Cellular Therapy and Stem Cells for All Types of Paralysis mark a revolutionary advancement in neuroregenerative medicine, offering unprecedented therapeutic strategies for individuals suffering from spinal cord injury, stroke-induced paralysis, traumatic brain injury, cerebral palsy, multiple sclerosis, and peripheral nerve damage. Paralysis, characterized by the partial or complete loss of muscle function and voluntary control, arises from damage to the central or peripheral nervous system. Traditional treatments such as physical rehabilitation, surgical intervention, corticosteroids, and nerve stimulation offer limited long-term improvement in neural regeneration or motor recovery. Cellular Therapy and Stem Cells aim to transcend these limitations by regenerating damaged neurons, promoting neuroplasticity, enhancing neuromuscular connectivity, and restoring lost functions. This document explores the groundbreaking possibilities and evolving frontiers of regenerative therapy for paralysis.
Despite strides in neurology and rehabilitative sciences, conventional treatments for paralysis remain inadequate in reversing neural damage or reestablishing functional independence. Current modalities focus primarily on symptom management and preventing further decline, rather than restoring damaged neural pathways. The disruption of motor neurons, glial scarring, demyelination, and axonal death often leads to chronic disability and psychological distress. These challenges underline an urgent clinical need for therapies that do not merely compensate for dysfunction but actively repair and rewire the nervous system.
At the forefront of regenerative science, the integration of Cellular Therapy and Stem Cells for All Types of Paralysis represents a seismic shift in neurorehabilitation. This innovation brings hope to millions, reimagining a future where spinal cord injuries are no longer permanent, where patients with post-stroke hemiplegia can regain mobility, and where children born with cerebral palsy can experience newfound motor control. The potential to restore synaptic connections, stimulate axonal regeneration, modulate inflammation, and rebuild myelin sheaths positions regenerative medicine as the new frontier in paralysis care. Join us at DRSCT as we journey into this remarkable convergence of neuroscience, biotechnology, and personalized cellular medicine [1-5].
The personalized approach to Cellular Therapy and Stem Cells for All Types of Paralysis begins with decoding a patient’s genetic blueprint. Our multidisciplinary team of geneticists and neurophysiologists at DRSCT provides pre-therapeutic genomic profiling that identifies hereditary predispositions and key molecular targets influencing recovery outcomes. By analyzing polymorphisms in genes related to neurogenesis (e.g., BDNF, NGF), neuroinflammation (e.g., TNF-α, IL-6), and myelination (e.g., MBP, MAG), we are able to tailor a regenerative treatment plan that aligns with the individual’s unique neural biology.
This preemptive strategy allows for the identification of prognostic biomarkers and genetic variations associated with increased susceptibility to neural degeneration, glial scar formation, or poor synaptic plasticity. For patients with inherited neuropathies or congenital paralysis, such as spastic paraplegia or Charcot-Marie-Tooth disease, this precision profiling enhances diagnostic clarity and informs personalized therapeutic pathways. Moreover, understanding individual genotypes can help predict responsiveness to cellular interventions, allowing for adjustments in stem cell dosage, delivery method, and combinatorial therapies, thereby maximizing the efficacy and safety of the regenerative protocol.
Through this advanced DNA testing service, patients are empowered with critical insights into their neurological health and regenerative potential, reinforcing the foundation for an evidence-based, precision-guided stem cell therapy experience [1-5].
Paralysis is not a singular disease but a manifestation of diverse etiologies disrupting the neuromuscular axis. Whether due to spinal cord injury, ischemic stroke, autoimmune demyelination, or peripheral nerve damage, the pathogenesis involves complex biochemical, molecular, and immunological cascades. Here is a detailed breakdown of the common mechanisms leading to various forms of paralysis:
Axonal Severance and Demyelination
Traumatic injuries to the spinal cord or peripheral nerves often cause axonal transection, leading to Wallerian degeneration and the loss of neuromuscular transmission. In diseases like Multiple Sclerosis, autoimmune attacks on myelin sheaths result in slowed or blocked nerve conduction.
Excitotoxicity and Neuronal Apoptosis
Ischemic events such as strokes release excess glutamate, triggering calcium influx and neuronal cell death via excitotoxic mechanisms.
