Barry J. Hoffer’s research while affiliated with Cleveland University and other places

What is this page?


This page lists works of an author who doesn't have a ResearchGate profile or hasn't added the works to their profile yet. It is automatically generated from public (personal) data to further our legitimate goal of comprehensive and accurate scientific recordkeeping. If you are this author and want this page removed, please let us know.

Publications (375)


Targeting neuroinflammation: 3-monothiopomalidomide a new drug candidate to mitigate traumatic brain injury and neurodegeneration
  • Article
  • Full-text available

June 2025

·

27 Reads

Journal of Biomedical Science

Shih Chang Hsueh

·

·

·

[...]

·

Background Traumatic Brain Injury (TBI) is a major risk factor for neurodegenerative disorders such as Parkinson’s disease (PD) and Alzheimer’s disease (AD), with neuroinflammation playing a critical role in the secondary cell death that exacerbates the initial injury. While targeting neuroinflammation holds significant therapeutic promise, clinical trials of available anti-inflammatory agents have fallen short. 3-Mono-thiopomalidomide (3-MP), a novel immunomodulatory imide drug (IMiD), was designed to curb inflammation without the adverse effects of traditional IMiDs and was evaluated across models involving neuroinflammation. Methods 3-MP anti-inflammatory activity was evaluated across cellular (RAW 264.7, IMG cells) and mouse studies following lipopolysaccharide (LPS)-challenge (for pro- and anti-inflammatory cytokines/chemokines), and mice subjected to controlled cortical impact (CCI) moderate traumatic brain injury (TBI). 3-MP human cereblon binding, including neosubstrate and molecular modeling evaluation, as well as chicken teratogenicity, ex vivo mouse and human stability studies, and mouse pharmacokinetics were appraised. Results 3-MP binds human cereblon, a key protein in the E3 ubiquitin ligase complex, without triggering downstream cascades leading to thalidomide-like teratogenicity in chicken embryos. 3-MP reduces pro-inflammatory markers in LPS-stimulated mouse macrophage and microglial cell cultures, and lowers pro-inflammatory cytokine/chemokine levels in plasma and brain of mice challenged with systemic LPS without lowering anti-inflammatory IL-10. 3-MP readily enters brain following systemic administration, and achieves a brain/plasma concentration ratio of 0.44–0.47. 3-MP mitigates behavioral impairments and reduces activation of astrocytes and microglia in mice challenged with CCI TBI. Conclusion 3-MP represents a promising new class of thalidomide-like IMiDs with potent anti-inflammatory effects that offers potential for treating TBI and possibly other neurodegenerative diseases possessing a prominent neuroinflammatory component.

Download

Fig. 1 3-MP nanosuspension time-dependent stability. 3-MP was formulated in Tween-80/water (approx. 1:130 v/v), vortexed with yttrium-stabilized zirconia-silica beads, stored at 4 °C, sampled over a 98-day period and quantified by LC-MS
Fig. 2 3-MP ameliorates LPS-induced inflammation in human PBMCs. Human PBMCs (5 × 10 5 cells) dose-dependently challenged with LPS (0.1-100 ng/ml, 24 h) demonstrated a small elevation in LDH (A) and a substantial rise in TNF-α levels (C) in media versus vehicle-treated controls (CNT). Dose-dependent 3-MP treatment of PBMCs challenged with
Fig. 3 Effects of 3-MP on LPS-induced changes in proinflammatory and anti-inflammatory cytokines in plasma and cerebral cortex. Systemic administration of LPS (1 mg/kg, I.P.) to rats induced significant changes in pro-(TNF-α and IL-6) and anti-inflammatory cytokines (IL-10 and IL-13) at 4 h in both A plasma and B the brain (cerebral cortex). One-hour pre-treatment with 3-MP (low dose: 13.23 mg/kg; high dose: 26.47 mg/ kg, I.P.) and pomalidomide (25 mg/kg, I.P.) significantly miti-
Fig. 4 Ex vivo metabolic profile of 3-MP in rat and human plasma and liver microsomes. A Plasma incubation, and B liver incubation of 3-MP in the presence of the cofactor NADPH
Fig. 5 3-MP pharmacokinetics. 3-MP was administered to rats via the intravenous (I.V.) and oral (P.O.) routes, and time-dependent plasma levels were obtained and subjected to LC-MS/ MS analysis (n = 3/group). Pharmacokinetic profile of 3-MP following a 5 mg/kg dose by either the I.V. or P.O route. Time-dependent plasma concentrations of 3-MP (n = 3/group)

+2

3-Monothiopomalidomide, a new immunomodulatory imide drug (IMiD), blunts inflammation and mitigates ischemic stroke in the rat

