Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Andrew M. Tan is active.

Publication


Featured researches published by Andrew M. Tan.


The Journal of Neuroscience | 2009

BDNF- hypersecreting human mesenchymal stem cells promote functional recovery, axonal sprouting and protection of corticospinal neurons after spinal cord injury

Masanori Sasaki; Christine Radtke; Andrew M. Tan; Peng Zhao; Hirofumi Hamada; Kiyohiro Houkin; Osamu Honmou; Jeffery D. Kocsis

Transplantation of mesenchymal stem cells (MSCs) derived from bone marrow has been shown to improve functional outcome in spinal cord injury (SCI). We transplanted MSCs derived from human bone marrow (hMSCs) to study their potential therapeutic effect in SCI in the rat. In addition to hMSCs, we used gene-modified hMSCs to secrete brain-derived neurotrophic factor (BDNF-hMSCs). After a dorsal transection lesion was induced at T9, cells were microinjected on each side of the transection site. Fluorogold (FG) was injected into the epicenter of the lesion cavity to identify transected corticospinal tract (CST) neurons. At 5 weeks after transplantation, the animals were perfused. Locomotor recovery improvement was observed for the BDNF-hMSC group, but not in the hMSC group. Structurally there was increased sprouting of injured corticospinal tract and serotonergic projections after hMSC and BDNF-hMSC transplantation. Moreover, an increased number of serotonergic fibers was observed in spinal gray matter including the ventral horn at and below the level of the lesion, indicating increased innervation in the terminal regions of a descending projection important for locomotion. Stereological quantification was performed on the brains to determine neuronal density in primary motor (M1) cortex. The number of FG backfilled cells demonstrated an increased cell survival of CST neurons in M1 cortex in both the hMSC and BDNF-hMSC groups at 5 weeks, but the increase for the BDNF-hMSC group was greater. These results indicate that transplantation of hMSCs hypersecreting BDNF results in structural changes in brain and spinal cord, which are associated with improved functional outcome in acute SCI.


The Journal of Neuroscience | 2006

Antibodies against the NG2 Proteoglycan Promote the Regeneration of Sensory Axons within the Dorsal Columns of the Spinal Cord

Andrew M. Tan; Mario Colletti; Ann T. Rorai; J. H. Pate Skene; Joel M. Levine

The NG2 chondroitin sulfate proteoglycan inhibits axon growth in vitro. Levels of NG2 increase rapidly in the glial scars that form at sites of CNS injury, suggesting that NG2 may inhibit axon regeneration. To determine the functions of NG2, we infused mixtures of neutralizing or non-neutralizing anti-NG2 monoclonal antibodies into the dorsally transected adult rat spinal cord and analyzed the regeneration of ascending mechanosensory axons anatomically. At 1 week after injury, ascending sensory axons in control animals terminated caudal to the lesion within an area containing dense deposits of NG2 immunoreactivity. In animals treated with the neutralizing anti-NG2 antibodies, labeled axons penetrated the caudal border of the lesion and grew into and beyond the lesion center. The low intrinsic growth capacity of adult neurons may also limit the ability of damaged axons to regenerate. To enhance growth, we combined antibody treatment with a peripheral nerve conditioning lesion. After a conditioning lesion and treatment with control, non-neutralizing antibodies, many sensory axons grew into the lesion core. These axons did not grow past the rostral border of the lesion; rather, they grew along the dorsal surface of the spinal cord and within any remaining pieces of the dorsal roots. In contrast, combining a peripheral nerve conditioning lesion with neutralizing anti-NG2 antibodies resulted in sensory axon regeneration past the glial scar and into the white matter rostral to the injury site. The combinatorial approach used here that neutralizes extrinsic inhibition and increases intrinsic growth results in anatomically correct axon regeneration, a prerequisite for functional recovery.


