WR, Folk JE, Blass JP, Cooper AJL. Increased levels of gamma-glutamylamines inHuntington disease CSF. J Neurochem 2008;106:37–44. [PubMed: 18422943]

Jensen PH, Sorensen ES, Petersen TE, Gliemann J, Rasmussen LK. Residues in the synuclein consensusmotif of the alpha-synuclein fragment, NAC, participate in transglutaminase-catalysed cross-linkingto Alzheimer-disease amyloid β A4 peptide. Biochem J 1995;310:91–94. [PubMed: 7646476]

Johnson GVW, LeShoure R Jr. Immunoblot analysis reveals that isopeptide antibodies do not specificallyrecognize the ε-(γ-glutamyl)lysine bonds formed by transglutaminase activity. J Neurosci Methods2004;134:151–158. [PubMed: 15003381]

Johnson GVW, Cox TM, Lockhart JP, Zinnerman MD, Miller ML, Powers RE. Transglutaminase activityis increased in Alzheimer's disease brain. Brain Res 1997;751:323–329. [PubMed: 9099822]

Kahlem P, Terre C, Green H, Djian P. Peptides containing glutamine repeats as substrates fortransglutaminase-catalyzed cross-linking: relevance to diseases of the nervous system. Proc NatlAcad Sci USA 1996;93:14580–14585. [PubMed: 8962095]

Karpuj MV, Garren H, Slunt H, Price DL, Gusella J, Becher MW, Steinman L. Transglutaminaseaggregates huntingtin into nonamyloidogenic polymers, and its enzymatic activity increases inHuntington's disease brain nuclei. Proc Natl Acad Sci USA 1999;96:7388–7393. [PubMed:10377424]

Kawabe S, Ikuta T, Ohba M, Chida K, Ueda E, Yamanishi K, Kuroki T. Cholesterol sulfate activatestranscription of transglutaminase 1 gene in normal human keratinocytes. J Invest Dermatol1998;111:1098–1102. [PubMed: 9856823]

Kim SY, Grant P, Lee JH, Pant HC, Steinert PM. Differential expression of multiple transglutaminasesin human brain Increased expression and cross-linking by transglutaminases 1 and 2 in Alzheimer'sdisease. J Biol Chem 1999;274:30715–30721. [PubMed: 10521460]

Kim Y, Park YW, Lee YS, Jeoung D. Hyaluronic acid induces transglutaminase II to enhance cell motility;role of Rac1 and FAK in the induction of transglutaminase II. Biotechnol Lett 2008;30:31–39.[PubMed: 17680210]

Jeitner et al. Page 8

J Neurochem. Author manuscript; available in PMC 2009 September 28.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Konno T, Morii T, Shimizu H, Oiki S, Ikura K. Paradoxical inhibition of protein aggregation andprecipitation by transglutaminase-catalyzed intermolecular cross-linking. J Biol Chem2005;280:17520–17525. [PubMed: 15731111]

Krasnikov BF, Kim SY, McConoughey SJ, et al. Transglutaminase activity is present in highly purifiednonsynaptosomal mouse brain and liver mitochondria. Biochemistry 2005;44:7830–7843. [PubMed:15909997]

Kuemmerle S, Gutekunst CA, Klein AM, Li XJ, Li SH, Beal MF, Hersch SM, Ferrante RJ. Huntingtonaggregates may not predict neuronal death in Huntington's disease. Ann Neurol 1999;46:842–849.[PubMed: 10589536]

Kuncio GS, Tsyganskaya M, Zhu J, Liu SL, Nagy L, Thomazy V, Davies PJA, Zern MA. TNF-αmodulates expression of the tissue transglutaminase gene in liver cells. Am J Physiol1998;274:G240–G245. [PubMed: 9486175]

Lai TS, Tucker T, Burke JR, Strittmatter WJ, Greenberg CS. Effect of tissue transglutaminase on thesolubility of proteins containing expanded polyglutamine repeats. J Neurochem 2004;88:1253–1260.[PubMed: 15009681]