March 2025

·

57 Reads

·

1 Citation

GeroScience

An overactive neuroinflammatory response is often evident in the elderly and is a significant contributor to brain tissue damage following acute ischemic stroke. Such an inflammatory response is largely mediated by microglial cells and peripheral blood mononuclear cells (PBMCs). Classical anti-inflammatory agents have not proved clinically effective in mitigating the impact of ischemic stroke but have highlighted targets for new drug development, in particular excessive proinflammatory cytokine release. The immunomodulatory imide drug (IMiD) class has shown potential in reducing neuroinflammation and switching microglial phenotypic expression away from a proinflammatory to a regenerative anti-inflammatory one. 3-Monothiopomalidomide (3-MP), a new IMiD, has a brain/plasma concentration ratio of 0.5 to 0.6, an oral bioavailability of 38.5%, and a monophasic disappearance of half-life 3.2 h following oral administration. 3-MP pretreatment mitigates lipopolysaccharide (LPS)-induced inflammation in cellular human PBMCs and, in rat studies, 3-MP pretreatment lowers proinflammatory cytokine levels in the conditioned media and in plasma and the brain, respectively. Administered systemically to rats challenged with middle cerebral artery occlusion (MCAo) and reperfusion, 3-MP post-MCAo treatment reduced infarction volume; improved body asymmetry, a behavioral measure of stroke impact; and lowered inflammation. In summary, 3-MP exerted neuroprotective effects via anti-inflammatory actions against MCAo-induced ischemic injury and represents a therapeutic that warrants further investigation as a treatment for brain damage and related disorders associated with excessive inflammation.


Elevating brain GLP‐1 and GIP levels as a treatment strategy for neurodegenerative disorders

January 2025

·

36 Reads

Background Epidemiological studies report an elevated risk of neurodegenerative disorders, particularly Parkinson’s disease (PD), in patients with type 2 diabetes mellitus (T2DM) that is mitigated in those prescribed incretin mimetics or dipeptidyl peptidase 4 inhibitors (DPP‐4Is). Incretin mimetic repurposing appears promising in human PD and Alzheimer’s disease (AD) clinical trials. DPP‐4Is are yet to be evaluated in PD or AD human studies. Methods Incretin mimetics have been evaluated in multiple cellular/animal PD models, including in 6‐hdroxydopamine (6‐OHDA) rats, and have demonstrated efficacy. The clinically approved DPP‐4Is, sitagliptin (Januvia) and PF‐00734200 (gosogliptin/Saterex) were hence evaluated in the classic 6‐OHDA unilateral medial forebrain bundle rat PD model to evaluate their repurposing potential when administered as human equivalent oral doses. Equivalent doses then were administered to naive nonhuman primates (NHPs) to evaluate whether biomarkers of efficacy in rat could be reproduced in NHPs as a further translational step towards human studies. Pharmacokinetics, DPP‐4 inhibition, GLP‐1/GIP and dopamine levels, together with dopaminergic and neuroinflammatory markers, and GLP‐1/GIP receptor levels were quantified. Results Sitagliptin/PF‐00734,200 pre‐ or post‐treatment mitigated 6‐OHDA‐induced dopaminergic neurodegeneration, dopamine level loss and neuroinflammation, and augmented neurogenesis in lesioned substantia nigra pars compacta and in striatum, and reduced classical methamphetamine‐induced rotation in rats. This efficacy associated with 70‐80% plasma and 20‐30% brain DPP‐4 inhibition, and with elevated plasma and brain GLP‐1/GIP levels. Alike plasma/CSF DPP‐4 inhibition and elevated GLP‐1/GIP levels were determined in NHPs administered rat equivalent (human translational) sitagliptin doses. In relation to the drug targets, brain GLP‐1/GIP receptor protein levels were age‐dependently maintained in rodents, preserved in rats challenged with 6‐OHDA, and in humans with PD. Combined GLP‐1+GIP receptor activation in neuronal cultures resulted in neurotrophic/neuroprotective actions superior to single agonists alone – particularly when combined with a DPP‐4I Conclusions These studies support further evaluation of repurposing clinically approved drugs that elevate plasma/brain/CSF GLP‐1/GIP as a treatment strategy for neurodegenerative disorders. Incretin mimetics are already in clinical evaluation in PD and AD. Similarly, the repurposing of gliptins warrants evaluation both alone and in combination with an effective incretin mimetic.


