Fact-checked by Grok 2 weeks ago

Neuroregeneration

Neuroregeneration refers to the process of repairing, replacing, or regenerating damaged neural tissues, including neurons, , sheaths, and synaptic connections, to restore sensory, motor, emotional, and cognitive functions after injury or disease. This phenomenon encompasses mechanisms such as (the birth of new neurons), axonal regrowth, (formation of new synapses), and remyelination (reformation of insulation around axons), which collectively enable structural and functional recovery in the . While neuroregeneration occurs naturally to varying degrees across and nervous system compartments, it is particularly limited in the adult mammalian (CNS), contrasting with the more robust repair capacity of the peripheral nervous system (PNS). In the peripheral nervous system (PNS), neuroregeneration is facilitated by supportive glial cells known as Schwann cells, which clear axonal debris through , secrete like (NGF), and form bands of Büngner to guide regenerating axons at a rate of approximately 1 mm per day. Successful PNS repair often leads to functional recovery, as seen in injuries to nerves like the or ulnar, though outcomes depend on injury severity, with gaps exceeding 2 cm posing significant challenges due to donor site morbidity in autografts and incomplete reinnervation. Pharmacological agents such as and , along with stem cell therapies using mesenchymal stem cells, have shown promise in enhancing PNS regeneration by promoting axonal growth and reducing inflammation. Conversely, regeneration is inherently restricted by intrinsic neuronal factors (e.g., downregulated growth programs) and extrinsic barriers, including glial scars formed by reactive that deposit inhibitory proteoglycans (CSPGs), as well as myelin-associated inhibitors like Nogo-A. These obstacles result in minimal spontaneous repair after conditions such as , , or neurodegenerative diseases like Alzheimer's and Parkinson's, where progressive loss exacerbates functional deficits. is confined to niche regions like the and , relying on signaling pathways such as and AKT, but overall CNS repair remains slow due to poor waste clearance and long lifespans. Recent advances in neuroregeneration research focus on overcoming these limitations through multifaceted strategies, including transplantation (e.g., neural and mesenchymal stem cells) to replace lost cells and modulate immune responses, biomaterials like hydrogels and scaffolds for bridging injury gaps, and techniques such as epidural stimulation to enhance motor recovery. editing tools like , combined with (e.g., , BDNF), and pharmacological interventions (e.g., chondroitinase ABC to degrade CSPGs) have demonstrated preclinical efficacy in promoting axonal regeneration and functional restoration, with ongoing clinical trials evaluating safety and outcomes in humans as of 2025. In 2025, notable progress includes FDA IND clearances in June for (iPSC)-based therapies targeting , , and (ALS), as well as a phase 1/2a trial in November demonstrating potential in restoring vision for age-related (AMD). Despite these progresses, challenges persist, including ethical concerns with embryonic s, tumorigenicity risks, and the need for personalized approaches to achieve translational success.

Fundamentals of Neuroregeneration

Definition and Biological Significance

Neuroregeneration refers to the regrowth or repair of damaged nervous tissues, cells, or cell products, encompassing the of new neurons, axons, synapses, and glial cells to restore neural structure and function after or . This process contrasts sharply with neurodegeneration, which involves the progressive structural or functional loss of neurons, often leading to irreversible decline in nervous system integrity. The biological significance of neuroregeneration lies in its potential to reinstate sensory, motor, and cognitive capabilities compromised by neural damage, thereby mitigating long-term disability and improving . For example, in peripheral nerve injuries such as transections from , neuroregeneration facilitates axonal regrowth and functional recovery, allowing restoration of sensation and movement in affected limbs. In contrast, central nervous system injuries like spinal cord typically result in limited regeneration, causing enduring sensory and motor deficits due to intrinsic repair constraints. From an evolutionary perspective, mammals display heterogeneous regenerative abilities, with robust repair in nervous system but minimal in the central, differing markedly from non-mammalian vertebrates like and salamanders that exhibit extensive neuroregenerative capacities. , for instance, generate new neurons across nearly all regions throughout adulthood, enabling full recovery from injuries such as damage or lesions. Similarly, salamanders can regenerate neuronal populations and repair nerve fibers in the , , and , restoring tissue architecture and function post-injury. These differences highlight evolutionary trade-offs, where mammalian neural may prioritize stability over regeneration to support advanced cognitive processes. Key historical milestones in understanding neuroregeneration trace back to the early , when Santiago Ramón y Cajal's microscopic observations revealed the constrained regenerative potential of adult mammalian neurons, famously declaring that "in adult centers the nerve paths are something fixed, ended, immutable; everything may die, nothing may be regenerated." This insight, derived from studies of neural histology and injury responses, established foundational principles for modern and spurred ongoing efforts to enhance regenerative therapies.

Core Cellular and Molecular Mechanisms

Neuroregeneration involves a coordinated interplay among key cellular players, including neurons, glial cells such as astrocytes, oligodendrocytes, and Schwann cells, and immune cells like macrophages, which collectively mediate degeneration and subsequent regrowth processes. Neurons initiate intrinsic growth programs following injury, while glia provide structural support and modulate the microenvironment; for instance, astrocytes in the central nervous system (CNS) can promote repair through secretion of supportive factors but also contribute to inhibitory scarring. Immune cells, particularly macrophages, clear debris during degeneration and release cytokines that influence axonal sprouting and remyelination. At the molecular level, growth-associated proteins like GAP-43 play a pivotal role in axonal regeneration by regulating cytoskeletal dynamics and motility, enabling the extension of neurites post-injury. , including (NGF) and (BDNF), bind to receptors (TrkA and TrkB, respectively), activating downstream signaling cascades such as PI3K/Akt and MAPK/ERK pathways that enhance neuronal survival, axon elongation, and . These factors are upregulated in response to injury, creating a permissive environment for regrowth, though their efficacy diminishes in the adult mammalian CNS due to inhibitory cues. The process unfolds in distinct stages: initial degeneration of the distal axon segment triggers retrograde signaling to the cell body, promoting survival and growth preparation; this is followed by axonal extension guided by dynamic growth cones that sense extracellular cues via integrins and Rho GTPases for pathfinding. Synapse reformation then occurs through activity-dependent mechanisms, involving presynaptic vesicle release and postsynaptic receptor clustering, often facilitated by neurotrophins. Elevating intracellular cyclic AMP (cAMP) levels, for example via pharmacological agents, enhances intrinsic growth capacity by overriding inhibitory signals and promoting transcription of regeneration-associated genes like SPRR1a. Genetic and epigenetic factors further regulate regenerative potential; in mammals, inhibition of the PTEN/mTOR pathway has been shown to boost axonal regrowth by increasing activity, which drives protein synthesis essential for growth cones, as demonstrated in optic nerve injury models where PTEN deletion extended axons over long distances. Epigenetic modifications, such as histone acetylation via HDAC inhibitors, also reprogram gene expression to mimic developmental growth states, enhancing neuronal plasticity. Schwann cells in the peripheral nervous system briefly dedifferentiate to support these mechanisms by proliferating and forming bands of Büngner.