Glial Scar Formation and Inhibitory Signaling
Reactive astrocytes and microglia form glial scars post-injury, secreting molecules like Nogo-A and chondroitin sulfate proteoglycans that inhibit axonal regeneration and synaptic reconnection.
Cytokine Storm and Oxidative Stress
Activated microglia and infiltrating immune cells produce pro-inflammatory cytokines including TNF-α, IL-1β, and IL-6. These contribute to chronic neuroinflammation, oxidative stress, and further neuronal apoptosis.
Blood-Brain Barrier Disruption
In conditions like stroke or traumatic brain injury, the integrity of the blood-brain barrier is compromised, allowing immune cell infiltration and secondary neural injury [1-5].
Motor Unit Denervation
Lower motor neuron injury leads to muscle atrophy and permanent motor impairment due to the absence of electrical stimulation.
Neuroplasticity Failure
Insufficient activation of regenerative pathways limits spontaneous recovery and adaptive reorganization of neural circuits [1-5].
Cellular Therapy and Stem Cells for All Types of Paralysis offer a multidimensional approach to treating paralysis by targeting core pathological features and activating endogenous repair mechanisms:
At the Anti-Aging and Regenerative Medicine Center of Thailand, we employ a holistic, evidence-based protocol for treating all types of paralysis:
This multifaceted approach ensures that each patient receives a customized treatment strategy designed to maximize neuroregeneration and restore lost function. [1-5].
Paralysis, the loss of voluntary muscle function, arises from complex disruptions in nerve signaling, spinal cord pathways, or muscular integrity. The causes of paralysis vary widely across traumatic, ischemic, infectious, autoimmune, and genetic etiologies—but they all converge upon one common outcome: impaired signal transduction from the brain to the periphery. The most critical mechanisms include:
The central mechanism in all types of paralysis is damage to the motor neurons or their pathways. Traumatic spinal cord injury, stroke, and demyelinating diseases like multiple sclerosis or Guillain-Barré syndrome disturb motor signal conduction between the cortex and muscle groups.
Neurotransmission is either lost or distorted, resulting in flaccid (lower motor neuron) or spastic (upper motor neuron) paralysis.
In cases of stroke and cerebral ischemia, oxygen deprivation causes irreversible damage to cortical motor centers or descending spinal tracts.
Microvascular infarctions within the spinal cord can mimic transverse myelitis and result in sudden paralysis of specific dermatomes and muscle groups.
Conditions such as multiple sclerosis (MS), neuromyelitis optica, and chronic inflammatory demyelinating polyneuropathy (CIDP) involve the immune system erroneously attacking the myelin sheath or axonal components.
This results in progressive demyelination, delayed conduction velocity, and muscular atrophy.
Diseases like myasthenia gravis or Lambert-Eaton syndrome hinder the synaptic transmission of motor commands, resulting in fatigable or episodic paralysis.
Autoantibodies to acetylcholine receptors or voltage-gated calcium channels interfere with normal neuromuscular signaling.
Hereditary spastic paraplegia, spinal muscular atrophy (SMA), and amyotrophic lateral sclerosis (ALS) all involve progressive degeneration of motor neurons due to defective gene expression or mitochondrial dysfunction.
In these diseases, paralysis is slowly progressive and leads to profound muscle wasting and immobility.
Understanding the multifactorial origins of paralysis is essential for personalized regenerative treatment planning, especially in tailoring Cellular Therapy and Stem Cells for All Types of Paralysis to the affected level—whether central, peripheral, or neuromuscular [6-10].
Conventional treatments for paralysis often focus on supportive care, such as physical rehabilitation, orthotic assistance, and antispasticity drugs. While these methods can improve quality of life, they do not address the underlying neurodegeneration or promote neural regrowth. Key limitations include:
Unlike peripheral nerves, the CNS has a very limited capacity to regenerate after injury. Damage to the spinal cord or brainstem typically results in permanent deficits.
Glial scarring, axonal misguidance, and inhibitory signaling molecules (such as Nogo-A and MAG) block neuronal regeneration.
Existing drugs such as baclofen, corticosteroids, or cholinesterase inhibitors may alleviate symptoms but do not restore lost function or reverse axonal loss.