Fig. 1 Weight drop induced mild traumatic brain injury and caused cortical function impairment in mice. A An illustration of the weight-drop-induced motor and sensory cortex injury site. B The protein levels of CCL5 in the cortex after 1-, 4-, and 7 days of injury (dpi) were detected by ELISA assay (sham vs. 1 dpi, p = 0.0420; sham vs. 4 dpi, p = 0.0177; sham vs. 7dpi, p = 0.0281. Data were presented as mean ± SEM and analyzed by t-test following Mann-Whitney test). C The mNSS score of both WT and CCL5-KO mice showed mild brain injury. (WT sham vs. TBI, p = 0.0038; KO sham vs. TBI, p < 0.0001; WT-TBI vs. KO-TBI, p < 0.0001). The motor function of 4 groups of mice included falling time from the accelerating Rotarod (WT sham vs. TBI, p < 0.0001; KO sham vs. TBI, p < 0.0001; WT-TBI vs. KO-TBI, p = 0.0002) (D) and foot faults with beam walking (WT sham vs. TBI, p = 0.0011; KO sham vs. TBI, p = 0.0019; WT-TBI vs. KO-TBI, p < 0.0001) (E). Sensory function was analyzed by limb sticker removal (WT sham vs. TBI, p = 0.0003; KO sham vs. TBI, p = 0.0173; WT-TBI vs. KO-TBI, p = 0.0399) (F). Data was analyzed from both paws. (n = 7-9 animals in C-F) Data in C-F were analyzed by two-way ANOVA and presented as mean ± SEM. The time of induced brain injury (mild traumatic brain injury, mTBI)
Fig. 2 The recovery of axonal injury was impaired in the CCL5-KO cortex after mild TBI. Golgi staining revealed the axon and spine structures in WT and CCL5-KO mouse cortex with sham treatment and mild TBI -14 and − 28 days of injury (dpi). A, F The representative images of neurites and spine structures in WT and CCL5-KO mouse cortex; boxed regions were enlarged on the right. Black arrowheads point to the normal dendritic spines, and white arrows point to swollen neurites and spines. Scale bar = 1 mm and 20 μm. B, G The number of intersections (WT sham vs. 14 dpi, p < 0.0001; WT 14 dpi vs. 28 dpi, p < 0.0001; KO sham vs. 14 dpi, p < 0.0001; KO 14 dpi vs. 28 dpi, p < 0.0001. Data were analyzed by two-way ANOVA and presented as mean ± SEM), C, H total intersections (WT sham vs. 14 dpi, p = 0.0018; WT 14 dpi vs. 28 dpi, p = 0.0493; KO sham vs. 14 dpi, p < 0.0001; KO 14 dpi vs. 28 dpi, p = 0.0004.), D, I spine density (WT sham vs. 14 dpi, p < 0.0001; WT 14 dpi vs. 28 dpi, p < 0.0001; WT sham vs. 28 dpi, no significant difference, NS; KO sham vs. 14 dpi, p < 0.0001; KO 14dpi vs. 28 dpi, p = 0.0399; KO sham vs. 28 dpi, p < 0.0001.), and E, J swollen spines (WT sham vs. 14 dpi, p < 0.0001; WT 14 dpi vs. 28 dpi, p < 0.0001; WT sham vs. 28 dpi, p = 0.0231; KO sham vs. 14 dpi, p = 0.0004; KO 14dpi vs. 28dpi, p = 0.0229; KO sham vs. 28 dpi, p < 0.0001.) were quantified in different groups of mice (n = 10 in each group). Data in C-E and H-J were analyzed by unpaired t-test and presented as mean ± SEM. K, L The expression of synaptic proteins -PSD95 and synaptophysin in different groups of WT and CCL5-KO mouse cortex, including sham, 4, 7, 14, and 28 dpi, was analyzed by western blot. Quantification of results from 3 independent mouse samples in each group is listed above the images of protein blots in 2 K-L. (KO PSD95: sham vs. 4 dpi, p = 0.0015; sham vs. 7 dpi, p = 0.0038; sham vs. 14 dpi, p = 0.0147; sham vs. 28 dpi, p = 0.0025. KO Synaptophysin: sham vs. 4 dpi, p = 0.0126; sham vs. 7 dpi, p = 0.0019; sham vs. 14 dpi, p = 0.0089; sham vs. 28 dpi, p = 0.0186. Data were presented as mean ± SEM and analyzed by t-test following Mann-Whitney test)
Fig. 5 Both pretreatment and post-treatment with CCL5 enhanced neurite and synapse growth and myelination-related signaling pathways in injured cortical tissue. A Volcano plot of significant DEPs between mTBI and TBI + CCL5 pretreatment (PreL5) CCL5-KO mouse cortex. B Volcano plot of significant DEPs between mTBI and TBI + CCL5 post-treatment (PostL5) CCL5-KO mouse cortex. DEPs: p-value < 0.05 in comparison to TBI, respectively. Colored points represent: log2 ratio > 0 upregulated protein (red) and log2 ratio < 0 downregulated protein (blue). Selected axonogenesis, neuritogenesis, synaptogenesis, and myelination pathway-related proteins are highlighted as indicated (Red: myelinations, Green: axonogenesis, neuritogenesis, and synaptogenesis, Yellow: overlapping). C Venn diagram comparing DEPs between the TBI group, TBI + CCL5 pretreatment (PreL5), and TBI + CCL5 post-treatment (PostL5) groups. 54 identified proteins (46 up regulated, 8 down regulated) were affected by both treatments. D Identified GO terms from each of the three GO groups (Green bar: biological process, Red bar: cellular component, Blue bar: molecular function. Blue character: neuron function-related) were shown. The strength of enrichment of each GO term was indicated by the Log10 p-value (X-axis). E IPA analysis identified affected diseases and functions in the nervous system category (Z-score value indicated that functions are predicted to be activated (red). Selected IPA canonical pathways in the nervous system identified significant DEPs related to axon (F), synapse (G), neuron development (H), and myelination (I) related signaling pathways in both PreL5 and PostL5 treatments. The Z-score in different identified pathways was shown as a gradient of yellow to red. See also Supplementary Figs. 3, 4, and 5
CCL5 is essential for axonogenesis and neuronal restoration after brain injury