Regeneration in the Peripheral Nervous System

Axonal Regrowth and Wallerian Degeneration

Following peripheral in the peripheral nervous system (PNS), the distal segment of the severed undergoes , an orchestrated process of breakdown that clears cellular debris to prepare the pathway for regeneration. This degeneration begins with the loss of axonal integrity due to calcium influx and activation of proteases like calpains, leading to cytoskeletal fragmentation within 24-48 hours in rodents. , the myelinating of the PNS, dedifferentiate rapidly, proliferate, and initiate of and axonal fragments, peaking around 4 days post-injury. Resident and recruited macrophages join this effort, with hematogenous macrophages peaking at 3-7 days to efficiently remove the majority of debris through lysosomal degradation, creating a permissive environment for axonal regrowth. This debris clearance is essential, as persistent fragments would otherwise inhibit regeneration by releasing inhibitory molecules. The axonal regrowth process in the PNS commences shortly after degeneration, with the proximal axon stump sealing and forming a within hours to days. This dynamic structure, driven by polymerization and extension, advances at a rate of approximately 1-3 mm per day in mammals, allowing directed elongation toward target tissues. Guidance occurs via bands of Büngner, aligned columns of Schwann cells that form within the cleared endoneurial tubes and provide topographic cues, components like , and such as (NGF) to support outgrowth. This Schwann cell-derived support enhances neuronal survival and growth cone navigation. Several factors influence the success of axonal regrowth in the PNS. The proximity of the injury site to the neuronal cell body plays a critical role; injuries closer to the cell body (proximal lesions) result in longer regeneration distances, increasing the of target muscle and poorer functional outcomes compared to distal injuries. Larger diameters, often associated with motor fibers, correlate with faster regeneration rates and higher reinnervation success due to greater intrinsic growth capacity and metabolic support. Age-related declines further impair regeneration, with slower advance and reduced observed in older individuals, leading to incomplete recovery. Experimental studies in demonstrate the potential for near-complete functional recovery in short-distance PNS injuries. For instance, in models of sciatic nerve transection with minimal end-to-end gaps and immediate repair, axons regrow effectively, restoring walking patterns and muscle function within weeks, with grasping ability recovered by 7-12 weeks depending on repair method. These findings highlight the PNS's robust regenerative capacity when injury gaps are small, achieving up to 80-90% target reinnervation in proximal stumps.

Supportive Role of Schwann Cells

Schwann cells, the primary glial cells of the peripheral nervous system, play a pivotal role in supporting axonal regeneration following . Upon peripheral nerve damage, which initiates , Schwann cells in the distal stump rapidly respond by proliferating severalfold to repopulate the denervated region, although this proliferation is not strictly essential for effective regeneration. Concurrently, both myelinating and non-myelinating Schwann cells dedifferentiate into a specialized repair , characterized by the upregulation of immature markers such as L1 and NCAM and the downregulation of -associated genes like Egr2 and P0. This dedifferentiation enables them to clear and axonal debris through , a process that begins within 5–7 days post-injury and is often assisted by infiltrating macrophages, thereby creating a permissive environment for regrowth. In their repair state, Schwann cells actively secrete a variety of components and trophic factors to guide and nurture regenerating . They produce and , which facilitate axon adhesion and extension along the endoneurial tubes. Additionally, Schwann cells upregulate the expression of , including glial cell line-derived neurotrophic factor (GDNF), (BDNF), and (NGF), which promote neuronal survival, axonal outgrowth, and target muscle reinnervation. These secreted factors collectively enhance the intrinsic growth capacity of during the regenerative process. Schwann cells further contribute by forming structured regenerative niches that direct axonal navigation. Within the preserved , they align longitudinally to create bands of Büngner—elongated columns of dedifferentiated cells that serve as physical guides for sprouting axons toward their distal targets. Once axons reach appropriate end-organs, Schwann cells redifferentiate, sorting axons into bundles and initiating remyelination to restore conduction velocity and functional integrity. However, the supportive efficacy of s can diminish in pathological conditions, such as chronic nerve injuries or . Prolonged in chronic injuries leads to a progressive loss of the repair , with s failing to maintain proliferative and trophic capabilities, resulting in poorer axonal regrowth. In , induces dysfunction through , mitochondrial impairment, and reduced signaling via pathways like Akt/, which inhibits proliferation, impairs dedifferentiation, and decreases secretion of such as BDNF and NGF. These alterations contribute to delayed regeneration and persistent neuropathy.

Regeneration in the Central Nervous System

Intrinsic Limitations to Neuronal Repair

Adult neurons in the central nervous system (CNS) exhibit limited regenerative capacity compared to those in the peripheral nervous system (PNS), primarily due to intrinsic cellular constraints that hinder axonal regrowth after injury. Unlike PNS neurons, which robustly reactivate growth programs following axotomy, CNS neurons fail to mount a sufficient intrinsic response, resulting in stalled regeneration and persistent functional deficits. This disparity arises from a combination of downregulated genetic programs, maturational changes, and metabolic limitations that prioritize neuronal stability over repair in the mature CNS. A key intrinsic limitation is the downregulation of regeneration-associated genes (RAGs) in adult CNS neurons, which diminishes their ability to initiate and sustain axonal outgrowth. In the PNS, injury triggers the expression of numerous RAGs, such as small proline-rich repeat protein 1A (SPRR1A), which is upregulated over 60-fold in axotomized sensory neurons to promote extension; however, this and other RAGs like GAP-43 and ATF3 show markedly reduced or absent induction in CNS neurons post-. This subdued RAG response contributes to the failure of CNS axons to regrow effectively, as these genes encode proteins essential for cytoskeletal remodeling and advancement. Studies in models confirm that artificially elevating RAG expression can partially restore regenerative potential, underscoring the neuron-intrinsic nature of this barrier. Maturation and aging further exacerbate these limitations through a developmental switch that represses regenerative programs to maintain circuit stability. During early development, CNS neurons express high levels of growth-promoting factors, but as maturity sets in, like RE1-silencing (REST), also known as neuron-restrictive silencer factor (NRSF), suppress these programs by binding to regulatory elements in promoters. REST acts as a master repressor, silencing genes critical for growth in adult neurons and preventing the reactivation of embryonic-like regenerative states after injury. This switch is evolutionarily conserved and linked to aging, where cumulative epigenetic modifications reinforce REST-mediated inhibition, making older CNS neurons even less responsive to damage. Network analyses in injured CNS models have identified REST as a central in this repressive circuitry, highlighting its role in blocking intrinsic repair pathways. Metabolic barriers also impose significant intrinsic constraints, as CNS axons face higher demands for long-distance regrowth than their PNS counterparts. Mature CNS axons, often myelinated and spanning greater distances, require substantial ATP for mitochondrial and biosynthetic processes during regeneration, but intrinsic deficits in production—such as impaired and —lead to an crisis post-injury. In contrast, PNS axons benefit from more efficient local supply via supportive , enabling sustained regrowth. Experimental interventions that boost neuronal , like enhancing mitochondrial function, have shown modest improvements in CNS axon extension, indicating that these metabolic hurdles are manipulable yet fundamentally limit unaided repair. Evidence from genetic models demonstrates that these intrinsic factors can be overcome to promote partial CNS regeneration. In optic nerve crush experiments using conditional knockout mice, deletion of the phosphatase and tensin homolog (PTEN) gene activates the mTOR signaling pathway, leading to robust regrowth of retinal ganglion cell axons up to several millimeters beyond the injury site—far exceeding spontaneous recovery. When combined with other manipulations like SOCS3 deletion, PTEN loss sustains long-term axon elongation, reaching contralateral visual targets and partially restoring visual function. These findings illustrate that intrinsic growth programs remain latent in adult CNS neurons and can be unlocked by targeting key regulators, offering insights into potential therapeutic avenues without addressing extrinsic inhibitors.

Glial Scar Formation and Its Effects

Following (CNS) injury, such as trauma, reactive astrocytes undergo hypertrophy and proliferation, leading to the formation of a that serves as a structural response to damage. This scar is primarily composed of reactive astrocytes expressing elevated levels of intermediate filament proteins, including (GFAP) and , which provide cytoskeletal support and enable cellular elongation. Additionally, the scar incorporates an (ECM) rich in inhibitory molecules, such as chondroitin sulfate proteoglycans (CSPGs; e.g., neurocan, brevican, phosphacan, and versican), hyaluronan, and tenascin-C, secreted by the astrocytes and infiltrating fibroblasts. The process of glial scar formation unfolds rapidly post-injury. Within hours, are activated by damage-associated signals like ATP and proinflammatory cytokines (e.g., TNF-α, IL-6), initiating and upregulation of GFAP. and of these cells begin 1–2 days after , peaking at 3–5 days, as extend processes to align parallel to the injury axis and form a dense by 7–10 days. Over 1–2 weeks to several months, the matures into a compact barrier, physically compartmentalizing the core and chemically enriching the with growth-inhibitory components. The exerts dual effects on CNS repair. Beneficially, it isolates the injury site, limiting the spread of , inflammatory cells, and fibrotic tissue to protect adjacent healthy from secondary damage. However, detrimentally, the acts as a physical and chemical barrier, impeding axonal regrowth by blocking penetration through its dense astrocytic processes and , where CSPGs bind and inhibit advancement via interactions with receptors like PTPσ and LAR. Studies modulating the have highlighted its inhibitory dominance. Enzymatic degradation of CSPGs using chondroitinase ABC (ChABC) partially reduces the scar's chemical barriers without fully ablating the astrocytic structure, promoting axonal sprouting, regeneration across the lesion, and improved functional recovery in models; for instance, intrathecal ChABC administration post-injury upregulated regeneration-associated genes like GAP-43 and enhanced locomotor outcomes.

Barriers to Neuroregeneration

Extracellular Matrix Inhibitors

In the (CNS), the (ECM) undergoes significant remodeling following , leading to the deposition of inhibitory molecules that hinder axonal regeneration. These non-myelin-based ECM components, primarily proteoglycans and glycoproteins, create a repressive environment by altering substrate adhesion, activating intracellular signaling cascades that collapse growth cones, and increasing tissue stiffness around the lesion site. Unlike myelin-associated inhibitors, which signal through specific neuronal receptors to limit outgrowth, ECM inhibitors exert broader effects on cellular migration and process extension in the post-injury scar. Chondroitin sulfate proteoglycans (CSPGs), such as aggrecan and neurocan, are among the most prominent ECM inhibitors upregulated in the after CNS trauma. These molecules bind to neuronal receptors like protein tyrosine phosphatase sigma (PTPσ), triggering the activation of the RhoA/Rho-associated kinase () pathway, which leads to actin destabilization and collapse, thereby preventing axonal extension. This inhibitory signaling is particularly potent in the mature CNS, where CSPG expression correlates with reduced plasticity and regeneration failure following (SCI). Keratan sulfate proteoglycans (KSPGs) contribute to the inhibitory milieu by accumulating in the , where they enhance matrix stiffness and disrupt neuronal adhesion and migration. Expressed by reactive , , and oligodendrocyte progenitors post-injury, KSPGs form a dense barrier that physically impedes axonal penetration and promotes a non-permissive environment for repair. Their role in scar formation has been linked to suppressed regeneration, with depletion strategies showing potential to alleviate these effects. Additional ECM factors, including tenascin-C and , further establish inhibitory gradients in the injured CNS by modulating cell-matrix interactions and . Tenascin-C, upregulated after , promotes neuroinflammatory cascades that exacerbate secondary damage and limit axonal , while , often in complex with proteoglycans, contributes to compartmentalization and reduced tissue permeability for regenerating fibers. Therapeutic approaches targeting these inhibitors, such as enzymatic degradation of CSPGs with chondroitinase ABC (ChABC), have demonstrated significant promotion of axonal regrowth and functional recovery in animal models, including , by digesting chains and attenuating downstream inhibitory signals. Sustained ChABC delivery enhances locomotor outcomes and , highlighting its translational potential.

Myelin-Associated Growth Inhibitors

Myelin-associated growth inhibitors are proteins embedded in or associated with the sheaths of the (CNS) that actively suppress axonal regeneration following injury, contributing to the poor reparative capacity of the CNS compared to the peripheral nervous system. These molecules, primarily produced by , form a collective "myelin brake" that halts advance and neurite extension, thereby limiting functional recovery after conditions like . Key examples include Nogo-A, -associated (MAG), and oligodendrocyte (OMgp), which signal through shared neuronal receptors to activate intracellular pathways that destabilize the . Nogo-A, a membrane-bound protein highly expressed in , is one of the most potent myelin-derived inhibitors of axonal growth. It exerts its effects primarily through two domains: the Nogo-66 , which binds to the Nogo-66 receptor 1 (NgR1) on neurons, and a Nogo-A-specific 20-amino-acid that signals independently via . Upon binding to NgR1, Nogo-A forms a receptor complex with LINGO-1 and co-receptors such as p75NTR or , leading to activation of the RhoA/ROCK pathway; this cascade promotes , growth cone collapse, and inhibition of neurite outgrowth. Genetic ablation of Nogo-A in mice enhances compensatory sprouting in the but yields limited improvements in long-distance axonal regeneration, underscoring its role in modulating plasticity rather than outright repair. Myelin-associated glycoprotein (MAG), a transmembrane sialic acid-binding protein present in both CNS and myelin, inhibits axonal regeneration by interacting with specific s on the neuronal surface. MAG binds to gangliosides GD1a and GT1b, triggering inhibitory signaling through receptors including NgR1, NgR2, and , which activate downstream cascades that collapse growth cones and suppress neurite extension in mature neurons. studies demonstrate that MAG potently inhibits outgrowth from cerebellar and adult neurons, with its effects reversed by anti-MAG antibodies, suggesting a direct role in the failure of CNS regeneration. Notably, MAG's inhibitory action is age-dependent, promoting outgrowth in neonatal neurons while suppressing it in adults, which may reflect developmental shifts in ganglioside expression. Oligodendrocyte myelin glycoprotein (OMgp), a glycosylphosphatidylinositol (GPI)-anchored protein enriched in CNS myelin, contributes to the inhibitory milieu by binding to NgR1 and PirB receptors on neurons. This interaction induces growth cone collapse and potently inhibits neurite outgrowth in cultured neurons, accounting for a significant portion of the overall inhibitory activity of CNS myelin extracts. Like Nogo-A and MAG, OMgp signals through the NgR1 complex to activate RhoA-mediated pathways, reinforcing the collective suppression of axonal regeneration post-injury. Deletion of OMgp in animal models promotes serotonergic sprouting but does not substantially enhance long-tract regeneration, indicating its primary influence on local plasticity. The clinical relevance of these inhibitors is highlighted by therapeutic efforts targeting Nogo-A, such as neutralizing antibodies like NG101, which have been tested in phase 2b trials for acute cervical . In a randomized, double-blind of 129 patients, intrathecal NG101 was safe and well-tolerated, with no significant overall improvement in upper extremity motor scores (delta 1.37 points), but post-hoc analyses revealed modest functional gains in motor-incomplete subgroups, including enhanced voluntary activation and . A September 2025 re-analysis of the trial data using electrophysiological stratification further indicated greater functional recovery in patients with preserved somatosensory evoked potentials (SSEPs) treated with NG101 compared to . These results suggest potential for anti-Nogo therapies in select patients, though broader efficacy remains under investigation.

Therapeutic Strategies for Neuroregeneration

Cellular Replacement Approaches

Cellular replacement approaches in neuroregeneration aim to restore lost neuronal or glial populations by generating new cells through endogenous or transplanting exogenous cell-derived neurons. These strategies address the limited intrinsic regenerative capacity of the (CNS) by leveraging cells or direct fate conversion to produce functional replacements that can integrate into neural circuits. Key methods include glial-to-neuron , grafting, and direct neuronal , each offering distinct advantages in avoiding the tumorigenic risks associated with pluripotent intermediates. In vivo glial-to-neuron reprogramming involves converting resident or other glial cells into neurons using viral delivery of transcription factors, such as NeuroD1, directly within the injured brain. Studies in models have demonstrated that NeuroD1 overexpression in leads to their transition into neuron-like cells, though these exhibit limited functional maturity, with induced cells showing neuronal and marker expression in the following injury. However, conversion efficiency remains limited in certain regions like the , with only partial astrocyte-to-neuron shifts observed, highlighting challenges in achieving widespread . Neural stem cell grafting utilizes sources such as induced pluripotent cells (iPSCs) or fetal neural to transplant cells that differentiate into neurons post-implantation. iPSC-derived neural cells (NSCs) have shown promise in preclinical models, but engraftment rates are typically low, with fewer than 5% of transplanted cells surviving long-term due to hostile CNS environments including and ischemia. Enhancing vascularization through co-transplantation with endothelial cells or organoid formats improves survival and differentiation, as demonstrated in iPSC-derived cerebral s grafted into brains, where vascular integration boosted neural maturation. Fetal grafts, while effective in historical Parkinson's trials, face ethical and scalability issues, prompting a shift toward iPSC sources for autologous therapies. As of 2025, Phase I/II clinical trials of iPSC-derived cells for are underway, showing promise in safety and efficacy. Direct neuronal reprograms non-neuronal cells, such as fibroblasts, into neurons using small molecules, vectors, or transcription factors like Ascl1 and Brn2, bypassing pluripotency to minimize risks. This approach avoids the tumorigenicity associated with iPSC intermediates, as it does not involve proliferative progenitor states, reducing mutation accumulation and tumor formation potential. Small molecule cocktails have successfully induced and , yielding neurons with electrophysiological properties suitable for CNS repair, and methods enable targeted conversion . Compared to iPSC-based methods, direct preserves age-related epigenetic signatures, potentially enhancing therapeutic relevance for age-associated neurodegeneration. Functional outcomes from these approaches have been particularly notable in models, where grafted dopaminergic neurons form synapses and restore circuit activity. Human iPSC-derived dopaminergic neurons transplanted into mouse exhibit pre- and postsynaptic integration, rescuing motor deficits through release and behavioral normalization. reprogramming in glial cells has similarly produced neurons that form functional connections, contributing to circuit repair without graft overgrowth. These results underscore the potential for cellular replacement to achieve therapeutic efficacy, though scalability and immune compatibility remain hurdles for clinical translation.

Pharmacological and Biomaterial Interventions

Pharmacological interventions in neuroregeneration target intracellular signaling pathways to enhance the intrinsic growth capacity of neurons, overcoming inhibitory environments in the central and peripheral nervous systems. RhoA/ROCK signaling, activated by myelin-associated inhibitors and cues, limits axonal outgrowth by promoting actomyosin contractility and collapse. Inhibition of this pathway using C3 transferase, a specific RhoA inactivator, has been shown to promote significant axonal regeneration in rodent models of , with treated animals exhibiting enhanced regrowth compared to controls. Similarly, pharmacological ROCK inhibitors like Y-27632 enhance neurite extension in cultured neurons exposed to inhibitory substrates, demonstrating dose-dependent improvements in dynamics. PTEN, a that negatively regulates the PI3K/AKT/ pathway, suppresses intrinsic growth programs in adult neurons; conditional deletion or inhibition of PTEN via small molecules such as bpV(HOpic) boosts signaling, leading to robust axon regeneration in crush models, where regrowing axons extend over 500 μm beyond the lesion site in mice. These approaches highlight the potential of targeting conserved signaling hubs to reinstate developmental-like growth states in mature neurons. Delivery of neurotrophic factors represents another key pharmacological strategy, providing sustained trophic support to promote neuronal survival and axonal elongation while mitigating rapid degradation of these proteins. (BDNF), which activates TrkB receptors to stimulate PI3K/AKT and MAPK/ERK pathways, has been effectively delivered via nanoparticles for prolonged release in injury models. Chitosan-based nanoparticles encapsulating BDNF enable slow release over weeks, resulting in enhanced axonal growth in neurons, with treated cultures showing 2-3 fold increases in neurite length compared to free BDNF applications. In peripheral models, such as sciatic nerve transection, nanoparticle-mediated BDNF delivery improves functional recovery, correlating with increased myelination and reduced . These systems address the short of , ensuring localized and temporally controlled signaling to support regeneration without systemic side effects. Biomaterial interventions provide structural guidance and biochemical cues to bridge injury gaps, mimicking the extracellular matrix (ECM) to facilitate axonal navigation and reduce scar formation. In the peripheral nervous system (PNS), collagen-based nerve conduits serve as FDA-approved alternatives to autografts for gaps up to 5 cm, promoting Schwann cell migration and vascularization to support axon regrowth. For instance, type I collagen tubes filled with permissive gels have demonstrated superior regeneration in canine sciatic nerve defects, with histological analysis revealing aligned axonal bundles and near-normal conduction velocities at 12 months post-implantation. Hydrogels, such as hyaluronic acid or fibrin-based matrices, offer tunable injectability and degradation profiles for PNS bridging, enhancing sprout formation by 40-60% in rodent models through incorporation of laminin or fibronectin motifs. In the central nervous system (CNS), where regeneration is more restricted, ECM-mimicking scaffolds like agarose or alginate hydrogels loaded with chondroitinase ABC degrade inhibitory glycosaminoglycans, allowing modest corticospinal tract sprouting in spinal cord injury paradigms. These biomaterials not only physically guide axons but also modulate the inflammatory milieu, fostering a pro-regenerative niche. Combinatorial therapies integrate pharmacological and biomaterial approaches to synergistically address multiple barriers, yielding greater regenerative outcomes than single modalities. For example, combining anti-Nogo-A antibodies, which neutralize myelin-associated growth inhibitors by blocking NgR1 receptor activation, with BDNF administration promotes extensive optic nerve regeneration in adult mice, where dual treatment results in axons projecting over 1 mm past the crush site, far exceeding either therapy alone. In Phase II clinical trials for acute optic neuritis, such as the RENEW study evaluating opicinumab (an anti-LINGO-1 antibody targeting a related inhibitory pathway), adjunctive use with corticosteroids showed trends toward improved visual evoked potential recovery, underscoring the potential of inhibitor neutralization paired with supportive factors. These strategies leverage orthogonal mechanisms—disinhibition plus growth promotion—to amplify intrinsic repair capacity, as evidenced by enhanced functional connectivity in preclinical imaging studies.

Clinical Progress and Challenges

Approved Treatments and Outcomes

Approved treatments for neuroregeneration primarily target (PNS) injuries through surgical interventions, while (CNS) options remain limited to neuroprotective agents that modestly slow disease progression rather than fully restoring . In the PNS, autografts serve as the gold standard for repairing significant gaps in traumatic injuries, involving the harvest and implantation of a patient's own segment to bridge the defect and facilitate axonal regrowth. Success rates for sensory recovery with autografts typically range from 70% to 90% in suitable cases, such as digital repairs, where patients achieve meaningful protective sensation or better. For compressive neuropathies like , surgical release of the transverse carpal ligament yields high efficacy, with over 90% of patients experiencing substantial symptom relief and sensory improvement within months post-procedure. In the CNS, approved interventions focus on neuroprotection to mitigate further degeneration, as true regenerative therapies are not yet available. , approved by the FDA in 1995 and remaining a cornerstone treatment for (ALS), inhibits glutamate and extends survival by 2 to 3 months on average while slowing functional decline. , approved in 2017, acts as a free radical scavenger to reduce in ALS, demonstrating a 33% slower decline in physical function over 24 weeks in early-stage patients. For acute (SCI), high-dose has been used since the 1990s to reduce secondary and , with the NASCIS-2 trial showing modest improvements in motor function at 6 weeks and 1 year when administered within 8 hours of injury. However, current guidelines as of 2025, including those from the American Association of Neurological Surgeons, do not recommend its routine use due to risks of complications like infection and outweighing benefits in many cases. Outcomes for these treatments highlight the disparity between PNS and CNS repair capacities, often measured using standardized scales. In PNS repairs, such as autografts or decompressions, 70% to 82% of patients achieve meaningful motor or sensory recovery, defined as Medical Research Council (MRC) grades M3 or higher for motor function and S3 or better for sensation, enabling return to daily activities. For carpal tunnel release, functional improvement occurs in 80% to 90% of cases, with most patients regaining near-normal grip strength and sensation by 6 months. In contrast, CNS outcomes are more limited; for ALS, riluzole and edaravone together yield partial stabilization in 30% to 50% of patients, extending time to ventilator dependence but rarely reversing deficits. For acute SCI, the American Spinal Injury Association (ASIA) Impairment Scale shows conversion to a better grade (e.g., from complete to incomplete injury) in approximately 27% of cases with methylprednisolone or supportive care, with partial motor recovery below the injury level in fewer than 10% of complete injuries, underscoring persistent barriers to regeneration. Post-2020 advancements include expanded approvals for , such as (Qalsody) in 2023 for SOD1-mutated cases, which reduces light chain levels as a of neuronal damage by up to 60% at 6 months, indicating neuroprotective effects without full regeneration. For , therapies remain in clinical trials rather than fully approved, but phase II/III data demonstrate long-term safety over 2 years, with 50% to 70% of patients reporting reduced pain and improved nerve conduction velocities due to enhanced axonal repair and anti-inflammatory actions. These interventions collectively emphasize symptom and partial preservation over robust regeneration, with ongoing monitoring via scales like for SCI revealing sustained but incomplete functional gains in responsive subgroups.

Emerging Research Directions

Recent advancements in have introduced light-activated ion channels, such as , to precisely modulate neuronal activity and promote regrowth following injury. These tools enable targeted stimulation of growth-promoting pathways, enhancing regeneration in preclinical models by activating intracellular signaling cascades like JAK2/STAT3. In studies from 2023 to 2025, optogenetic approaches have demonstrated the ability to induce remodeling and receptor-mediated dynamics in cultured neurons, facilitating directed extension. Translation from models, where optogenetics has illuminated mechanisms of robust repair, to mammals has shown promise, with transcranial stimulation promoting regeneration in rodent models and restoring functional connections in situ. However, challenges include limited penetration in deep structures and the need for delivery, which may limit clinical translation. Gene editing technologies, particularly CRISPR-Cas9, are being explored to silence inhibitory genes that hinder neuroregeneration, such as those encoding Nogo-A and myelin-associated glycoprotein (MAG), which restrict axonal sprouting in the adult mammalian . By targeting these genes, CRISPR-Cas9 disrupts inhibitory signaling, allowing for enhanced neurite outgrowth in preclinical settings. Recent 2023-2025 studies have applied to edit genes involved in regenerative barriers, including those linked to formation, resulting in improved functional recovery in rodent models of neurodegeneration and injury. Although primate-specific data remain limited, extensions from rodent experiments indicate enhanced axon extension when combining gene editing with supportive interventions, highlighting a pathway toward translational therapies. Key challenges involve off-target editing risks, efficient delivery across the blood-brain barrier, and long-term safety in humans. Human (iPSC)-derived neural organoids represent a transformative bioengineered platform for studying neuroregeneration, offering three-dimensional models that recapitulate architecture and cellular interactions absent in traditional animal systems. These organoids enable high-throughput testing of regenerative therapies by simulating responses and evaluating axon regrowth in a human-specific context, thereby addressing key species differences in regenerative capacity—such as the limited mammalian repair compared to . Developments from 2023 to 2025 have integrated vascularization and immune components into these models, improving their physiological relevance for assessing regeneration post- and identifying novel pro-regenerative factors. By bridging translational gaps, iPSC organoids facilitate the screening of interventions that promote synaptic reconnection and neuronal , accelerating the shift from bench to bedside. Nonetheless, variability between organoids, issues, and ethical concerns regarding iPSC sourcing persist as hurdles. The integration of (AI) and analytics is revolutionizing neuroregeneration research by leveraging algorithms to predict outcomes based on injury profiles, including lesion severity, inflammatory markers, and genetic factors. These predictive models analyze multimodal datasets from and to forecast regenerative potential, enabling personalized therapeutic strategies. In 2023-2025 studies, AI has demonstrated high accuracy in anticipating functional recovery after spinal cord injuries through routine bloodwork patterns, identifying biomarkers that correlate with axon regrowth and motor restoration. Furthermore, AI-driven platforms are expediting by simulating regenerative responses and prioritizing compounds that enhance , with systematic reviews underscoring their role in overcoming data scarcity in preclinical trials. Challenges include potential biases in training data, the need for larger diverse datasets, and interpretability of AI decisions to ensure clinical trust.

