A Small Molecule Mimetic of the Humanin Peptide as a Candidate for

Nov 21, 2017 - Department of Orthopedic Surgery, DGSOM, UCLA, Los Angeles, California 90095, United States. § Department of Orthopedic Surgery, Unive...
0 downloads 6 Views 1MB Size
Subscriber access provided by READING UNIV

Letter

A Small Molecule Mimetic of the Humanin Peptide as a Candidate for Modulating NMDA-Induced Neurotoxicity Mohammad Parvez Alam, Tina Bilousova, Patricia Spilman, Kanagasabai Vadivel, Dongsheng Bai, Denis Evseenko, and Varghese John ACS Chem. Neurosci., Just Accepted Manuscript • DOI: 10.1021/acschemneuro.7b00350 • Publication Date (Web): 21 Nov 2017 Downloaded from http://pubs.acs.org on November 23, 2017

Just Accepted “Just Accepted” manuscripts have been peer-reviewed and accepted for publication. They are posted online prior to technical editing, formatting for publication and author proofing. The American Chemical Society provides “Just Accepted” as a free service to the research community to expedite the dissemination of scientific material as soon as possible after acceptance. “Just Accepted” manuscripts appear in full in PDF format accompanied by an HTML abstract. “Just Accepted” manuscripts have been fully peer reviewed, but should not be considered the official version of record. They are accessible to all readers and citable by the Digital Object Identifier (DOI®). “Just Accepted” is an optional service offered to authors. Therefore, the “Just Accepted” Web site may not include all articles that will be published in the journal. After a manuscript is technically edited and formatted, it will be removed from the “Just Accepted” Web site and published as an ASAP article. Note that technical editing may introduce minor changes to the manuscript text and/or graphics which could affect content, and all legal disclaimers and ethical guidelines that apply to the journal pertain. ACS cannot be held responsible for errors or consequences arising from the use of information contained in these “Just Accepted” manuscripts.

ACS Chemical Neuroscience is published by the American Chemical Society. 1155 Sixteenth Street N.W., Washington, DC 20036 Published by American Chemical Society. Copyright © American Chemical Society. However, no copyright claim is made to original U.S. Government works, or works produced by employees of any Commonwealth realm Crown government in the course of their duties.

Page 1 of 24 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Chemical Neuroscience

A Small Molecule Mimetic of the Humanin Peptide as a Candidate for Modulating NMDA-Induced Neurotoxicity Mohammad Parvez Alam†, Tina Bilousova†, Patricia Spilman†, Kanagasabai Vadivel‡, Dongsheng Bai†, Chris J. Elias†, Denis Evseenko§, and Varghese John†* Affiliations: †

Drug Discovery Laboratory, Department of Neurology, UCLA, Los Angeles, CA, 90095, USA Department of Orthopedic Surgery, DGSOM, UCLA, Los Angeles, CA, 90095, USA § Department of Orthopedic Surgery, University of Southern California (USC), Los Angeles, CA, 90033, USA ‡

ACS Paragon Plus Environment

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 2 of 24

ABSTRACT: Humanin (HN), a 24-amino acid bioactive peptide, has been shown to increase cell survival of neurons after exposure to Aβ and NMDA-induced toxicity and thus could be beneficial in the treatment of Alzheimer’s disease (AD). The neuroprotection by HN is reported to be primarily through its agonist binding properties to the gp130 receptor. However, peptidic nature of HN presents challenges in its development as a therapeutic for AD. We report here for the first time the elucidation of the binding site of Humanin (HN) peptide to gp130 receptor extracellular domain through modeling and the synthesis of small molecule mimetics that interact with the HN binding site on the gp130 receptor and provide protection against NMDA-induced neurotoxicity in primary hippocampal neurons. A brain permeable small molecule mimetic was identified through exploratory medicinal chemistry using microfluidic flow chemistry to facilitate the synthesis of new analogs for screening and SAR optimization.