September 2024

·

57 Reads

·

7 Citations

Journal of Biomedical Science

Background Traumatic brain injury (TBI) causes axon tearing and synapse degradation, resulting in multiple neurological dysfunctions and exacerbation of early neurodegeneration; the repair of axonal and synaptic structures is critical for restoring neuronal function. C-C Motif Chemokine Ligand 5 (CCL5) shows many neuroprotective activities. Method A close-head weight-drop system was used to induce mild brain trauma in C57BL/6 (wild-type, WT) and CCL5 knockout (CCL5-KO) mice. The mNSS score, rotarod, beam walking, and sticker removal tests were used to assay neurological function after mTBI in different groups of mice. The restoration of motor and sensory functions was impaired in CCL5-KO mice after one month of injury, with swelling of axons and synapses from Golgi staining and reduced synaptic proteins-synaptophysin and PSD95. Administration of recombinant CCL5 (Pre-treatment: 300 pg/g once before injury; or post-treatment: 30 pg/g every 2 days, since 3 days after injury for 1 month) through intranasal delivery into mouse brain improved the motor and sensory neurological dysfunctions in CCL5-KO TBI mice. Results Proteomic analysis using LC-MS/MS identified that the “Nervous system development and function”-related proteins, including axonogenesis, synaptogenesis, and myelination signaling pathways, were reduced in injured cortex of CCL5-KO mice; both pre-treatment and post-treatment with CCL5 augmented those pathways. Immunostaining and western blot analysis confirmed axonogenesis and synaptogenesis related Semaphorin, Ephrin, p70S6/mTOR signaling, and myelination-related Neuregulin/ErbB and FGF/FAK signaling pathways were up-regulated in the cortical tissue by CCL5 after brain injury. We also noticed cortex redevelopment after long-term administration of CCL5 after brain injury with increased Reelin positive Cajal-Rerzius Cells and CXCR4 expression. CCL5 enhanced the growth of cone filopodia in a primary neuron culture system; blocking CCL5’s receptor CCR5 by Maraviroc reduced the intensity of filopodia in growth cone and also CCL5 mediated mTOR and Rho signalling activation. Inhibiting mTOR and Rho signaling abolished CCL5 induced growth cone formation. Conclusions CCL5 plays a critical role in starting the intrinsic neuronal regeneration system following TBI, which includes growth cone formation, axonogenesis and synaptogensis, remyelination, and the subsequent proper wiring of cortical circuits. Our study underscores the potential of CCL5 as a robust therapeutic stratagem in treating axonal injury and degeneration during the chronic phase after mild brain injury. Graphical Abstract


Serotonergic Regulation of Synaptic Dopamine Levels Mitigates L-DOPA-Induced Dyskinesia in a Mouse Model of Parkinson’s Disease

June 2024

·

121 Reads

·

2 Citations

Background The serotonin (5-HT) system can manipulate the processing of exogenous L-DOPA in the DA-denervated striatum, resulting in the modulation of L-DOPA-induced dyskinesia (LID). Objective To characterize the effects of the serotonin precursor 5-hydroxy-tryptophan (5-HTP) or the serotonin transporter (SERT) inhibitor, Citalopram on L-DOPA-induced behavior, neurochemical signals, and underlying protein expressions in an animal model of Parkinson’s disease. Methods MitoPark (MP) mice at 20 weeks of age, subjected to a 14-day administration of L-DOPA/Carbidopa, displayed dyskinesia, referred to as LID. Subsequent investigations explored the effects of 5-HT-modifying agents, such as 5-HTP and Citalopram, on abnormal involuntary movements (AIMs), locomotor activity, neurochemical signals, serotonin transporter activity, and protein expression in the DA-denervated striatum of LID MP mice. Results 5-HTP exhibited duration-dependent suppressive effects on developing and established LID, especially related to abnormal limb movements observed in L-DOPA-primed MP mice. However, Citalopram, predominantly suppressed abnormal axial movement induced by L-DOPA in LID MP mice. We demonstrated that 5-HTP could decrease L-DOPA-upregulation of DA turnover rates while concurrently upregulating 5-HT metabolism. Additionally, 5-HTP was shown to reduce the expressions of p-ERK and p-DARPP-32 in the striatum of LID MP mice. The effect of Citalopram in alleviating LID development may be attributed to downregulation of SERT activity in the dorsal striatum of LID MP mice. Conclusions While both single injection of 5-HTP and Citalopram effectively mitigated the development of LID, the difference in mitigation of AIM subtypes may be linked to the unique effects of these two serotonergic agents on L-DOPA-derived DA and 5-HT metabolism.