Lesort M, Attanavanich K, Zhang J, Johnson GVW. Distinct nuclear localization and activity of tissuetransglutaminase. J Biol Chem 1998;273:11991–11994. [PubMed: 9575137]

Lesort M, Chun W, Johnson GVW, Ferrante RJ. Tissue transglutaminase is increased in Huntington'sdisease brain. J Neurochem 1999;73:2018–2027. [PubMed: 10537061]

Lesort M, Lee M, Tucholski J, Johnson GVW. Cystamine inhibits caspase activity Implications for thetreatment of polyglutamine disorders. J Biol Chem 2003;278:3825–3830. [PubMed: 12458211]

Lin MT, Beal MF. Mitochondrial dysfunction and oxidative stress in neurodegenerative diseases. Nature2006;443:787–795. [PubMed: 17051205]

Lorand L, Graham RM. Transglutaminases: crosslinking enzymes with pleiotropic functions. Nat RevMol Cell Biol 2003;4:140–156. [PubMed: 12563291]

Mastroberardino PG, Iannicola C, Nardacci R, et al. ‘Tissue’ transglutaminase ablation reduces neuronaldeath and prolongs survival in a mouse model of Huntington's disease. Cell Death Differ 2002;9:873–880. [PubMed: 12181738]

Mattson MP. Calcium and neurodegeneration. Aging Cell 2007;6:337–350. [PubMed: 17328689]Medvedev A, Saunders NA, Matsuura H, Chistokhina A, Jetten AM. Regulation of the transglutaminase

I gene Identification of DNA elements involved in its transcriptional control in tracheobronchialepithelial cells. J Biol Chem 1999;274:3887–3896. [PubMed: 9920944]

Miller CC, Anderton BH. Transglutaminase and the neuronal cytoskeleton in Alzheimer's disease. JNeurochem 1986;46:1912–1922. [PubMed: 2871133]

Miller ML, Johnson GVW. Transglutaminase cross-linking of the tau protein. J Neurochem1995;65:1760–1770. [PubMed: 7561874]

Murthy SN, Wilson JH, Lukas TJ, Kuret J, Lorand L. Cross-linking sites of the human tau protein, probedby reactions with human transglutaminase. J Neurochem 1998;71:2607–2614. [PubMed: 9832162]

Nagy L, Saydak M, Shipley N, et al. Identification and characterization of a versatile retinoid responseelement (retinoic acid receptor response element-retinoid X receptor response element) in the mousetissue transglutaminase gene promoter. J Biol Chem 1996;271:4355–4365. [PubMed: 8626785]

Nemes Z, Devreese B, Steinert PM, Van Beeumen J, Fesus L. Cross-linking of ubiquitin, HSP27, parkin,and alpha-synuclein by γ-glutamyl-ε-lysine bonds in Alzheimer's neurofibrillary tangles. FASEB J2004;18:1135–1137. [PubMed: 15132984]

Pardin C, Pelletier JN, Lubell WD, Keillor JW. Cinnamoyl inhibitors of tissue transglutaminase. J OrgChem 2008a;73:5766–5775. [PubMed: 18582115]

Pardin C, Roy I, Lubell WD, Keillor JW. Reversible and Competitive Cinnamoyl Triazole Inhibitors ofTissue Transglutaminase. Chem Biol Drug Des 2008b;72:189–196. [PubMed: 18715232]

Park KC, Chung KC, Kim YS, Lee J, Joh TH, Kim SY. Transglutaminase 2 induces nitric oxide synthesisin BV-2 microglia. Biochem Biophys Res Commun 2004;323:1055–1062. [PubMed: 15381106]

Pastuszko A, Wilson DF, Ereciska M. A role for transglutaminase in neurotransmitter release by rat brainsynaptosomes. J Neurochem 1986;46:499–508. [PubMed: 2867126]

Jeitner et al. Page 9

J Neurochem. Author manuscript; available in PMC 2009 September 28.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Perry MJ, Mahoney SA, Haynes LW. Transglutaminase C in cerebellar granule neurons: regulation andlocalization of substrate cross-linking. Neuroscience 1995;65:1063–1076. [PubMed: 7617162]