KEYWORDS: Humanin, gp130 receptor, NMDA, excitotoxicity, primary neurons, microfluidics

2

ACS Paragon Plus Environment

Page 3 of 24 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Chemical Neuroscience

Alzheimer’s disease (AD) is the most prevalent age-related dementia and currently approved therapeutics provide only temporary symptomatic relief. Therefore new approaches to therapeutic development are urgently needed. Neuronal cell death is a key feature of AD pathology and factors leading to neuronal loss are many and not completely elucidated.1

In 2001, Hashimoto et al. identified Humanin (HN) a 24-amino acid peptide from occipital lobe of postmortem AD patient brain tissue and showed that it protected neurons from amyloid-beta (Aβ)related toxicity.2 Muzumdar and coworkers reported a decrease in the endogenous HN plasma level with age, suggesting that decreased HN levels may be linked to cognitive decline during aging.3 The potent HN analog HNG, with a substitution of serine-14 to glycine, showed neuroprotection against Aβ1-42induced toxicity in primary neurons at nanomolar levels.2 HN exerts its function through binding to both extracellular receptors and intracellular binding partners.4 The proposed extracellular binding partners include G-protein coupled receptors, formyl peptide receptor like (FPRL)-1 and 2;5,

6

and a trimeric

receptor complex of gp130, ciliary neurotrophic factor receptor α (CNTFR), and the IL-27 receptor subunit, WSX-1.7

Hashimoto and coworkers reported that HN confers its protection against Aβ1-42 neurotoxicity through agonism of gp130 and that is required for HN-mediated protection against Aβ1-42-induced death. In F11 neurohybrid cells transfected with amyloid precursor protein (APP) with the familial AD mutant V642IAPP, there is significant neuronal cell death in vitro. Treatment with HN protects against this cell death, but this protection is eradicated if the F11 cells are also treated with a gp130 neutralizing antibody. In addition, the study shows that activation of STAT3 is essential for the HN activity, thus treatment of the F11 cells with HN resulted in up-regulation of the tyrosine phosphorylation of STAT3 (pTyr7053

ACS Paragon Plus Environment

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 4 of 24

STAT3).7 Immunofluorescence (IF) labeling in aged Tg2576 AD model mice showed that pSTAT3 levels were significantly reduced in the dentate gyrus (DG) and CA1 brain regions of vehicle-treated mice while treatment intranasally with the HN derivative peptide Colivelin (CLN) restored pSTAT3 levels completely up to levels seen in non-transgenic mice.8 Furthermore, HNG has been shown to act through the gp130/IL6ST receptor complex to activate STAT3, AKT and ERK1/2 signaling pathways.9 These results support the reports, which suggest that an increase in gp130 signaling may be of benefit in AD.10 Others have suggested that the anti-AD therapeutic benefits of HN could be through interaction with residues (17–28) of monomeric Aβ that results in prevention of Aβ oligomerization and provides neuroprotection.11, 12

Recent studies have shown that HN can rescue neurons from NMDA-mediated toxicity in a manner similar to the NMDA receptor antagonist MK801, but the mechanism of the rescue by HN is not through antagonism of the NMDA receptor.13 Excitotoxicity through NMDA receptor over-activation is associated with disease progression in AD,14 therefore over the years there has been significant drug discovery efforts to identify new NMDA receptor antagonists for treatment of AD and other neurological disorders such as Parkinson’s disease, cerebral ischemia, and stroke wherein excitotoxicity is thought to play a role. However, such antagonists have been associated with significant adverse effects and neurotoxicity. A partial antagonist of the NMDA receptor called memantine (Namenda®) was approved for AD as a symptomatic therapy over a decade ago but provides only limited benefit.15

We show here - for the first time - that HN acts to rescue the NMDA-mediated toxicity in primary hippocampal neurons through its interaction with the domains-4 and -5 (D4-D5) of the gp130 receptor. Our discovery was based on computational modeling of HN peptide and the extracellular domains of 4

ACS Paragon Plus Environment

Page 5 of 24 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Chemical Neuroscience

gp130. Interestingly, through screening and exploratory medicinal chemistry approaches we identified a small molecule series that also binds to gp130 receptor in the same binding site between domains D4-D5 as HN. The small molecule mimetics, like HN, rescue neurons from NMDA-mediated toxicity. Furthermore, our modeling analysis shows that a known antagonist, SC144,16 of gp130 also binds to the same HN binding site and reverses the neuroprotection conferred by these mimetics providing experimental evidence of their direct interaction with gp130 pathway. Thus, these small molecules are HN mimetics revealing a new mechanism for suppressing NMDA receptor over-activation and excitotoxicity for new therapeutic development in neurodegenerative disorders such as AD.

RESULTS AND DISCUSSION

Our discovery efforts for small molecule HN mimetic began with a screening hit (RCGD)-423 (compound 1), which was found to be a regulator of cartilage growth and differentiation. This compound was identified by our collaborator at UCLA through an HTS effort using the 200,000 UCLA screening library, to find compounds that increase articular chondrocyte activation and facilitate joint repair. Compound 1 was shown to bind gp130 initiating a signaling cascade comprising activation of the JAKSTAT3 pathway, phosphorylation of STAT3 (pSTAT3), and ultimately regulation of gene expression in chondrocytes (Shkhyan et al. submitted manuscript) and hair follicle stem cells.17 Given that HN has been shown to elicit its neuroprotective effects through gp130 receptor agonism,7 we wanted to use compound 1 as a starting lead to conduct exploratory medicinal chemistry to find gp130 interacting HN mimetics. We rationally designed and synthesized compounds 2–8 and evaluated their protective effects against NMDA-induced neurotoxicity.