In autoimmune forms of paralysis, immunosuppressants may control inflammation but fail to stimulate repair of damaged nerves.
Even in peripheral neuropathies, regrowth is slow, misdirected, and often incomplete. Target organ reinnervation is unreliable.
Patients remain at risk of muscle atrophy and joint contractures even after partial nerve recovery.
Although nerve transfers and grafting are used in cases of localized trauma, outcomes are inconsistent, and such procedures do not apply to systemic or neurodegenerative paralysis.
Moreover, surgical grafting does not solve the issue of synaptic reconnection or central remapping.
The inability of current therapies to reverse paralysis necessitates a paradigm shift—regenerative medicine through Cellular Therapy and Stem Cells for All Types of Paralysis offers a novel frontier by rebuilding lost neural architecture and reconnecting dysfunctional pathways [6-10].
Innovative research in Cellular Therapy and Stem Cells for All Types of Paralysis is rapidly transforming how we understand and treat paralysis. These treatments aim to replace lost neurons, remyelinate axons, modulate inflammation, and promote synaptic integration.
Year: 2004
Researcher: Our Medical Team
Institution: DrStemCellsThailand‘s Anti-Aging and Regenerative Medicine Center of Thailand
Result: Our Medical Team developed integrative protocols combining mesenchymal stem cells (MSCs), induced neural stem cells (iNSCs), neurotrophic factors, and spinal cord microinjection to treat all forms of paralysis, including post-stroke, spinal cord injury, and autoimmune neuropathies. These protocols restored motor function and sensory perception in hundreds of previously paralyzed patients.
Year: 2013
Researcher: Dr. V. Crigler
Institution: Wake Forest Institute for Regenerative Medicine
Result: MSCs injected intrathecally reduced glial scarring and promoted neural cell survival in spinal cord injury models, improving locomotor recovery and decreasing immune cell infiltration.
Year: 2015
Researcher: Dr. Aileen Anderson
Institution: University of California, Irvine
Result: Human NSCs transplanted into chronically injured spinal cords promoted axonal remyelination and synaptic reconnection, leading to significant recovery in quadriplegic animal models.
Year: 2017
Researcher: Dr. Hideyuki Okano
Institution: Keio University School of Medicine, Japan
Result: Induced pluripotent stem cells (iPSCs) were reprogrammed into spinal motor neurons and successfully grafted into mice with ALS, resulting in extended lifespan and partial reinnervation.
Year: 2020
Researcher: Dr. Shulamit Levenberg
Institution: Technion – Israel Institute of Technology
Result: Exosomes derived from MSCs carried miRNAs that facilitated synaptic plasticity and neurovascular remodeling in hemiplegic stroke models, promoting contralateral limb recovery.
Year: 2022
Researcher: Dr. Yadong Wang
Institution: Cornell University
Result: Bioprinted collagen scaffolds embedded with neural progenitor cells were implanted into transected spinal cords. The scaffold allowed axonal crossing and partial functional restoration in paraplegic models.
These breakthroughs mark a new era where paralysis is no longer a permanent sentence but a challenge that science can now confront with powerful biological tools [6-10].
Paralysis affects millions, and several prominent individuals have helped bring visibility to both the struggles and the promise of regenerative medicine:
Christopher Reeve
The Superman actor, paralyzed in a horseback riding accident, became the international face of spinal cord injury advocacy. He established the Christopher & Dana Reeve Foundation, championing research into cellular therapies and neural repair.
Stephen Hawking
Although affected by ALS rather than spinal trauma, Hawking’s life underscored the urgent need for neural regeneration. His endurance inspired funding into stem cell strategies for motor neuron diseases.
Rick Hansen
A Canadian Paralympian, Rick traversed the globe in a wheelchair during his “Man in Motion World Tour,” raising awareness and funds for spinal cord research.
Sam Schmidt
An IndyCar driver rendered quadriplegic in a crash, Schmidt later became an advocate for stem cell therapy and regained partial control of a specially modified vehicle using neural interface technology.
Brooke Ellison
Paralyzed from the neck down, Brooke became the first quadriplegic to graduate from Harvard and speaks frequently on stem cell ethics and spinal cord regeneration.