Piacentini M, Martinet N, Beninati S, Folk JE. Free and protein-conjugated polyamines in mouseepidermal cells Effect of high calcium and retinoic acid. J Biol Chem 1988;263:3790–3794.[PubMed: 3346223]

Pinto JT, Van Raamsdonk JM, Leavitt BR, Hayden MR, Jeitner TM, Thaler HT, Krasnikov BF, CooperAJL. Treatment of YAC128 mice and their wild-type littermates with cystamine does not lead to itsaccumulation in plasma or brain: implications for the treatment of Huntington disease. J Neurochem2005;94:1087–1101. [PubMed: 15992377]

Polakowska RR, Graf BA, Falciano V, LaCelle P. Transcription regulatory elements of the first introncontrol human transglutaminase type I gene expression in epidermal keratinocytes. J Cell Biochem1999;73:355–369. [PubMed: 10321835]

Rasmussen LK, Sorensen ES, Petersen TE, Gliemann J, Jensen PH. Identification of glutamine and lysineresidues in Alzheimer amyloid beta A4 peptide responsible for transglutaminase-catalysedhomopolymerization and cross-linking to α 2M receptor. FEBS Lett 1994;338:161–166. [PubMed:7905838]

Ritter SJ, Davies PJA. Identification of a transforming growth factor-beta1/bone morphogenetic protein4 (TGF-beta1/ BMP4) response element within the mouse tissue transglutaminase gene promoter. JBiol Chem 1998;273:12798–12806. [PubMed: 9582307]

Sárvari M, Karpati L, Fésüs L, Deli L, Muszbek L, Nemes Z. Competitive enzyme-linked immunosorbentassay for N ε γ-glutamyl lysine. Anal Biochem 2002;311:187–190. [PubMed: 12470680]

Selkoe DJ, Abraham C, Ihara Y. Brain transglutaminase: in vitro crosslinking of human neurofilamentproteins into insoluble polymers. Proc Natl Acad Sci USA 1982a;79:6070–6074. [PubMed: 6136967]

Selkoe DJ, Ihara Y, Salazar FJ. Alzheimer's disease: insolubility of partially purified paired helicalfilaments in sodium dodecyl sulfate and urea. Science 1982b;215:1243–1245. [PubMed: 6120571]

Sieradzan KA, Mann DM. The selective vulnerability of nerve cells in Huntington's disease. NeuropatholAppl Neurobiol 2001;27:1–21. [PubMed: 11298997]

Sohn J, Kim TI, Yoon YH, Kim JY, Kim SY. Novel transglutaminase inhibitors reverse the inflammationof allergic conjunctivitis. J Clin Invest 2003;111:121–128. [PubMed: 12511595]

Stack EC, Ferro JL, Kim J, et al. Therapeutic attenuation of mitochondrial dysfunction and oxidativestress in neurotoxin models of Parkinson's disease. Biochim Biophys Acta 2008;1782:151–162.[PubMed: 18206128]

Tucholski J, Kuret J, Johnson GVW. Tau is modified by tissue transglutaminase in situ: possiblefunctional and metabolic effects of polyamination. J Neurochem 1999;73:1871–1880. [PubMed:10537045]

Van Raamsdonk JM, Pearson J, Bailey CD, Rogers DA, Johnson GVW, Hayden MR, Leavitt BR.Cystamine treatment is neuroprotective in the YAC128 mouse model of Huntington disease. JNeurochem 2005;95:210–220. [PubMed: 16181425]

Vermes I, Steur EN, Jirikowski GF, Haanen C. Elevated concentration of cerebrospinal fluid tissuetransglutaminase in Parkinson's disease indicating apoptosis. Mov Disord 2004;19:1252–1254.[PubMed: 15368613]

Wang J, Wang CE, Orr A, Tydlacka S, Li SH, Li XJ. Impaired ubiquitin-proteasome system activity inthe synapses of Huntington's disease mice. J Cell Biol 2008;180:1177–1189. [PubMed: 18362179]