References

  1. [1]
    Neurodegenerative Diseases: Regenerative Mechanisms and Novel ...
    Neuroregeneration can also be defined as the progressive structural and functional recovery of the damaged nervous system over time. Damage to the central ...
  2. [2]
    Neuroregeneration and plasticity: a review of the physiological ...
    Neuroregeneration involves synthesizing new neurons and connections, providing extra resources in the long term to replace those damaged by the injury.
  3. [3]
    Peripheral Nerve Regeneration: Opportunities and Challenges - PMC
    Feb 8, 2023 · This article aims to provide a critical overview of current research on peripheral nerve regeneration and future perspectives.Pharmacotherapy · Stem Cell Therapy · Artificial Nerve Conduits
  4. [4]
    A Comprehensive Review of the Role of Stem Cells in ...
    Aug 22, 2024 · This review provides a comprehensive analysis of the role of stem cells in neuroregeneration, exploring different types of stem cells.
  5. [5]
    Advancements in neuroregenerative and neuroprotective therapies ...
    This comprehensive review aims to provide an up-to-date and thorough overview of the latest neuroregenerative and neuroprotective therapies currently under ...
  6. [6]
    Neuroregeneration - an overview | ScienceDirect Topics
    Neuroregeneration is the regrowth or repair of the nervous tissue by generating new neurons, axons, synapses, and glial cells.
  7. [7]
    Neuroregeneration in neurodegenerative disorders - BMC Neurology
    Jun 23, 2011 · Neuroregeneration is a relatively recent concept that includes neurogenesis, neuroplasticity, and neurorestoration - implantation of viable cells as a ...Missing: definition biological
  8. [8]
    A comparative view of regenerative neurogenesis in vertebrates - PMC
    Compared with mammals, teleost fish such as zebrafish and medaka show significantly greater rates of neurogenesis, as new neurons are generated in most brain ...
  9. [9]
    Homeostatic and regenerative neurogenesis in salamanders
    Salamanders can replace neuronal populations, repair damaged nerve fibers and restore tissue architecture in retina, brain and spinal cord, leading to ...
  10. [10]
    Cajal, the neuronal theory and the idea of brain plasticity - PMC
    This paper reviews the importance of Cajal's neuronal theory (the Neuron Doctrine) and the origin and importance of the idea of brain plasticity that emerges ...
  11. [11]
    Wallerian degeneration: gaining perspective on inflammatory events ...
    In this review, we first provide a brief historical perspective, discussing how peripheral nerve injury (PNI) may have caused World War I.
  12. [12]
    The Drama of Wallerian Degeneration: The Cast, Crew, and Script
    Nov 23, 2021 · In this review, we describe our current understanding of Wallerian degeneration, focusing on the molecular players and mechanisms that mediate ...
  13. [13]
    Axon Regeneration in the Peripheral and Central Nervous Systems
    Peripheral nervous system (PNS) axons readily regenerate, allowing recovery of function after peripheral nerve damage.
  14. [14]
    Regenerative Medicine: A New Horizon in Peripheral Nerve Injury ...
    Mar 31, 2025 · Local MSC injection increases axonal fiber counts, improves electromotor recovery, and enhances nerve regeneration. It also has immunological ...
  15. [15]
    Challenges and Opportunities for Regeneration in the Peripheral ...
    Successful peripheral nerve repair depends not only upon delivery of axons to the periphery, but also upon re-establishment of appropriate connections between ...
  16. [16]
    A novel nerve transection and repair method in mice - Nature
    Dec 10, 2020 · The better functional recovery following nerve transection with minimal gap suggests that close approximation of the severed nerve ends and ...
  17. [17]
    Assessment of Functional Recovery of Sciatic Nerve in Rats ...
    The objective of this study was to assess the functional recovery of sciatic nerve in rats subjected to different fluences of low-level laser therapy (LLLT).
  18. [18]
  19. [19]
  20. [20]
  21. [21]
    Schwann Cells: Development and Role in Nerve Repair - PMC
    Schwann cells develop from the neural crest in a well-defined sequence of events. This involves the formation of the Schwann cell precursor and immature ...
  22. [22]
  23. [23]
  24. [24]
  25. [25]
    What makes a RAG regeneration associated? - Frontiers
    The reduced neuron-intrinsic injury-induced RAG response is an important reason for the failure of CNS regeneration. An interesting point to consider is why ...
  26. [26]
    A Gene Network Perspective on Axonal Regeneration - PMC
    Nov 22, 2011 · Axotomy of peripheral neurons results in the activation of hundreds of neuron-intrinsic regeneration-associated genes (RAGs), whereas in many ...
  27. [27]
    Small Proline-Rich Repeat Protein 1A Is Expressed by Axotomized ...
    Feb 15, 2002 · The small proline-rich repeat protein 1A (SPRR1A) is not detectable in uninjured neurons but is induced by >60-fold after peripheral axonal damage.
  28. [28]
    Axonal Regeneration: Underlying Molecular Mechanisms and ...
    After nerve injury, ATF3 activates and targets RAGs, including cJUN, Hsp27, and Cap-23, which is functionally related to Gap-43, and Sprr1a to induce axonal ...
  29. [29]
    Transcription factor network analysis identifies REST/NRSF as an ...
    Jul 29, 2022 · These analyses identify a role for REST as an upstream suppressor of the intrinsic regenerative program in the CNS.
  30. [30]
    The Master Negative Regulator REST/NRSF Controls Adult ...
    Jun 29, 2011 · Our work identifies REST/NRSF as a master negative regulator of adult NSC differentiation and offers a potential molecular target for neuroregenerative ...
  31. [31]
    Restoring cellular energetics promotes axon regeneration and ...
    Axonal regeneration in the central nervous system (CNS) is a highly energy demanding process. Extrinsic insults and intrinsic restrictions lead to an energy ...
  32. [32]
    Nerve regeneration: Specific metabolic demands? - ScienceDirect
    During development, peripheral nerve axons grow a long way from the neuronal cell bodies to distant target organs like muscle or skin.
  33. [33]
    Long-Distance Axon Regeneration in the Mature Optic Nerve
    Nov 17, 2010 · The combination of PTEN deletion plus Zymosan (Zymo) plus CPT–cAMP resulted in far greater regeneration than any other treatments (D). E–H, ...
  34. [34]
    CNS Injury, Glial Scars, and Inflammation - PubMed Central - NIH