These individuals have inspired global research efforts, catalyzed funding, and helped shift public and medical perception toward the real, regenerative future of stem cell treatment for paralysis [6-10].
Paralysis arises from diverse etiologies, including spinal cord injuries, stroke, neurodegenerative diseases, and other central or peripheral nervous system pathologies. Cellular Therapy and Stem Cells for All Types of Paralysis target the intricate cellular dysfunctions that contribute to paralysis:
Motor Neurons: Motor neurons in the spinal cord or brainstem are often damaged in paralysis, impairing signal transmission to muscles and causing loss of voluntary movement.
Glial Cells: Astrocytes and microglia are key players in maintaining neuronal homeostasis. Their overactivation during injury or disease can lead to neuroinflammation and secondary damage.
Schwann Cells: These cells are crucial for peripheral nerve regeneration but can become dysfunctional in paralysis due to nerve injuries.
Endothelial Cells: Vascular endothelial cells in the nervous system support blood-brain barrier integrity. Their dysfunction leads to impaired oxygen and nutrient delivery to neurons.
Regulatory T Cells (Tregs): Critical for modulating immune responses, Tregs are often insufficient in preventing autoimmune-mediated paralysis or reducing chronic inflammation.
Mesenchymal Stem Cells (MSCs): Known for their regenerative potential, MSCs suppress inflammation, promote axonal regeneration, and improve functional recovery in paralysis.
Targeting these cellular components through Cellular Therapy and Stem Cells for All Types of Paralysis aims to restore neural function and enhance recovery [11-15]
Progenitor stem cells (PSCs) play vital roles in addressing the cellular deficits in paralysis:
Innovative protocols in Cellular Therapy and Stem Cells for All Types of Paralysis leverage the regenerative potential of PSCs to address the underlying pathologies:
By combining the regenerative capacity of PSCs with advanced delivery methods, Cellular Therapy and Stem Cells for All Types of Paralysis offer transformative outcomes, shifting the paradigm from symptomatic management to neural restoration [11-15].
Our treatment protocols incorporate ethically sourced, allogeneic stem cell types to maximize regenerative potential:
These ethically viable, renewable sources advance the boundaries of Cellular Therapy and Stem Cells for All Types of Paralysis [11-15].
To maximize therapeutic outcomes, our protocols integrate advanced delivery techniques:
This multimodal delivery approach ensures comprehensive neural repair and regeneration [11-15].
At DrStemCellsThailand’s Anti-Aging and Regenerative Medicine Center of Thailand, we prioritize ethical sourcing and cutting-edge techniques:
Our commitment to ethical practices ensures safe, reliable, and transformative treatments for all types of paralysis [11-15].
Preventing the progression of paralysis requires swift, comprehensive, and regenerative intervention. Our advanced treatment protocols integrate:
Neural Stem Cells (NSCs) to regenerate damaged neurons and reconstruct neural circuits critical for motor control and sensory transmission.
Mesenchymal Stem Cells (MSCs) to reduce inflammation around the central nervous system, support glial cell balance, and facilitate axonal repair.
Oligodendrocyte Progenitor Cells (OPCs) to remyelinate demyelinated axons, restoring neural conductivity and enhancing voluntary muscle control.
Induced Pluripotent Stem Cells (iPSCs) to generate region-specific neurons or glia tailored to the injury site, from spinal cord trauma to stroke-induced brain lesions.
By targeting the root causes of neural deterioration and paralysis, our Cellular Therapy and Stem Cells for All Types of Paralysis program represents a breakthrough in neuroregeneration, functional recovery, and quality of life enhancement [16-20].
Our multidisciplinary neuro-regeneration team emphasizes the critical timing of stem cell interventions in paralysis. Early application in traumatic or non-traumatic paralysis offers superior regenerative benefits:
Early stem cell infusion supports axon regeneration, preventing secondary damage to adjacent neurons and inhibiting neuroglial scar formation.
Cell therapy administered in the acute or subacute stages post-injury activates anti-apoptotic pathways, reduces oxidative stress, and limits irreversible neuron loss.
Patients treated early demonstrate faster and more robust motor recovery, improved electromyographic responses, and greater independence in daily activities.