Chronic inflammation with microglia senescence at basal forebrain: impact on cholinergic deficit in Alzheimer’s brain hemodynamics

June 2024

·

27 Reads

·

3 Citations

Brain Communications

Cholinergic innervation in brain is involved in modulating neurovascular function including cerebral blood flow hemodynamics in response to neuronal activity. Cholinergic deficit is associated with pathophysiology in Alzheimer’s disease, albeit the etiology remains to clarify. In current study, neocortex cerebral blood flow response to acetylcholine was evaluated by Laser-Doppler Flowmetry in 3xTgAD Alzheimer’s disease model) and wild type mice of two age groups. The peak of cerebral blood flow to acetylcholine (i.v.) from baseline levels (% ΔrCBF) were higher in young 3xTgAD vs. in wild type mice (48.35; CI:27.03-69.67 vs. 22.70; CI:15.5-29.91, p < 0.05); this was reversed in old 3xTgAD mice (21.44; CI:2.52-40.35 vs. 23.25; CI:23.25-39). Choline acetyltransferase protein was reduced in neocortex, while cerebrovascular reactivity to acetylcholine was preserved in young 3xTgAD mice. This suggests of endogenous acetylcholine deficit and possible cholinergic denervation from selected cholinergic nuclei within basal forebrain. The early deposition of tauopathy moieties (mutant hTau and pTau181) and its coincidence in cholinergic cell clusters, were observed occasionally at basal forebrain of 3xTgAD mice including substantia innominate, nucleus Basalis of Meynert and nucleus of horizontal limb diagonal band of Broca. A prominent feature was microglia interacting tauopathy and demonstrated a variety of morphology changes particularly when located in proximity to tauopathy. The microglia ramified phenotype was reduced as evaluated by the ramification index and Fractal analysis. Increased microglia senescence, identified as SASP (senescence-associated secretory phenotype), was colocalization with p16Ink4ɑ, a marker of irreversible cell cycle arrest in old 3xTgAD vs. wild type mice (p = 0.001). The p16Ink4ɑ was also observed in neuronal cells bearing tauopathy within basal forebrain of 3xTgAD mice. TNF-ɑ, the pro-inflammatory cytokine elevated persistently in microglia (Peason’s correlation coefficient = 0.62), and the loss of cholinergic cells in vulnerable basal forebrain environment was indicated by image analysis in 3xTgAD mice, which linked to the cholinergic deficits in neocortex rCBF hemodynamics. Our study revealed the early change of CBF hemodynamics to acetylcholine in 3xTgAD model. As a major effector of brain innate immune activation, microglia SASP with age related disease progression is indicative of immune cell senescence, which contributes to chronic inflammation and cholinergic deficits at basal forebrain. Targeting neuroinflammation and senescence may mitigate cholinergic pathophysiology in Alzheimer’s disease.



Fig. 1 Changes in TH protein expression at three MitoPark and PT320-treated MP mouse PD stages. Results show at the level of the substantia nigra pars compacta (SNc), TH starts to decline during the middle stage. The PT320 treatment showed significant differences at the late stage. The TH expression of the striatum area showed little change at the early stage but substantial changes with PT320-treated groups in the middle and late stages, as compared to untreated MitoPark animals. A Representative immunofluorescent images of TH expression in the SNc. Scale bar: 0.5 mm (B) Western blot results of TH expression and C The statistical results in SNc. D Representative immunofluorescent images of TH expression in the striatum. Scale bar: 1 mm (E) Western blot results of TH expression and F The statistical results in striatum. One-way analysis of variance (ANOVA) followed by Bonferroni post hoc test for multiple comparisons. WT vs MP or MP+PT320: *, p < 0.05; **, p < 0.005; ***, p < 0.001; MP vs MP+PT320: #, p < 0.05; ##, p < 0.005. (N = 3-5)
Fig. 4 TEM analysis of mitochondrial structural changes in the striatum region in wild-type (WT), MitoPark, and PT320-treated MitoPark mice. The mitochondrial structures can be divided into four types as detailed in Results. Type I is the normal structure of healthy mitochondria. Type IV is the most damaged mitochondria, with disrupted and discontinuous outer membranes and deficient cristae. The red arrows indicate the different types of mitochondria. A Representative TEM images of four types of mitochondrial morphologies. Scale bar: 500 nm (B) Representative TEM images of different stages of the mitochondria morphologies in three groups. Scale bar: 1 µm (C) Percent of mitochondria type in early stage. D Percent of mitochondria type in middle stage. E Percent of mitochondria type in late stage. One-way analysis of variance (ANOVA) followed by Bonferroni post hoc test for multiple comparisons. WT vs MP or MP+PT320: *, p < 0.01; **, p < 0.05; ***, p < 0.001; MP vs MP+PT320: #, p < 0.01; ##, p < 0.05; ###, p < 0.001. (N = 3)
Fig. 7 The mitochondrial complex II is composed of Succinate dehydrogenase complex subunits A, B, C, and D (SDHA, SDHB, SDHC, and SDHD). The data here shows inconsistencies between the protein levels and RNA levels, indicating that the protective effects of PT320 may not directly affect the mitochondrial complex. A Protein level results of SDHA, SDHB, SDHC, and SDHD. B The heat map of the complex II associated RNA expression in Next-Generation Sequencing (NGS) data. C The real-time polymerase chain reaction (rtPCR) results of complex II composed genes. (N = 3-6)
Fig. 8 Results of 6 selected genes which are mitochondrial morphology controllers. Although the fold change was not significant, we choose the genes which the FDR p-value <0.05 to conduct qPCR analysis, indicating a potential involvement in mitochondrial morphology control. Confirmation of the RNA levels and protein expressions of Opa1, Mfn2, and Fis1, with FDR p-values <0.05. The results indicate that PT320 might have a major influence on Opa1 and Fis1, but not on Mfn2. A Summary of NGS results of selected genes. B The qPCR results of the Opa1, Mfn2, and Fis1 genes. C Protein expression of Opa1, Mfn2, and Fis1. (N = 3-6)
Attenuating mitochondrial dysfunction and morphological disruption with PT320 delays dopamine degeneration in MitoPark mice