    The formation of a histologically apparent glial scar, composed of astrocytes and connective tissue elements, is one of the most studied but poorly understood ...
  35. [35]
    Dissecting the Dual Role of the Glial Scar and Scar-Forming ...
    As a major component of the SCI lesion, the glial scar is primarily composed of scar-forming astrocytes and plays a crucial role in spinal cord regeneration.
  36. [36]
    Chondroitinase ABC promotes functional recovery after spinal cord ...
    Apr 11, 2002 · We show that intrathecal treatment with ChABC degraded CS-GAG at the injury site, upregulated a regeneration-associated protein in injured neurons, and ...
  37. [37]
    Chondroitin sulfate proteoglycans prevent immune cell phenotypic ...
    May 25, 2022 · Chondroitin sulfate proteoglycans (CSPGs) act as potent inhibitors of axonal growth and neuroplasticity after spinal cord injury (SCI).
  38. [38]
    Advances in the Signaling Pathways Downstream of Glial-Scar Axon ...
    (A) Intracellularly, activation of PTPσ and LAR by CSPGs activate RhoA/ROCK signaling and inactivate Akt and Erk pathways. However, the two receptors use ...Missing: neuroregeneration | Show results with:neuroregeneration
  39. [39]
    Axon growth inhibition by RhoA/ROCK in the central nervous system
    Among these, CSPGs play important roles to inhibit axonal regeneration after injury. CSPGs include aggrecan, brevican, neurocan, NG2, phosphacan, and ...Rhoa/rock Signaling And Axon... · Myelin-Associated Inhibitors · Rhoa/rock Inhibition In...Missing: neuroregeneration | Show results with:neuroregeneration
  40. [40]
    Chondroitin Sulfate Proteoglycans in the Nervous System: Inhibitors ...
    The interaction of CSPGs and neurons activates the Rho-ROCK and/or protein kinase C (PKC) intracellular signaling cascades, which inhibit process extension. It ...2. Chondroitin Sulfate... · 4. Degenerative Lesions · 6. Cspg Deposition At Cns...Missing: neuroregeneration | Show results with:neuroregeneration
  41. [41]
    Brain keratan sulfate and glial scar formation - PubMed
    Glial scar is the physical barrier to axonal regeneration of injured neurons. ... It is known that keratan sulfate proteoglycans (KSPGs) are also augmented ...Missing: stiffness | Show results with:stiffness
  42. [42]
    [PDF] Spinal Cord Injury Elicits Expression of Keratan Sulfate ...
    Spinal Cord Injury Elicits Expression of Keratan Sulfate Proteoglycans by Macrophages, Reactive Microglia, and Oligodendrocyte Progenitors.Missing: stiffness | Show results with:stiffness<|separator|>
  43. [43]
    The Role of Tenascin-C in Tissue Injury and Repair After Stroke
    A matricellular protein tenascin-C (TNC) is considered to be an important inducer to promote neuroinflammatory cascades and the resultant pathology in stroke.
  44. [44]
    Hyaluronan hydrates and compartmentalises the CNS/PNS ...
    Jul 26, 2023 · Administered ITI ameliorates brain injury, prevents cell death and improves behavioural outcomes after neonatal hypoxic ischaemia in rats (Chen ...Missing: post- | Show results with:post-
  45. [45]
    Recent advances in the therapeutic uses of chondroitinase ABC
    This review focuses on recent advances in therapies using ChABC in animal models of SCI and peripheral nerve damage.
  46. [46]
    Chondroitinase ABC Promotes Axon Regeneration and Reduces ...
    Mar 24, 2021 · Many studies have shown that digestion of CSPGs with chondroitinase ABC (ChABC) can enhance axon growth and functional recovery after SCI.Abstract · Introduction · Results · Discussion
  47. [47]
    A Systematic Review and Meta-analysis of Chondroitinase ABC
    Jan 7, 2025 · This systematic review aims to investigate the efficiency of ChABC treatment in animal models of SCI for locomotor recovery. Materials and ...
  48. [48]
    Myelin-Associated Inhibitors in Axonal Growth After CNS Injury - PMC
    Mar 6, 2014 · Nogo, MAG and OMgp are three prototypical myelin inhibitors that signal through multiple neuronal receptors to exert growth inhibition.Missing: seminal | Show results with:seminal
  49. [49]
    A novel role for myelin-associated glycoprotein as an inhibitor of ...
    We demonstrate that the myelin-associated glycoprotein (MAG), a transmembrane protein of both CNS and PNS myelin, strongly inhibits neurite outgrowth.
  50. [50]
  51. [51]
  52. [52]
    Direct Neuronal Reprogramming: Bridging the Gap Between Basic ...
    Direct neuronal reprogramming is an innovative new technology that involves the conversion of somatic cells to induced neurons (iNs) without passing through ...
  53. [53]
    Neurons derived from NeuroD1-expressing astrocytes transition ...
    Jul 25, 2025 · Abstract. In vivo conversion of nonneuronal cells into neurons is a proposed strategy to replace neurons lost to CNS injury or disease.
  54. [54]
    Revisiting astrocyte to neuron conversion with lineage tracing in vivo
    Oct 14, 2021 · A recent study showed that co-expression of NEUROD1 and DLX2 can efficiently convert astrocytes into local neurons in the adult mouse striatum ( ...
  55. [55]
    Translational considerations in injectable cell-based therapeutics for ...
    Aug 10, 2017 · However, existing cell-delivery approaches have shown limited success, with numerous studies showing fewer than 5% of injected cells persisting ...
  56. [56]
    Vascularization and Engraftment of Transplanted Human Cerebral ...
    Nov 12, 2018 · We hypothesized that transplantation of cerebral organoids derived from human ESC (hESC)/iPSC may enhance graft survival, neural differentiation ...
  57. [57]
    Engraftment and morphological development of vascularized human ...
    Oct 20, 2017 · Our vascularized iPSC-CM 3D-tissues may be a useful regenerative paradigm which will require additional expanded and long-term studies.
  58. [58]
    Direct cell reprogramming for tissue engineering and regenerative ...
    Feb 13, 2019 · Direct cell reprogramming, also called transdifferentiation, allows for the reprogramming of one somatic cell type directly into another.Missing: tumorigenicity | Show results with:tumorigenicity<|separator|>
  59. [59]
    Application of Small Molecules in the Central Nervous System Direct ...
    The use of small molecules to directly reprogram somatic cells into neurons provides a new therapeutic strategy for neurological diseases.Missing: avoiding | Show results with:avoiding
  60. [60]
    Transdifferentiation: a new promise for neurodegenerative diseases