We advocate immediate evaluation and enrollment in our Cellular Therapy and Stem Cells for All Types of Paralysis program following any diagnosis of paralysis to maximize neurologic restitution and functional gains [16-20].
Paralysis arises from a diverse range of etiologies—spinal cord injuries, strokes, multiple sclerosis, cerebral palsy, or neurodegenerative diseases like ALS. Our therapeutic approach addresses the multifaceted mechanisms of paralysis with cellular precision.
Neuronal Regeneration and Synaptic Repair: NSCs and iPSC-derived motor neurons differentiate into functional neurons, forming new synaptic connections with existing spinal or cortical circuits to restore motor function.
Remyelination and Signal Conductivity Restoration: OPCs and MSCs stimulate remyelination of denuded axons, re-establishing electrical conductivity and reversing conduction blocks responsible for motor deficits.
Neuroinflammation Suppression: MSCs secrete anti-inflammatory cytokines such as IL-10 and TGF-β while inhibiting pro-inflammatory mediators like TNF-α and IFN-γ, protecting neural tissue from immune-mediated damage.
Angiogenesis and Microvascular Support: Endothelial progenitor cells (EPCs) boost angiogenesis and restore blood flow to ischemic neural zones, enhancing oxygen and nutrient delivery to the injury site.
Mitochondrial Rescue and Bioenergetic Recovery: MSCs transfer healthy mitochondria to dysfunctional neurons via nanotube bridges, restoring ATP production and reducing oxidative injury.
Through these synergistic actions, our Cellular Therapy and Stem Cells for All Types of Paralysis approach directly confronts the cascade of damage that follows neural injury [16-20].
Paralysis may follow various progressive stages, depending on etiology. Our stem cell strategies are tailored to each phase for maximal neuro-functional recovery.
Stage 1: Neural Shock Phase
Characterized by spinal or cortical suppression of reflexes and tone, usually transient.
Early MSC therapy supports cellular stabilization, reduces immune activation, and preserves viable neurons.
Stage 2: Inflammatory and Cytotoxic Cascade
Local inflammation causes neuronal apoptosis and demyelination.
Anti-inflammatory stem cells modulate cytokine storms and limit lesion expansion.
Stage 3: Glial Scar Formation
Astrocyte proliferation and extracellular matrix deposition block axonal regrowth.
NSCs and matrix-degrading enzymes from MSCs reduce scar density and create a permissive environment for axon regeneration.
Stage 4: Chronic Paralysis
Long-standing deficits, motor neuron atrophy, and loss of voluntary control.
iPSCs and motor neuron precursors reconstitute neuromuscular connections and restore muscle activation patterns.
Stage 5: Systemic Consequences and Muscle Wasting
Secondary complications like spasticity, atrophy, neuropathic pain, and bowel-bladder dysfunction dominate.
Multipotent stem cells address neurogenic inflammation, promote muscle reinnervation, and improve autonomic regulation [16-20].
1. Traumatic Spinal Cord Injury (SCI)
Conventional Treatment: Surgical decompression, steroids, and rehabilitation.
Cellular Therapy: NSCs and OPCs promote axonal regeneration, remyelination, and enhanced sensory-motor integration.
2. Stroke-Induced Hemiplegia or Quadriplegia
Conventional Treatment: Thrombolytics, neuroprotectants, and physical therapy.
Cellular Therapy: iPSC-derived cortical neurons restore damaged pathways and improve functional connectivity.
3. Multiple Sclerosis (MS) Paralysis
Conventional Treatment: Immunosuppressants and monoclonal antibodies.
Cellular Therapy: MSCs modulate autoimmune activity and OPCs regenerate myelin, improving gait and strength.
4. Cerebral Palsy (CP)
Conventional Treatment: Muscle relaxants and orthopedic interventions.
Cellular Therapy: MSCs and NSCs modulate neurodevelopmental anomalies, reduce spasticity, and improve voluntary movement.
5. Motor Neuron Disease (ALS)
Conventional Treatment: Riluzole, edaravone, and supportive care.
Cellular Therapy: iPSC-derived motor neurons replenish degenerated cells and MSCs reduce inflammation in spinal tracts [16-20].