April 2024

·

98 Reads

·

6 Citations

Journal of Biomedical Science

Background Mitochondria are essential organelles involved in cellular energy production. Changes in mitochondrial function can lead to dysfunction and cell death in aging and age-related disorders. Recent research suggests that mitochondrial dysfunction is closely linked to neurodegenerative diseases. Glucagon-like peptide-1 receptor (GLP-1R) agonist has gained interest as a potential treatment for Parkinson's disease (PD). However, the exact mechanisms responsible for the therapeutic effects of GLP-1R-related agonists are not yet fully understood. Methods In this study, we explores the effects of early treatment with PT320, a sustained release formulation of the GLP-1R agonist Exenatide, on mitochondrial functions and morphology in a progressive PD mouse model, the MitoPark (MP) mouse. Results Our findings demonstrate that administration of a clinically translatable dose of PT320 ameliorates the reduction in tyrosine hydroxylase expression, lowers reactive oxygen species (ROS) levels, and inhibits mitochondrial cytochrome c release during nigrostriatal dopaminergic denervation in MP mice. PT320 treatment significantly preserved mitochondrial function and morphology but did not influence the reduction in mitochondria numbers during PD progression in MP mice. Genetic analysis indicated that the cytoprotective effect of PT320 is attributed to a reduction in the expression of mitochondrial fission protein 1 (Fis1) and an increase in the expression of optic atrophy type 1 (Opa1), which is known to play a role in maintaining mitochondrial homeostasis and decreasing cytochrome c release through remodeling of the cristae. Conclusion Our findings suggest that the early administration of PT320 shows potential as a neuroprotective treatment for PD, as it can preserve mitochondrial function. Through enhancing mitochondrial health by regulating Opa1 and Fis1, PT320 presents a new neuroprotective therapy in PD. Graphical Abstract


DPP-4 inhibitors sitagliptin and PF-00734,200 mitigate dopaminergic neurodegeneration, neuroinflammation and behavioral impairment in the rat 6-OHDA model of Parkinson’s disease

April 2024

·

72 Reads

·

12 Citations

GeroScience

Epidemiological studies report an elevated risk of Parkinson's disease (PD) in patients with type 2 diabetes mellitus (T2DM) that is mitigated in those prescribed dipeptidyl peptidase 4 (DPP-4) inhibitors. With an objective to characterize clinically translatable doses of DPP-4 inhibitors (gliptins) in a well-characterized PD rodent model, sitagliptin, PF-00734,200 or vehicle were orally administered to rats initiated either 7-days before or 7-days after unilateral medial forebrain bundle 6-hydroxydopamine (6-OHDA) lesioning. Measures of dopaminergic cell viability, dopamine content, neuroinflammation and neurogenesis were evaluated thereafter in ipsi- and contralateral brain. Plasma and brain incretin and DPP-4 activity levels were quantified. Furthermore, brain incretin receptor levels were age-dependently evaluated in rodents, in 6-OHDA challenged animals and human subjects with/without PD. Cellular studies evaluated neurotrophic/neuroprotective actions of combined incretin administration. Pre-treatment with oral sitagliptin or PF-00734,200 reduced methamphetamine (meth)-induced rotation post-lesioning and dopaminergic degeneration in lesioned substantia nigra pars compacta (SNc) and striatum. Direct intracerebroventricular gliptin administration lacked neuroprotective actions, indicating that systemic incretin-mediated mechanisms underpin gliptin-induced favorable brain effects. Post-treatment with a threefold higher oral gliptin dose, likewise, mitigated meth-induced rotation, dopaminergic neurodegeneration and neuroinflammation, and augmented neurogenesis. These gliptin-induced actions associated with 70–80% plasma and 20–30% brain DPP-4 inhibition, and elevated plasma and brain incretin levels. Brain incretin receptor protein levels were age-dependently maintained in rodents, preserved in rats challenged with 6-OHDA, and in humans with PD. Combined GLP-1 and GIP receptor activation in neuronal cultures resulted in neurotrophic/neuroprotective actions superior to single agonists alone. In conclusion, these studies support further evaluation of the repurposing of clinically approved gliptins as a treatment strategy for PD. Supplementary Information The online version contains supplementary material available at 10.1007/s11357-024-01116-0.