    Aug 6, 2018 · Transdifferentiation can be achieved by cell-permeable small molecules without the need for viral transduction. Induced neural progenitor cells ...Missing: tumorigenicity | Show results with:tumorigenicity
  61. [61]
    Dopaminergic Progenitors Derived From Epiblast Stem Cells ...
    Apr 6, 2020 · Comparison of human primary with human iPS cell-derived dopaminergic neuron grafts in the rat model for Parkinson's disease. Stem Cell Rev.
  62. [62]
    Exercise promotes the functional integration of human stem cell ...
    Apr 24, 2025 · Human pluripotent stem cell (hPSC)-derived dopamine neurons can functionally integrate and reverse motor symptoms in Parkinson's disease models, ...
  63. [63]
    Peripheral Nerve Protection Strategies: Recent Advances and ...
    Apr 24, 2025 · Autologous nerve grafts remain the gold standard for repair; however, outcomes are frequently suboptimal due to donor site morbidity and ...
  64. [64]
    Peripheral Nerve Graft Surgery: Techniques and Recovery
    Sensory nerve grafts have success rates around 70–90%, while motor nerve recovery is lower, often 50–70%. In one study of 385 nerve repairs, including sensory, ...How Does Nerve Graft Surgery... · 1. Autograft (autologous... · How Is Nerve Graft Surgery...
  65. [65]
    Carpal tunnel surgery success rates explained - Practice Plus Group
    Sep 11, 2023 · The success rate of carpal tunnel surgery is over 90%. Delve into the factors that could affect the success of your carpal tunnel release.Carpal tunnel surgery: an... · Understanding success rates...Missing: sensory | Show results with:sensory
  66. [66]
    Medications for Treating ALS - The ALS Association
    The FDA approved Radicava as an IV treatment for ALS in 2017 followed by an oral suspension in 2022. Radicava is intended to slow the loss of physical function ...
  67. [67]
    Approved Drugs for ALS - Your ALS Guide
    There are currently two drugs that have been approved in the United States to treat people living with ALS—in addition to one that treats people who have a ...
  68. [68]
    A Randomized, Controlled Trial of Methylprednisolone or Naloxone ...
    May 17, 1990 · We conclude that in patients with acute spinal-cord injury, treatment with methylprednisolone in the dose used in this study improves neurologic recovery.
  69. [69]
    Spinal Cord Injuries Treatment & Management - Medscape Reference
    Mar 4, 2024 · The guidelines recommend that methylprednisolone not be used for the treatment of acute SCI within the first 24–48 hours following injury. The ...
  70. [70]
    Peripheral nerve repair throughout the body with processed nerve ...
    Feb 26, 2020 · Overall, 82% meaningful recovery (MR) was achieved across sensory, mixed, and motor nerve repairs up to gaps of 70 mm. No related adverse events ...
  71. [71]
    Changes in electrophysiological parameters after open carpal ... - NIH
    Aug 28, 2012 · From the 17 evaluated hand with moderate, moderate to severe and severe CTS, severity improvement was reported in 82.3% 6 months and in 88.2% 9 ...Missing: recovery rate
  72. [72]
    188 American Spinal Injury Association (ASIA) Impairment...
    RESULTS: Improvement of at least 1 AIS grade from presentation was noted in 27% of patients (mean improvement of 0.3 AIS grades, p < 0.001) upon discharge. ...Missing: repair | Show results with:repair
  73. [73]
    Drug for rare form of ALS, based in part on WashU research ...
    Drug for rare form of ALS, based in part on WashU research, approved by FDA. Washington University neurologist led international trials of the drug, tofersen.
  74. [74]
    Latest Treatments for Neuropathy: Exploration & Study Data (2025)
    Overall, MSCs show promise in treating neuropathy by promoting nerve regeneration, reducing inflammation, and providing neuroprotection. However, more research ...
  75. [75]
    Transcranial Optogenetic Stimulation Promotes Corticospinal Tract ...
    Jun 30, 2025 · Transcranial optogenetic stimulation promotes corticospinal tract axon regeneration to repair spinal cord injury by activating the JAK2/STAT3 pathway.
  76. [76]
    Optogenetic control of receptor-mediated growth cone dynamics in ...
    Jan 28, 2025 · Here, we demonstrate the use of an optogenetic tool to activate receptor signaling locally to induce actin-mediated growth cone remodeling in neurons.
  77. [77]
    Single-cell analysis of innate spinal cord regeneration identifies ...
    Aug 15, 2024 · This study provides a comprehensive resource of the cells and mechanisms that direct spinal cord regeneration and establishes zebrafish as a model of ...
  78. [78]
    Precise rewiring of corticospinal axons and spinal interneurons via ...
    Aug 1, 2025 · This in situ optogenetic modulation successfully demonstrated the restoration of spinal motor circuits by rebuilding functional connections ...<|separator|>
  79. [79]
    Advances of Genome Editing with CRISPR/Cas9 in ... - ResearchGate
    Dec 6, 2023 · Advances of Genome Editing with CRISPR/Cas9 in Neurodegeneration: The Right Path towards Therapy. MDPI. Biomedicines. December 2023; 11(12):3333.
  80. [80]
    CRISPR and Artificial Intelligence in Neuroregeneration
    The purpose of this review is to bring together findings from recent developments in genome editing and computational approaches, which center around the ...
  81. [81]
    Generation of Neural Organoids and Their Application in Disease ...
    May 24, 2025 · Organoids generated from patient-derived stem cells can model the pathological processes of various neurological disorders, such as autism, ...Missing: neuroregeneration | Show results with:neuroregeneration
  82. [82]
    Bioengineering tools for next-generation neural organoids
    We review recent progress in using bioengineering methods to improve neural organoid formation and analysis by leveraging microfabrication, biomaterials, 3D ...Missing: neuroregeneration testing differences
  83. [83]
    Artificial Intelligence and Machine Learning in Neuroregeneration
    AI-driven drug discovery holds promise for identifying neuroregenerative therapies. Nonetheless, addressing existing limitations remains crucial in this rapidly ...
  84. [84]
    AI breakthrough finds life-saving insights in everyday bloodwork
    Sep 23, 2025 · AI-powered analysis of routine blood tests can reveal hidden patterns that predict recovery and survival after spinal cord injuries.
  85. [85]
    Artificial Intelligence and Machine Learning in Neuroregeneration
    Jun 27, 2025 · These technologies aid in diagnosing diseases and facilitating functional recovery through robotics and targeted stimulation. AI-driven drug ...