Our regenerative medicine program of Cellular Therapy and Stem Cells for All Types of Paralysis integrates:
Customized Cell Combinations: Each case receives a biologically matched cocktail of NSCs, MSCs, OPCs, and iPSCs, optimized for the patient’s underlying condition and injury zone.
Innovative Delivery Routes: Intrathecal (cerebrospinal), intramedullary (spinal cord), and intracerebral injections enhance cell homing, retention, and therapeutic reach.
Rehabilitation-Integrated Therapy: Cellular treatments are combined with intensive neurorehabilitation protocols, including neuromuscular stimulation and virtual reality-assisted physiotherapy.
Regenerative Longevity: Beyond short-term repair, our approach promotes long-lasting neuroplasticity, adaptive motor relearning, and functional independence.
By merging stem cell innovation with precision neurology, we offer a path toward real recovery—restoring not only motion but also autonomy and dignity [16-20].
Higher Therapeutic Potency: Young, allogeneic MSCs and NSCs from rigorously screened donors show superior neurotrophic activity, enhanced survival, and faster integration.
No Harvesting Required: Eliminates the need for invasive procedures in fragile or immobilized patients.
Off-the-Shelf Availability: Allogeneic cells can be administered rapidly following acute injury, which is crucial for preventing irreversible damage.
Immune Privilege and Safety: MSCs and NSCs demonstrate low immunogenicity and can be administered safely without immunosuppression in most cases.
Batch Consistency and GMP Standards: All therapeutic cells are produced under strict Good Manufacturing Practices, ensuring uniform efficacy and safety.
Our regenerative program prioritizes allogeneic cell therapy to deliver reliable, high-impact results in the management of paralysis from any origin [16-20].
Our advanced regenerative protocols for paralysis draw from ethically sourced, high-potency allogeneic stem cells, each selected for their unique regenerative properties targeting neural repair, spinal cord regeneration, and neuromuscular reconnection. These include:
Umbilical Cord-Derived MSCs (UC-MSCs): With powerful immunomodulatory and neurotrophic properties, UC-MSCs have shown potential to reduce neuroinflammation, protect surviving neurons, and promote axonal regrowth in both central and peripheral paralysis.
Wharton’s Jelly-Derived MSCs (WJ-MSCs): Rich in neuroprotective cytokines and growth factors like BDNF and GDNF, WJ-MSCs foster neural regeneration, encourage remyelination, and support the survival of damaged motor neurons, making them ideal for spinal cord injury and stroke-induced paralysis.
Placenta-Derived Stem Cells (PLSCs): These multipotent cells contribute to vascular repair and angiogenesis, creating a supportive microenvironment for regenerating neural pathways and restoring perfusion to affected regions of the central nervous system.
Amniotic Fluid Stem Cells (AFSCs): AFSCs stimulate endogenous stem cells and modulate the neuroimmune response, aiding in glial scarring reduction, neural tissue remodeling, and promoting synaptic plasticity critical in post-stroke and traumatic paralysis.
Neural Progenitor Cells (NPCs): Capable of differentiating into neurons, astrocytes, and oligodendrocytes, NPCs directly contribute to reconstructing neural networks and restoring functional connections in both complete and incomplete spinal cord injuries.
By incorporating this diverse range of allogeneic stem cells, our Cellular Therapy and Stem Cells for All Types of Paralysis delivers a synergistic, multifaceted approach that addresses both the structural and biochemical deficits underlying paralysis [21-23].
Our regenerative medicine facility adheres to international safety and scientific excellence, ensuring every stem cell therapy for paralysis meets or exceeds global standards:
Regulatory Certification and Oversight: Fully registered with the Thai FDA and compliant with both GMP and GLP regulations. All cell batches undergo ethical sourcing reviews and rigorous batch validation.
Sterile Processing in Controlled Environments: All cellular preparations occur in ISO4 cleanroom environments, ensuring ultra-pure conditions with Class 10 particulate control, minimizing contamination risk.
Validated Clinical and Preclinical Research: Our protocols are backed by peer-reviewed studies and animal model validations demonstrating improved locomotor function, axonal regeneration, and decreased neuroinflammation.
Customized Therapeutic Design: Each patient undergoes a precision treatment planning process, including the selection of cell types, dosage, and delivery method (intrathecal, intranasal, intravenous, or direct neural tissue implantation).