Fig. 1 Nonhuman primate protocol. Vehicle or one of three doses of sitagliptin (5, 20, or 100 mg/kg) was administered orally once daily for 6 consecutive days to rhesus monkeys (7-20 kg weight, mixed gender, 12 to 19 years of age, n = 5 to 7 per group) to attain steady-state conditions prior to a classical oral glucose load, which was administered on the "test day" (day 6) to initiate endogenous incretin (GLP-1 and GIP) release, since incretin release from L and K cells is stimulated by food presence within the gastrointestinal tract. Time-
Fig. 3 CSF sitagliptin levels in nonhuman primates. CSF dose-dependent sitagliptin pharmacokinetics in NHPs and CSF/plasma concentration ratio were obtained at 3 and 5 h post sitagliptin oral administration. All values are mean ± SEM
Fig. 7 Combined DPP-4 inhibition and incretins mitigate 6-OHDA-mediated cell loss in cultured SH-SY5Y neuronal cells. (A) Representative photomicrographs indicate that challenge with 100 µM 6-OHDA (left lower panel) reduced cell density as compared to the control (left upper panel) (scale bar = 100 µm). (A, right panels) Cell nuclei were stained with TOPRO-3 following 24-h drug treatment/6-OHDA challenge. Cell density across 96-well plates was measured by a LiCor Odyssey image system. Three representative TOPRO-3 images were obtained from cells administered vehicle, 6-OHDA, and 6-OHDA + GLP-1 + DPP-4 inhibitor (DPP-4I: PF-000734,200,
Sitagliptin elevates plasma and CSF incretin levels following oral administration to nonhuman primates: relevance for neurodegenerative disorders

March 2024

·

45 Reads

·

2 Citations

GeroScience

The endogenous incretins glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) possess neurotrophic, neuroprotective, and anti-neuroinflammatory actions. The dipeptidyl peptidase 4 (DPP-4) inhibitor sitagliptin reduces degradation of endogenous GLP-1 and GIP, and, thereby, extends the circulation of these protective peptides. The current nonhuman primate (NHP) study evaluates whether human translational sitagliptin doses can elevate systemic and central nervous system (CNS) levels of GLP-1/GIP in naive, non-lesioned NHPs, in line with our prior rodent studies that demonstrated sitagliptin efficacy in preclinical models of Parkinson’s disease (PD). PD is an age-associated neurodegenerative disorder whose current treatment is inadequate. Repositioning of the well-tolerated and efficacious diabetes drug sitagliptin provides a rapid approach to add to the therapeutic armamentarium for PD. The pharmacokinetics and pharmacodynamics of 3 oral sitagliptin doses (5, 20, and 100 mg/kg), equivalent to the routine clinical dose, a tolerated higher clinical dose and a maximal dose in monkey, were evaluated. Peak plasma sitagliptin levels were aligned both with prior reports in humans administered equivalent doses and with those in rodents demonstrating reduction of PD associated neurodegeneration. Although CNS uptake of sitagliptin was low (cerebrospinal fluid (CSF)/plasma ratio 0.01), both plasma and CSF concentrations of GLP-1/GIP were elevated in line with efficacy in prior rodent PD studies. Additional cellular studies evaluating human SH-SY5Y and primary rat ventral mesencephalic cultures challenged with 6-hydroxydopamine, established cellular models of PD, demonstrated that joint treatment with GLP-1 + GIP mitigated cell death, particularly when combined with DPP-4 inhibition to maintain incretin levels. In conclusion, this study provides a supportive translational step towards the clinical evaluation of sitagliptin in PD and other neurodegenerative disorders for which aging, similarly, is the greatest risk factor.


Citations (72)


... Unlike 3,6′-DTP that rapidly disappears on addition to mouse and human plasma samples, 3-MP proved to be stable in plasma (Fig. 4A), in accord with its stability in rat plasma [64]. In in vivo studies in mouse, Fig. 4B shows the time-dependent plasma and brain drug levels achieved by 3-MP following its I.P. administration at doses of 26.47 mg/kg and 2.65 mg/kg. ...

Reference:

Targeting neuroinflammation: 3-monothiopomalidomide a new drug candidate to mitigate traumatic brain injury and neurodegeneration
3-Monothiopomalidomide, a new immunomodulatory imide drug (IMiD), blunts inflammation and mitigates ischemic stroke in the rat

GeroScience

... In contrast, the MARE groups at both time points showed a moderate trend toward positive correlation with very similar slopes. After injury, the chemokine CCL5 reduces oxidative stress, aids in neuronal ATP generation, and promotes axon and synapse regeneration 105 . However, CCL5 has also been shown to have pro-in ammatory effects on microglia, and its overexpression has been associated with memory impairment in mice 106,107 . ...

CCL5 is essential for axonogenesis and neuronal restoration after brain injury

Journal of Biomedical Science

... A genetic mouse model of Parkinson's disease with mitochondrial dysfunction selectively in dopaminergic neurons demonstrated that mitochondrial morphology defects preceded both neuronal impairment and motor deficits. These early mitochondrial abnormalities also responded to treatment, highlighting their potential as early indicators of disease progression and therapeutic efficacy 33 . To assess mitochondrial morphology, mitochondria were classified into four distinct types based on their morphology: type I mitochondria -healthy mitochondria, type II mitochondria -moderately affected mitochondria, type III mitochondria -severely affected mitochondria and type IV mitochondria -dysfunctional mitochondria exhibited disrupted outer membrane, deficiency in cristae, absence of the internal membrane structures 33 . ...