5

ACS Paragon Plus Environment

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 6 of 24

The ClusPro docking server was used to model the interactions between HN and the extracellular domains of gp130;18 this also revealed the HN binding domains D4-D5 as a potential high-affinity binding sites for the HN peptide in the gp130 extracellular region (Figure 1). In addition, for compounds 1–8 we performed modeling studies using the Swissdock program; these again revealed domains D4-D5 as a potential high-affinity binding site in the gp130 extracellular region (Figure 1, Supporting information, Figures S1–S7). The binding free energy of HN and compound 2 to gp130 D4D6 were determined to be -46.99 (± 9.65) and -18.61 (± 2.30) kcal/mol, respectively using the MM/PBSA method implemented in AMBER (see Supporting Information). The compound 2 remained bound to the gp130 D4D6 domain during the 50 ns molecular dynamic simulations similar to that seen with the larger 24 amino acid peptide HN and which has more interactions to the receptor due to its relatively larger size than 2. For the small molecule mimetic 2 this implies good affinity of compound 2 for gp130 receptor at the D4D6 domain, the calculated binding energy for compound 2 is ~2.5 fold lower than that of HN. The known gp130 antagonist, SC144, was also modeled in parallel to investigate its binding specificity (Supporting information, Figure S8). SC144 was found to have high-affinity binding for the same site in the D4-D5 domains. Thus, modeling reveals that the compounds 1–8, the gp130 antagonist SC144 and HN all bound to domains (D4-D5) of gp130 receptor.

Compound 1 and analogs belong to the 2-aminothiazoles family of compounds, and while many methods have been reported to synthesize 2-aminothiazoles, including solution, solid phase synthesis, a tandem one-pot aqueous phase synthesis19 and microwave promoted synthesis.20 Our method for 2aminothiazoles preparation involves Hantzsch condensation21 using 2-bromo-1-phenylethan-1-one (and its derivatives) and 1-(4-bromophenyl)thiourea (and its derivatives) in a microfluidic reactor without the use of a catalyst which is unprecedented (Table 1). 6

ACS Paragon Plus Environment

Page 7 of 24 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Chemical Neuroscience

We use a flow-chemistry approach that we have previously described,22 for our synthesis. Microfluidic flow chemistry facilitated rapid syntheses of the new analogs. This is a green chemistry approach with a cleaner reaction profile, smaller carbon footprint, higher yields, and quick scalability with minimum reaction optimization enabling us to quickly gain SAR insights into the optimal gp130 binding molecules for neuroprotection. The reactions are rapid, proceeding to completion in a few minutes and products are obtained in good yields, in milligram quantities. As a result of these efforts, we report the synthesis of new chemical entities (NCEs) such as N-(2,4-difluorophenyl)-4-(pyridin-3-yl)thiazol-2amine (compound 6). With a number of analogs in hand, we next determined the ability of the small molecules to confer neuroprotection against NMDA-induced toxicity (Table 1). In this study, the cytotoxicity of NMDA was quantified by measuring the activity of lactate dehydrogenase (LDH) released from primary cultured hippocampal neurons into the media and confirmed by living cell density analysis using calcein staining. Our results showed that NMDA treatment (100 µM) for 2.5 h caused cytotoxicity in the primary neurons in vitro and triggered release of LDH (Table 1 and Figure 2A). LDH level in the NMDA treatment group was about 26% higher than in the DMSO control. MK801, a noncompetitive antagonist of NMDA receptor, was used as a positive control in these studies.23 As shown in Table 1 and Figure 2A, the addition of MK801 significantly reduced, by 43 %, the release of LDH from NMDA-treated neurons. We evaluated different batches of HN and HNG from different vendors in our assay and only one of them (Tocris Biosciences, cat# 5154, SI) was active and exhibited protection against NMDA-induced neurotoxicity (Supplementary Figure S9). Neither HN nor the compounds by itself had any effect on LDH levels from neurons which were not treated with NMDA. However, the release of LDH was significantly inhibited in NMDA-treated neurons when treated with the HN mimetics. Neurons were pretreated with compounds for 16 h before exposure to NMDA. 7

ACS Paragon Plus Environment

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 8 of 24

The 2-aminothiazole derivatives showed varied neuroprotective activity depending on the aryl substitution on either side of the central aminothiazole core. As shown in Table 1 the levels of NMDA induced LDH release was inhibited by 20% on treatment with our initial screening hit 1 while greater inhibition of the LDH release was seen with the HN mimetic analogs 2–8 at 10 µM. The levels of NMDA induced LDH release was inhibited by 21% on treatment with compound 3, the pyridine analog of compound 2. While compound 4, decrease the LDH levels by 32% compound 5 with two fluoro groups at the ortho and para - position inhibited LDH release by 43%, similar to the activity of MK801. Compound 6 is the pyridine analog of compound 5, and it inhibited NMDA induced LDH release similar to what observed for compound 3. Compound 7 with the phenethyl group showed similar inhibition as compound 3 and compound 6. Introduction of fluoro group at para position in both the rings, compound 8, showed slightly worse inhibition of LDH release as compared to when the fluoro group was just present only in one ring. Compound 2 was one of the best and its neuroprotective activity was similar to MK801. The known gp130 antagonist, SC144, treatment reversed the neuroprotection provided by compounds 2 or 8 against NMDA-induced neurotoxicity (Figure 2A). Compound 2 was assessed for its downstream signaling based on its effects on STAT3, AKT and ERK activation as previously reported upon HNG treatment in HEK293 cells, SH-SY5Y cells and primary hippocampal neurons.9 For our experiments we used the potent HN analog HNG1724 in SH-SY5Y cells and primary hippocampal neurons. Treatment of the cells with HNG17 and compound 2 (10µM) in serum free media were performed for varying time periods as indicated in supplementary Figure S11. In SH-SY5Y cells, compound 2 treatment showed a 2-fold increase (as measured using the ratio of densitometry values for the p-STA3 and STAT3 bands) in phosphorylation of STAT3 within 10 minutes at its regulatory Tyr705 site (Supplementary Figure S11A), this is similar to that seen with HNG17 at 5 minutes. However, in primary hippocampal neuronal cultures the p-STAT3 levels were below levels of 8

ACS Paragon Plus Environment

Page 9 of 24 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Chemical Neuroscience

detection both for HNG17 and compound 2 at all time points (Supplementary Figure S11B), this could be due lower cross reactivity of the p-STAT3 antibody for the mouse protein. Compound 2 treatment showed 9-fold increase in phosphorylation of AKT at its regulatory Thr308 site in the serum free media condition in SH-SY5Y cells (Supplementary Figure S11A) after 15 minutes incubation and 4-fold increase in primary cortical neurons for the same time (Supplementary Figure S11B). These increases in p-AKT were greater than that seen with HNG17 in these cells. Compound 2 treatment caused a rapid 2fold increase in phosphorylation of ERK1/2 at its regulatory Thr202/Tyr204 site in the serum free media condition in SH-SY5Y cell (Supplementary Figure S11A) and 1.2 fold in primary cortical neurons (Supplementary Figure S11B), similar increase were seen with HNG17 in the SH-SY5Y cells and less so in the primary neuronal cells. In SH-SY5Y cells, co-treatment of compound 2 with gp130 antagonist, SC144, inhibited phosphorylation of STAT3, AKT and ERK1/2, this effect is similar to what was reported on treatment with the HN analog HNG treatment and with the anti-gp130 antibody or the gp130 antagonist SC144.9 This data supports the downstream signaling effects of compound 2 primarily through its interaction with the gp130 receptor. Furthermore, co-treatment of compound 2 with AG490, a JAK inhibitor, greatly reduced the AKT phosphorylation indicating compound 2 acts upstream to JAK. In contrast, ERK1/2 phosphorylation did not change much, as there are alternate pathways involved besides through the trimeric gp130 complex involving ciliary neurotrophic factor receptor α (CNTFR), and the IL-27 receptor subunit, WSX-1, for activation of the ERK pathway.

9

These data support

compound 2 being a HN mimetic at the gp130 receptor.