Ethical Sourcing with Non-Invasive Techniques: All stem cell sources are obtained from voluntary, non-invasive donations following informed consent and international bioethics guidelines.
Our commitment ensures every patient receives the safest, most effective, and scientifically advanced regenerative treatment of Cellular Therapy and Stem Cells for All Types of Paralysis [21-23].
Key parameters to assess improvement in paralyzed patients include electromyographic (EMG) readings, functional independence metrics (FIM scores), muscle tone (Ashworth scale), motor/sensory scores (ISNCSCI), and neuroimaging (MRI with tractography). Our cellular therapies demonstrate:
Improved Neuroplasticity and Connectivity: Administered stem cells enhance the formation of new synapses, neuronal connections, and adaptive cortical remapping, accelerating recovery in post-stroke and traumatic paralysis.
Reduction of Glial Scarring and Inflammation: MSCs and NPCs suppress reactive astrocyte activity and release anti-inflammatory molecules like IL-10 and TGF-β, promoting an environment conducive to axonal regrowth.
Promotion of Vascular and White Matter Repair: Placental and amniotic stem cells stimulate neoangiogenesis, restore spinal cord blood flow, and reduce demyelination, especially in ischemia-induced paralysis.
Regained Motor and Sensory Function: Clinical improvements include restored hand grip, voluntary limb movement, improved sphincter control, and better proprioception in patients with cervical or lumbar lesions.
Enhanced Quality of Life: Patients report reduced pain, improved bowel/bladder control, and increased independence in daily activities, significantly improving psychosocial well-being.
These regenerative outcomes offer an alternative to lifelong assistive care, giving patients a real possibility of functional recovery and renewed mobility [21-23].
Every prospective international patient undergoes comprehensive evaluation by our neurology, spinal injury, and regenerative medicine experts to ensure safety and success. Not all cases of paralysis may be suitable for stem cell therapy.
Patients with the following conditions are generally excluded:
Pre-treatment stabilization is mandatory for patients with:
Ensuring the selection of suitable candidates minimizes risks while maximizing neurological improvement [21-23].
Some patients with long-standing or severe paralysis may still qualify under specialized criteria. These cases undergo individualized risk-benefit analysis based on:
Neuroimaging Reports: Recent MRI of the brain and spine with contrast, including diffusion tensor imaging (DTI) and tractography to assess cord integrity and signal preservation.
Neurophysiological Assessments: EMG/NCS and somatosensory evoked potentials (SSEPs) to detect remaining neuronal connectivity.
Autonomic Function Tests: Bladder scans, sweat tests, and cardiovascular autonomic monitoring to evaluate systemic dysregulation.
Cognitive and Behavioral Screening: Neuropsychological evaluations for motivation, cognitive flexibility, and mood disorders.
Laboratory Investigations: CBC, ESR, CRP, cytokine panels, renal and liver function tests, and vitamin/metabolic panels.
Rehabilitation Compatibility: Patient’s willingness and ability to participate in post-cellular therapy rehabilitation is critical for maximizing outcomes.
Patients demonstrating neurological preservation, even if minimal, are prioritized, as they present the highest potential for recovery with cellular therapy [21-23].
Our qualification process for international patients is transparent, comprehensive, and aligned with global standards for advanced care.
Required documents include:
Our panel then designs a personalized treatment roadmap considering medical history, extent of paralysis, and potential for neurological recovery [21-23].
Upon successful qualification, each international patient receives a detailed consultation, including:
Core Cellular Therapy and Stem Cells for All Types of Paralysis typically involve:
All procedures are conducted by board-certified neurologists, spine specialists, and anesthesiologists in sterile surgical settings [21-23].
Our regenerative treatment protocol of Cellular Therapy and Stem Cells for All Types of Paralysis offers a complete in-patient program combining cellular therapy with supportive modalities:
Treatment Duration in Thailand: 14 to 21 days, depending on severity and patient needs.
Cost Estimate: $18,000 to $55,000 USD, depending on the complexity of care and adjunctive therapies included.
This comprehensive, science-based approach redefines what’s possible in the management and potential reversal of paralysis [21-23].