Yan ZH, Noonan S, Nagy L, Davies PJA, Stein JP. Retinoic acid induction of the tissue transglutaminasepromoter is mediated by a novel response element. Mol Cell Endocrinol 1996;120:203–212.[PubMed: 8832580]

Zainelli GM, Ross CA, Troncoso JC, Muma NA. Transglutaminase cross-links in intranuclear inclusionsin Huntington disease. J Neuropathol Exp Neurol 2003;62:14–24. [PubMed: 12528814]

Zainelli GM, Dudek NL, Ross CA, Kim SY, Muma NA. Mutant huntingtin protein: a substrate fortransglutaminase 1, 2, and 3. J Neuropathol Exp Neurol 2005;64:58–65. [PubMed: 15715085]

Zemaitaitis MO, Lee JM, Troncoso JC, Muma NA. Transglutaminase-induced cross-linking of tauproteins in progressive supranuclear palsy. J Neuropathol Exp Neurol 2000;59:983–989. [PubMed:11089576]

Jeitner et al. Page 10

J Neurochem. Author manuscript; available in PMC 2009 September 28.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Zemaitaitis MO, Kim SY, Halverson RA, Troncoso JC, Lee JM, Muma NA. Transglutaminase activity,protein, and mRNA expression are increased in progressive supranuclear palsy. J Neuropathol ExpNeurol 2003;62:173–184. [PubMed: 12578227]

Zhang W, Johnson BR, Suri DE, Martinez J, Bjornsson TD. Immunohistochemical demonstration oftissue transglutaminase in amyloid plaques. Acta Neuropathol 1998;96:395–400. [PubMed:9797004]

Jeitner et al. Page 11

J Neurochem. Author manuscript; available in PMC 2009 September 28.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Fig. 1.Effect of cysteamine, cysteine, hypotaurine, and taurine on TG activity. TG activity in thepresence of cysteamine (□), cysteine (■) hypotaurine (○), or taurine (●) was determined bymeasuring the incorporation of radiolabeled putrescine into N,N-dimethylcasein as describedby Jeitner et al. (2005) using tritiated putrescine and his-tagged guinea pig TG 2 (Gillet etal. 2004). The data are depicted as percent of the control (21 910 ± 1731 dpm per tube) andrepresent the mean ± SEM of four separate experiments. The data with cysteamine atconcentrations ≥ 2.5 mM and taurine at 5 and 10 mM were significantly different from that ofthe control (p < 0.05, paired t-test).

Jeitner et al. Page 12

J Neurochem. Author manuscript; available in PMC 2009 September 28.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Fig. 2.Effect of cyst(e)amine on dopamine-induced macroautophagy. SH SY5Y cells at 70%confluence in a 75 cm2 flask were detached with 0.05% trypsin : verscene (1 : 1), then collectedinto 50 mL 10% fetal bovine serum, 90% Dulbecco's modified eagle medium prior to seedingonto 24 multi-well plates at 1 mL per well. Twenty-four hours later the cells were treated witheither cystamine (■) or cysteamine (□) for 2 h. The cells were then incubated for an additional24 h together with 10−4 M dopamine to induce macroautophagy as described by Gomez-Santoset al. (2003). The data from these studies are depicted as the mean ± SEM of four separateexperiments. The cells were also treated with cystamine for 4 h then washed three times withHank's balanced salt solution (HBBS) at 37°C, followed by 24 h incubation with 100 μMdopamine (gray open circles). The data from these studies represent the mean of two individualexperiments that did not vary by more than 5% of the mean. At the end of the incubations, thecells were washed twice with HBSS at 37°C then incubated with 250 μL of 1 mg/mL 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-tetrazolium bromide in HBSS for 30 min at 37°C, for thedetermination of viability. The resulting formazan precipitates were then solubilized with 200μL DMSO. The viability of cells treated with cystamine at 15 μM and 100 μM dopamine (■)was significantly different than that of cells treated with dopamine alone (p < 0.05, paired t-test.

Jeitner et al. Page 13

J Neurochem. Author manuscript; available in PMC 2009 September 28.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript