Introduction
Neurodegenerative disorders such as Alzheimer’s disease (AD) and Parkinson’s disease (PD) emerge from intersecting processes—proteostasis failure, maladaptive stress signaling, metabolic compromise, and inflammation—that culminate in progressive neuronal dysfunction and death. A central translational challenge is delivery: many candidate modulators of these pathways fail at the blood–brain barrier (BBB) or at the neuronal plasma membrane, narrowing the toolbox for probing causality in vivo. Cell-penetrating peptides (CPPs) have therefore attracted sustained research interest as modular shuttles capable of ferrying diverse cargos—peptides, proteins, nucleic acids, and small molecules—across biological barriers while retaining bioactivity at intracellular targets.
In parallel, the c-Jun N-terminal kinase (JNK) family has been implicated as a nodal stress relay that can tip cells toward apoptosis when activated chronically or excessively. JNKs also participate in physiological functions, including synaptic plasticity and development, implying that broad inhibition requires nuance in timing, isoform selectivity, and brain region context. Coupling CPP delivery platforms with JNK-directed inhibitory motifs has become a prominent experimental approach to test whether dampening JNK signaling can attenuate neuronal degeneration in models of AD, PD, and related conditions. The sections below synthesize these lines of inquiry with attention to mechanistic caveats and areas where evidence remains preliminary.
Reimagining Delivery: Modular CPP Platforms for Crossing Biological Barriers
Classical pharmacology is often thwarted by the BBB’s tight junctions and efflux transporters, and even when a compound gains CNS access, membrane impermeability can trap it extracellularly. CPPs—short, typically cationic or amphipathic sequences exemplified by TAT, penetratin, Arg-rich motifs, and Pep-1—offer a flexible scaffold to enhance translocation through endocytosis-dependent and, in some contexts, direct translocation routes. In experimental systems, CPPs have delivered kinase inhibitors, proteostasis modulators, and regulatory peptides into neurons and glia with measurable intracellular readouts. Their small size eases synthesis and conjugation to cargos, while sequence tuning can bias uptake mechanisms or subcellular localization. Notably, CPPs themselves may confer context-specific neuroprotective signals, complicating attribution of effects solely to the attached cargo. This duality underscores why rigorous controls—cargo-free CPPs, scrambled sequences, dose–response mapping, and orthogonal delivery methods—are essential when interpreting neuroprotection attributed to CPP conjugates.
Stress Relays at a Crossroads: JNK Isoforms in Neuronal Signaling
The JNK family (JNK-1/2/3) integrates inputs from oxidative stress, inflammatory cytokines, excitotoxicity, and proteotoxic species. Downstream, JNKs phosphorylate transcription factors (e.g., c-Jun) and cytosolic substrates, modulating apoptosis programs, autophagy flux, cytoskeletal dynamics, and synaptic remodeling. JNK-1 and JNK-2 are broadly expressed, whereas JNK-3 is enriched in brain and testis, motivating early neuro-centric inhibition efforts focused on JNK-3. Experimental inhibition of JNK-3 has yielded neuroprotective readouts in several paradigms; however, accumulating data suggest that combined modulation of multiple isoforms can more effectively blunt pro-apoptotic cascades triggered by chronic stressors. This broader strategy, while promising, must balance the risk of impairing homeostatic roles of JNKs in plasticity, development, and immune signaling. Isoform-selective, temporally controlled inhibition—rather than blanket suppression—therefore appears to be a rational design goal for future constructs.
Pathology Interface: Convergence of Aβ, Tau, and α-Syn with JNK Pathways
Misfolded protein species central to AD and PD pathophysiology interface bidirectionally with JNK signaling. In AD-like models, extracellular Aβ accumulation can activate JNK-3 and has been linked to shifts toward Aβ42 production; JNK activity may also intersect with tau phosphorylation states that influence microtubule stability and synaptic function. In PD models, α-synuclein burden and mitochondrial stress can engage JNK cascades, promoting apoptosis and neuroinflammation. These observations position JNK as a convergence hub rather than a solitary driver—its activation may both respond to and reinforce proteostatic stress, inflammation, and autophagy bottlenecks. Consequently, experimental JNK damping is being explored alongside strategies that normalize autophagy, bolster mitochondrial resilience, or modulate innate immune tone, with the hypothesis that multi-axis correction could be required to produce durable neuroprotection.
Cargo on Board: CPP–JNK Inhibitor Conjugates as Proof-of-Concept Tools
To interrogate whether intracellular JNK signaling is a tractable lever in vivo, investigators have fused CPPs (notably TAT) to peptide inhibitors that block JNK–substrate interactions (e.g., JNKI-1–derived motifs). These chimeras have shown the ability to access brain tissue and reduce readouts of neuronal death in cellular stress assays and in rodent models relevant to AD/PD-like pathology, ischemia, and excitotoxic injury. Some reports indicate improved synaptic markers, dampened neuroinflammation, and partial preservation of behavioral performance. At the same time, studies variably suggest that broader inhibition across JNK-1/2/3 might outperform JNK-3-focused designs, especially when apoptosis is driven by network-wide stressors. Open questions remain regarding pharmacokinetics, endosomal escape efficiency, regional exposure heterogeneity, and long-term consequences of repeated pathway suppression. These uncertainties motivate continued optimization of sequence composition, linker chemistry, and dosing paradigms, as well as head-to-head comparisons with non-CPP delivery systems.
Systems Perspective: Potential Benefits, Trade-offs, and Measurement Priorities
Reports of JNK inhibition in neurodegeneration models frequently note improvements consistent with a shift from pro-death to pro-survival signaling: increased autophagy competence and misfolded-protein clearance, reduced caspase activation, lower inflammatory cytokine signatures, and partial restoration of synaptic connectivity and gene-expression programs. Yet, because JNKs modulate development, plasticity, and immune responses, broad or chronic suppression could, in principle, impede adaptive remodeling or host defense. Future work may benefit from: (i) isoform- and substrate-selective inhibitors that spare beneficial nodes; (ii) spatiotemporal control (e.g., activity-dependent or pathology-triggered release); (iii) multiplex readouts (proteostasis, mitochondrial function, electrophysiology, and behavior) to capture trade-offs; and (iv) careful disentangling of CPP-intrinsic effects from cargo actions. Collectively, these priorities could clarify when, where, and how much JNK attenuation contributes to meaningful neuroprotection in complex disease milieus.
Conclusion
CPPs offer a versatile experimental platform to deliver otherwise non-permeant cargos into the CNS, enabling direct tests of intracellular hypotheses in living systems. Within this framework, targeting JNK signaling has emerged as a credible strategy to mitigate stress-driven apoptosis and secondary pathologies linked to Aβ, tau, and α-syn. Early studies with CPP–JNK inhibitor constructs suggest measurable protection across multiple models, particularly when inhibition spans relevant isoforms and is timed to pathophysiologic triggers. Nonetheless, JNKs serve essential physiological roles, and durable benefit will likely depend on refined selectivity, circuit-aware dosing, and combination strategies that address proteostasis, mitochondrial biology, and neuroimmune crosstalk in concert. Further research is needed.
References
- Keighron, N., Avazzadeh, S., Goljanek-Whysall, K., McDonagh, B., Howard, L., Ritter, T., & Quinlan, L. R. (2023). Extracellular vesicles, cell-penetrating peptides and miRNAs as future novel therapeutic interventions for Parkinson’s and Alzheimer’s disease. Biomedicines, 11(3), 728.
- Borsello, T., & Forloni, G. (2007). JNK signalling: a possible target to prevent Current pharmaceutical design, 13(18), 1875-1886.
- Abdelrahman, S., Hassan, H. A., Abdel-Aziz, S. A., Marzouk, A. A., Narumi, A., Konno, H., & Abdel-Aziz, M. (2021). JNK signaling as a target for anticancer therapy. Pharmacological Reports, 73, 405-434.
- Meloni, P., Craig, A. J., Milech, N.,Hopkins, R. M., Watt, P. M., & Knuckey, N.W. (2014). The neuroprotective efficacy of cell-penetrating peptides TAT, penetratin, Arg-9, and Pep-1 in glutamic acid, kainic acid, and in vitro ischemia injury models using primary cortical neuronal cultures. Cellular and molecular neurobiology, 34, 173-181.
- Graczyk, P. P. (2013). JNK inhibitors as anti-inflammatory and neuroprotective Future Medicinal Chemistry, 5(5), 539-551.
- Repici, M., & Borsello, T. (2007). JNK pathway as therapeutic target to prevent degeneration in the central nervous Hypoxia and Exercise, 145-155.
- Busquets, , Ettcheto, M., Cano, A., R. Manzine, P., Sanchez-Lopez, E., Espinosa-Jimenez, T., … & Camins, A. (2019). Role of c-Jun N-Terminal Kinases (JNKs) in epilepsy and metabolic cognitive impairment. International Journal of Molecular Sciences, 21(1), 255.
- Borsello, T., & Bonny, C. (2004). Use of cell- permeable peptides to prevent neuronal Trends in Molecular Medicine, 10(5), 239-244.
- Martinez Leo, E., Rojas Herrera, R. A., & Segura Campos, M. R. (2022). Biopeptides with Neuroprotective Effect in the Treatment of Neuroinflammation Induced by Adiposity-based Chronic Disease. Food Reviews International.
- Orejana, L., Barros-Miñones, L., Aguirre, N., & Puerta, E. (2013). Implication of JNK pathway on tau pathology and cognitive decline in a senescence-accelerated mouse model. Experimental gerontology, 48(6), 565-571.
- Bonny, C., Oberson, A., Negri, S., Sauser, C., & Schorderet, D. F. (2001). Cell- permeable peptide inhibitors of JNK: novel blockers of β-cell Diabetes, 50(1), 77-82.
Disclaimer: The information provided is intended solely for educational and scientific discussion. The compounds described are strictly intended for laboratory research and in-vitro studies only. They are not approved for human or animal consumption, medical use, or diagnostic purposes. Handling is prohibited unless performed by licensed researchers and qualified professionals in controlled laboratory environments.