Calcein (calcein-acetoxymethyl ester; calcein-AM), a highly lipophilic non-fluorescent and cell permeable compound, staining was used to evaluate the density of living primary neurons. Calcein-AM can be converted by intracellular esterases into calcein, an anionic fluorescent form, only labeling viable 9

ACS Paragon Plus Environment

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 10 of 24

cells, and thus provides both morphological and functional information about living neuronal cells. NMDA (100 µM, 2.5 h) alone induced a significant decrease in neuronal survival ─ more than 50% cell death observed. With MK801 pretreatment, the density of NMDA-treated neurons was rescued to levels better than DMSO-only control, this indicates that the toxicity was induced by NMDA receptor activation (Figure 2B-2D, Supplementary Figure S10). Furthermore, there was no difference in density of living neurons between the compound 2 group and the DMSO-only control group, suggesting that compound 2 could also reduce NMDA-induced toxicity similar to the direct antagonist, MK801.

The neuroprotective effects of the compound 2 make it a promising candidate for further testing in AD models. Since any potential therapeutic for AD needs to cross the blood-brain barrier (BBB), we further evaluated the brain permeability of compound 2, in mice. Mice were dosed orally at 10 or 30 mg/kg, or injected subcutaneously (SQ) at 10 mg/kg, and euthanized after 1, 2, 4, 6, and 8 hours post-dose. We found that at 2 h after SQ delivery at 10 mg/kg the brain Cmax was 161 ng/g while dosing at 30 mg/kg orally, however, resulted in the brain Cmax of 156 ng/g (0.57 µM) as shown in Figure 3. The brain to plasma ratio for 2 was ~4:1 for oral 30 mg/kg; and ~7.5:1 for 10 mg/kg SQ injection. Based on the good brain permeability of compound 2 it is a promising lead candidate to conduct proof-of-concept testing in a mouse model of NMDA-induced toxicity in future studies.

In the present study, we elucidate using modeling, the binding site of HN in the extracellular domains of the receptor gp130. Our results show that HN and small molecule mimetics binds to the same site at the interface of the D4-D5 domain and provide neuroprotection from the NMDA neurotoxicity in primary hippocampal neurons. The small molecule HN mimetics hold great promise towards developing an orally available bioactive small molecule that confers the benefits of the humanin peptide. HN has 10

ACS Paragon Plus Environment

Page 11 of 24 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Chemical Neuroscience

shown to have multiple beneficial effects in neurodegeneration, diabetes, and artherosclerosis, but due to its peptidic structure, presents challenges in its development as a therapeutic.25-27 Small molecule mimetics of HN are attractive agents for therapeutic development both in AD and other neurological disorders where HN has shown beneficial effects such as in traumatic brain injury (TBI),28 stroke,29 Aβinduced cerebrovascular dementia,30 and amyotrophic lateral sclerosis (ALS).4 Humanin has a number of different cytoprotective and metaboloprotective effects such as cardioprotection against cellular apoptosis during myocardial injury,31 along with that HN modulate systemic insulin sensitivity and glucose homeostasis.32 Given the good oral brain permeability of the HN mimetic compound 2, it represents a promising molecular tool for in vivo proof-of-concept testing as a neuroprotective agent in NMDA and Aβ-induced toxicity models. Further efforts will be made to explore SAR and expand our library of small molecule HN mimetics, while evaluating the underlying gp130 signaling mechanism. These agents could lead to a new pharmacological class of therapeutic agents for AD.

METHODS General Procedure for Synthesis. The chemical synthesis of compound 2 began from commercially available 2-bromo-1-phenylethan-1-one and 1-(4-fluorophenyl)thiourea. They were premixed in a separate vial and were pumped through a preheated glass microfluidic reactor maintained at 80°C and 2 bar pressure (Syrris Asia Flow Chemistry Module) at a 500 µL/min flow rate with a one minute residence time in the reactor using Syrris Asia pumps to afford compound 2 from the output stream. Other compounds were synthesized (Table 1) by similarly pumping corresponding starting material through the preheated glass microfluidic reactor. The general procedure for cell testing and pharmacokinetic analysis is provided in Supporting Information.

11

ACS Paragon Plus Environment

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 12 of 24

Statistical Analysis. All the data were expressed as the mean ± SEM. Significant differences were determined by Student’s t-tests, and one-way ANOVA followed by Dunnett's multiple comparisons test using GraphPad Prism 7 software. Values of * < 0.05, ** < 0.01, *** < 0.001 were considered statistically significant.

ASSOCIATED CONTENT Supporting Information The supporting information is available free of charge on the ACS Publication website at DOI: Supplementary data including biological methods, experimental procedures, western blot, additional in silico modeling figures, 1H NMR and 13C NMR of the synthesized compounds are provided (PDF).

AUTHOR INFORMATION Corresponding Author *Varghese John, PhD. Associate Professor, Drug Discovery Laboratory, Department of Neurology, UCLA,

3131

Reed,

710

Westwood

Plaza,

Los

Angeles,

CA,

90095,

USA.

E-mail:

[email protected]. Author Contributions M.P.A., V.J., and P.S. has wrote the manuscript. V.J. contributed to the design and interpretation of the experiments. M.P.A. synthesized the compounds, designed the experiments, helped T.B. with the biochemical experiments, and analyzed the data. T.B. has designed and performed primary neuronal culture, cell culture, biochemical experiments, and analyzed the data. K.V. has performed docking and molecular dynamics studies. D.B. has performed PK study. C.J.E. has contributed by performing

12

ACS Paragon Plus Environment

Page 13 of 24 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Chemical Neuroscience

western blot. D.E. has provided the structure of initial hit. All authors have given approval to the final version of the manuscript.

Funding Sources This research was supported by the funding provided to V.J. by the Mary S. Easton Center for Alzheimer’s disease Research

Notes The authors declare no competing financial interests.

ACKNOWLEDGEMENTS Mass Spectrometry Instrumentation was made available through the support of Dr. Greg Khitrov at the University of California, Los Angeles Molecular Instrumentation Center – Mass Spectrometry Facility in the Department of Chemistry.

13

ACS Paragon Plus Environment

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 14 of 24

REFERENCES [1] Pepeu, G., and Grazia Giovannini, M. (2017) The fate of the brain cholinergic neurons in neurodegenerative diseases, Brain Res 1670, 173-184. [2] Hashimoto, Y., Niikura, T., Tajima, H., Yasukawa, T., Sudo, H., Ito, Y., Kita, Y., Kawasumi, M., Kouyama, K., Doyu, M., Sobue, G., Koide, T., Tsuji, S., Lang, J., Kurokawa, K., and Nishimoto, I. (2001) A rescue factor abolishing neuronal cell death by a wide spectrum of familial Alzheimer’s disease genes and Ab, Proc. Natl. Acad. Sci.U. S. A. 98, 6336–6341. [3] Muzumdar, R. H., Huffman, D. M., Atzmon, G., Buettner, C., Cobb, L. J., Fishman, S., Budagov, T., Cui, L., Einstein, F. H., Poduval, A., Hwang, D., Barzilai, N., and Cohen, P. (2009) Humanin: a novel central regulator of peripheral insulin action, PLoS One 4, e6334. [4] Gong, Z., Tas, E., and Muzumdar, R. (2014) Humanin and age-related diseases: a new link?, Front Endocrinol (Lausanne) 5, 210. [5] Harada, M., Habata, Y., Hosoya, M., Nishi, K., Fujii, R., Kobayashi, M., and Hinuma, S. (2004) NFormylated humanin activates both formyl peptide receptor-like 1 and 2, Biochem Biophys Res Commun 324, 255-261. [6] Ying, G., Iribarren, P., Zhou, Y., Gong, W., Zhang, N., Yu, Z. X., Le, Y., Cui, Y., and Wang, J. M. (2004) Humanin, a newly identified neuroprotective factor, uses the G protein-coupled formylpeptide receptor-like-1 as a functional receptor, J Immunol 172, 7078-7085. [7] Hashimoto, Y., Kurita, M., Aiso, S., Nishimoto, I., and Matsuoka, M. (2009) Humanin inhibits neuronal cell death by interacting with a cytokine receptor complex or complexes involving CNTF receptor alpha/WSX-1/gp130, Mol Biol Cell 20, 2864-2873. [8] Chiba, T., Yamada, M., Sasabe, J., Terashita, K., Shimoda, M., Matsuoka, M., and Aiso, S. (2009) Amyloid-beta causes memory impairment by disturbing the JAK2/STAT3 axis in hippocampal neurons, Mol Psychiatry 14, 206-222. [9] Kim, S. J., Guerrero, N., Wassef, G., Xiao, J., Mehta, H. H., Cohen, P., and Yen, K. (2016) The mitochondrial-derived peptide humanin activates the ERK1/2, AKT, and STAT3 signaling pathways and has age-dependent signaling differences in the hippocampus, Oncotarget 7, 4689946912. [10] Matsuoka, M., and Hashimoto, Y. (2010) Humanin and the receptors for humanin, Mol Neurobiol 41, 22-28. [11] Maftei, M., Tian, X., Manea, M., Exner, T. E., Schwanzar, D., von Arnim, C. A., and Przybylski, M. (2012) Interaction structure of the complex between neuroprotective factor humanin and Alzheimer's beta-amyloid peptide revealed by affinity mass spectrometry and molecular modeling, J Pept Sci 18, 373-382. [12] Romeo, M., Stravalaci, M., Beeg, M., Rossi, A., Fiordaliso, F., Corbelli, A., Salmona, M., Gobbi, M., Cagnotto, A., and Diomede, L. (2017) Humanin Specifically Interacts with Amyloid-beta Oligomers and Counteracts Their in vivo Toxicity, J Alzheimers Dis 57, 857-871. [13] Cui, A. L., Li, J. Z., Feng, Z. B., Ma, G. L., Gong, L., Li, C. L., Zhang, C., and Li, K. (2014) Humanin rescues cultured rat cortical neurons from NMDA-induced toxicity not by NMDA receptor, ScientificWorldJournal 2014, 341529. [14] Sonkusare, S. K., Kaul, C. L., and Ramarao, P. (2005) Dementia of Alzheimer's disease and other neurodegenerative disorders--memantine, a new hope, Pharmacol Res 51, 1-17. [15] Matsunaga, S., Kishi, T., and Iwata, N. (2015) Memantine monotherapy for Alzheimer's disease: a systematic review and meta-analysis, PLoS One 10, e0123289. 14

ACS Paragon Plus Environment

Page 15 of 24 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Chemical Neuroscience

[16] Xu, S., Grande, F., Garofalo, A., and Neamati, N. (2013) Discovery of a novel orally active smallmolecule gp130 inhibitor for the treatment of ovarian cancer, Mol Cancer Ther 12, 937-949. [17] Flores, A., Schell, J., Krall, A. S., Jelinek, D., Miranda, M., Grigorian, M., Braas, D., White, A. C., Zhou, J. L., Graham, N. A., Graeber, T., Seth, P., Evseenko, D., Coller, H. A., Rutter, J., Christofk, H. R., and Lowry, W. E. (2017) Lactate dehydrogenase activity drives hair follicle stem cell activation, Nat Cell Biol. [18] Comeau, S. R., Gatchell, D. W., Vajda, S., and Camacho, C. J. (2004) ClusPro: a fully automated algorithm for protein-protein docking, Nucleic Acids Res 32, W96-99. [19] Madhav, B., Narayana Murthy, S., Anil Kumar, B. S. P., Ramesh, K., and Nageswar, Y. V. D. (2012) A tandem one-pot aqueous phase synthesis of thiazoles/selenazoles, Tetrahedron Letters 53, 3835-3838. [20] Kabalka, G. W., and Mereddy, A. R. (2006) Microwave promoted synthesis of functionalized 2aminothiazoles, Tetrahedron Letters 47, 5171-5172. [21] Hantzsch, A., and Weber, J. H. (1887) Ber Dtsch Chem Ges 20, 3118. [22] Alam, M. P., Jagodzinska, B., Campagna, J., Spilman, P., and John, V. (2016) C-O bond Formation in a Microfluidic Reactor: High Yield SNAr Substitution of Heteroaryl Chlorides, Tetrahedron Lett 57, 2059-2062. [23] Wong, E. H., Kemp, J. A., Priestley, T., Knight, A. R., Woodruff, G. N., and Iversen, L. L. (1986) The anticonvulsant MK-801 is a potent N-methyl-D-aspartate antagonist, Proc Natl Acad Sci U. S. A. 83, 7104-7108. [24] Chiba, T., Yamada, M., Hashimoto, Y., Sato, M., Sasabe, J., Kita, Y., Terashita, K., Aiso, S., Nishimoto, I., and Matsuoka, M. (2005) Development of a femtomolar-acting humanin derivative named colivelin by attaching activity-dependent neurotrophic factor to its N terminus: characterization of colivelin-mediated neuroprotection against Alzheimer's disease-relevant insults in vitro and in vivo, J Neurosci 25, 10252-10261. [25] Niikura, T., Sidahmed, E., Hirata-Fukae, C., Aisen, P. S., and Matsuoka, Y. (2011) A humanin derivative reduces amyloid beta accumulation and ameliorates memory deficit in triple transgenic mice, PLoS One 6, e16259. [26] Zhang, W., Zhang, W., Li, Z., Hao, J., Zhang, Z., Liu, L., Mao, N., Miao, J., and Zhang, L. (2012) S14G-humanin improves cognitive deficits and reduces amyloid pathology in the middle-aged APPswe/PS1dE9 mice, Pharmacol Biochem Behav 100, 361-369. [27] Chin, Y. P., Keni, J., Wan, J., Mehta, H., Anene, F., Jia, Y., Lue, Y. H., Swerdloff, R., Cobb, L. J., Wang, C., and Cohen, P. (2013) Pharmacokinetics and tissue distribution of humanin and its analogues in male rodents, Endocrinology 154, 3739-3744. [28] Wang, T., Huang, Y., Zhang, M., Wang, L., Wang, Y., Zhang, L., Dong, W., Chang, P., Wang, Z., Chen, X., and Tao, L. (2013) [Gly14]-Humanin offers neuroprotection through glycogen synthase kinase-3beta inhibition in a mouse model of intracerebral hemorrhage, Behav Brain Res 247, 132-139. [29] Xu, X., Chua, C. C., Gao, J., Hamdy, R. C., and Chua, B. H. (2006) Humanin is a novel neuroprotective agent against stroke, Stroke 37, 2613-2619. [30] Jung, S. S., and Van Nostrand, W. E. (2003) Humanin rescues human cerebrovascular smooth muscle cells from Abeta-induced toxicity, J Neurochem 84, 266-272. [31] Charununtakorn, S. T., Shinlapawittayatorn, K., Chattipakorn, S. C., and Chattipakorn, N. (2016) Potential Roles of Humanin on Apoptosis in the Heart, Cardiovasc Ther 34, 107-114. 15