Attenuating mitochondrial dysfunction and morphological disruption with PT320 delays dopamine degeneration in MitoPark mice

Journal of Biomedical Science

... The protective effects of other DPP4 inhibitors, such as saxagliptin, vildagliptin, and sitagliptin, against PD have been reported in prior studies (Svenningsson et al. 2016;Alrouji et al. 2023). Oral treatment with sitagliptin ameliorates the motor impairment and dopaminergic neuronal death induced by 6-OHDA lesions in rodents, which involves the preservation of GLP-1 (Yu et al. 2024). The present study provides evidence that sitagliptin modulates the striatal expression of proteins linked to mitochondrial function and cytoskeletal organisation in dyskinetic mice. ...

DPP-4 inhibitors sitagliptin and PF-00734,200 mitigate dopaminergic neurodegeneration, neuroinflammation and behavioral impairment in the rat 6-OHDA model of Parkinson’s disease

GeroScience

... In rodents, pomalidomide has been shown to have favorable permeability of the blood-brain barrier and exert its therapeutic effects by affecting inflammation-related circuits. In Parkinson's and Alzheimer's disease pomalidomide treatment reduced proinflammatory cytokine generation and loss of synapses and neurons and through this way, it caused a partial recovery of the disease (Palmas et al. 2022;Greig et al. 2023). Given pomalidomide's immunomodulatory properties and its capacity to cross the blood-brain barrier, it makes it a promising candidate for repurposing in neurodegenerative disorders (Munafò et al. 2020), including epileptic seizures. ...

Mitigating chronic neuroinflammation in Alzheimer mice by novel immunomodulatory imide drugs (IMiDs) conserves neuroplasticity and cognitive function and also alleviates neurodegeneration‐behavioral impairments in TBI‐challenged mice
  • Citing Article
  • December 2023

... LPS has been known as a ligand for Toll-like receptor 4 (TLR4), which is mainly expressed on microglia in the CNS, and thus microglia might be the main target of LPS in the CNS [73,74]. Consequently, establishing an in vitro model of LPS-induced neuroinflammation using cell lines is crucial for studying microglial behavior under inflammatory conditions [75]. The BV2 cell line, a classical cell line used to mimic microglia in experimental in vitro studies, can effectively respond to LPS stimulation and is widely used to study neuroinflammation and a variety of CNS degenerative diseases [76,77]. ...

The RhoA-ROCK1/ROCK2 Pathway Exacerbates Inflammatory Signaling in Immortalized and Primary Microglia

... 25 This feature represents a key advantage in mass spectrometry, where different imaging modalities can be used in a complementary fashion since molecules display different propensities for ionization. 26,27 The vast majority of MSI studies involve mapping the distribution of therapeutics and biomolecules, including lipids, 28,29,30 proteins, 31,32,33 peptides, 34 carbohydrates, 35,36 and other metabolites 37 . ...

MALDI-MSI of lipids in a model of breast cancer brain metastasis provides a surrogate measure of ischemia/hypoxia

Molecular and Cellular Biochemistry

... For this study, both cultured RAW 264.7 and IMG cells were pre-administered either vehicle or 3-MP (0.6μM to 60μM), and then challenged with LPS at a concentration of 20 ng/mL and 10 ng/mL, respectively, one hour later to induce inflammation. Following this procedure, these mouse immortalized macrophage/microglia cells provide a useful phenotypic model to screen for antiinflammatory drug action [44,62,63], as they share features with brain-derived microglial cells in response to a pro-inflammatory signal. After 24 h of LPS exposure, cellular viability along with levels of nitrite (a stable marker of nitric oxide generation) and TNF-α were quantified in both cell lines. ...

Novel, thalidomide-like, non-cereblon binding drug tetrafluorobornylphthalimide mitigates inflammation and brain injury

Journal of Biomedical Science

... A sustained-release exenatide agent, PT302, was found to sustain dopaminergic neurons after 6-OHDA lesioning in rats [48]. Interestingly, in a recent study in a PD mouse model, early treatment with PT320 ameliorated L-DOPA-induced dyskinesia, highlighting its possible ability to mitigate dopaminergic degeneration [49]. Notably, in another study, lixisenatide and liraglutide were found to be more effective regarding protection against MPTP-induced dopaminergic degeneration compared to exenatide [50]; however, these results need to be replicated [51]. ...

PT320, a Sustained-Release GLP-1 Receptor Agonist, Ameliorates L-DOPA-Induced Dyskinesia in a Mouse Model of Parkinson’s Disease

... Such failure has led to a closer examination of microglial dysfunction and the resulting chronic neuroinflammation as a potential target for slowing down AD progression [21]. Increasingly, neuroinflammation is not just considered as a secondary response to Aβ accumulation and tau pathology, but also as a causative factor in the etiology of AD [22]. Several studies imply that neuroinflammatory conditions develop much before the onset of clinical symptoms of AD, which has led to the hypothesis that neuroinflammation drives the disease progression independently of the Aβ plaques and neurofibrillary tangles, and interaction of these three factors exacerbates the disease progression [23]. ...

Targeting neuroinflammation reduces synaptic, neuronal and cognitive loss in 5xFAD Alzheimer mice.
  • Citing Article
  • December 2022