ACS Paragon Plus Environment

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 16 of 24

[32] Kim, S. J., Xiao, J., Wan, J., Cohen, P., and Yen, K. (2017) Mitochondrially derived peptides as novel regulators of metabolism, J Physiol 595, 6613-6621.

16

ACS Paragon Plus Environment

Page 17 of 24 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Chemical Neuroscience

Legends

Table

Table 1. Schematic for the microfluidic reactor for syntheses of compounds 1–8 and effect of small molecule HN mimetics on NMDA-triggered LDH release in primary hippocampal neurons.

17

ACS Paragon Plus Environment

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 18 of 24

Figures

Figure 1. The predicted binding site for HN, compound 2 and SC144 in the extracellular domains D4D6 of gp130. (A) HN, compound 2 and SC144 are predicted to bind to the interface region between extracellular domains 4 and 5 of gp130. The extracellular domains D4-D6 of gp130 and HN (green) are shown in the model. Compound 2 (magenta) and SC144 (cyan) are shown in stick representation. (B) Molecular surface representation of the gp130 extracellular domains 4-6 are shown with their electrostatic potential. (C) and (D) The gp130 residues involved in interaction with the compound 2 (magenta) and HN (green) are shown, respectively. Carbon atoms in the D4 domain are colored light brown while the carbon atoms in the D5 domain are shown in cyan. The residues within 5 Å radius of compound 2 or HN are shown in the models.

Figure 2. (A) Effect of co-treatment of gp130 antagonist, SC144, MK801 and compounds 2 and 8 on NMDA-triggered LDH release in cultured hippocampal neurons. NMDA increased LDH release (bar 2), and both MK801 and compound 2 rescued the LDH increase (bars 3 and 4, respectively). Antagonist SC144 prevented compound 2 rescue (bar 5). Compound 8 also rescued LDH and SC144 prevented this rescue (bars 6 and 7, respectively). Each bar represents mean ± SEM of four independent observations. Statistical significance was at P < 0.05. Effect of compound 2 on density of hippocampal neurons under NMDA-induced toxicity. Fluorescent microscopy was used to show representative calcein staining, and cellular density of each group. (B) Control (DMSO-only) density is shown. (C) A clear reduction of neuronal density is seen as a result of NMDA-induced toxicity. (D) With MK801 pretreatment, the density of NMDA-treated neurons was similar to the DMSO-only control. (E) Pretreatment with compound 2 also prevented NMDA-induced toxicity and the density of living neurons was similar to DMSO-only control. 18

ACS Paragon Plus Environment

Page 19 of 24 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Chemical Neuroscience

Figure 3. Compound 2 levels in mice brain and plasma. Mice were dosed orally at 10 or 30 mg/kg, or injected subcutaneously (SQ) at 10 mg/kg. Compound 2 is orally bioavailable at a dose of 30 mg/kg with a Cmax ~0.57 µM level in the brain at 1 h post dose.

19

ACS Paragon Plus Environment

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Table 1.

20

ACS Paragon Plus Environment

Page 20 of 24

Page 21 of 24 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Chemical Neuroscience

Figures Figure 1.

21

ACS Paragon Plus Environment

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Figure 2.

22

ACS Paragon Plus Environment

Page 22 of 24

Page 23 of 24 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Chemical Neuroscience

Figure 3.

23

ACS Paragon Plus Environment

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Table of Contents Artwork

24

ACS Paragon Plus Environment

Page 24 of 24