The Journal of Neuroscience | 2008

Neuropathic Pain Memory Is Maintained by Rac1-Regulated Dendritic Spine Remodeling after Spinal Cord Injury

Andrew M. Tan; Severine Stamboulian; Yu-Wen Chang; Peng Zhao; Avis B. Hains; Stephen G. Waxman; Bryan C. Hains

Localized increases in synaptic strength constitute a synaptic basis for learning and memory in the CNS and may also contribute to the maintenance of neuropathic pain after spinal cord injury (SCI) through the de novo formation or elaboration of postsynaptic dendritic structures. To determine whether SCI-induced dendritic spine remodeling contributes to neuronal hyperexcitability and neuropathic pain, we analyzed spine morphometry, localization, and functional influence in dorsal horn (DH) neurons in adult rats 1 month after sham surgery, contusion SCI, and SCI treated with a selective inhibitor of Rac1 activation, NSC23766. After SCI, DH neurons located in lamina IV–V exhibited increased spine density, redistributed spines, and mature spines compared with control neurons, which was associated with enhancement of EPSCs in computer simulations and hyperexcitable responsiveness to innocuous and noxious peripheral stimuli in unit recordings in vivo. SCI animals also exhibited symptoms of tactile allodynia and thermal hyperalgesia. Inhibition of the small GTP-binding protein Rac1 ameliorated post-SCI changes in spine morphology, attenuated injury-induced hyperexcitability of wide-dynamic range neurons, and progressively increased pain thresholds over a 3 d period. This suggests that Rac1 is an important intracellular signaling molecule involved in a spinal dendritic spine pathology associated with chronic neuropathic pain after SCI. Our report provides robust evidence for a novel conceptual bridge between learning and memory on the one hand, and neuropathic pain on the other.


The Journal of Pain | 2010

Unilateral Focal Burn Injury Is Followed by Long-Lasting Bilateral Allodynia and Neuronal Hyperexcitability in Spinal Cord Dorsal Horn

Yu-Wen Chang; Andrew M. Tan; Carl Y. Saab; Stephen G. Waxman

UNLABELLED Pain after burn injury can be intense and long lasting. Treatment is often ineffective, and there is a need for increased knowledge of the underlying pain mechanisms. In the present study, we established a unilateral partial-thickness burn injury model, which produces ipsilateral mechanical allodynia soon after injury, followed by contralateral allodynia. Chronic bilateral allodynia lasts up to 8 weeks postinjury in this model. In addition to the change in pain behavior, electrophysiological analyses showed that dorsal horn neurons become hyperexcitable and display significantly increased evoked activity with enlarged receptive fields, initially on the side ipsilateral to the injury, and subsequently on both sides of the spinal cord. It is known that, following nerve injury, activation of p38 mitogen-activated protein kinase (MAPK) pathways within spinal microglia contributes to the pathogenesis of pain. In our burn injury model, rapid and prolonged activation of phospho-p38-expressing microglia occurs bilaterally in the spinal cord dorsal horn. Taken together, these data demonstrate that a unilateral peripheral burn injury can produce long-lasting allodynia that can spread to the contralateral limb, together with dorsal horn neuronal hyperexcitability and microglial activation on both ipsilateral and contralateral sides of the spinal cord. Our results suggest that central neuropathic mechanisms can contribute to pain after burn injury. PERSPECTIVE Mechanisms contributing to pain following burn injury are incompletely understood. In a novel animal model of burn injury, we have demonstrated hyperexcitability of second-order sensory neurons, activation of microglia, and chronic bilateral pain following the burn injury. This work identifies potential therapeutic targets to alleviate pain after burn injury.


Molecular Therapy | 2013

Virus-mediated shRNA Knockdown of Nav1.3 in Rat Dorsal Root Ganglion Attenuates Nerve Injury-induced Neuropathic Pain

Omar Abdel Samad; Andrew M. Tan; Xiaoyang Cheng; Edmund Foster; Sulayman D. Dib-Hajj; Stephen G. Waxman

Neuropathic pain is a chronic condition that is often refractory to treatment with available therapies and thus an unmet medical need. We have previously shown that the voltage-gated sodium channel Na(v)1.3 is upregulated in peripheral and central nervous system (CNS) of rats following nerve injury, and that it contributes to nociceptive neuron hyperexcitability in neuropathic conditions. To evaluate the therapeutic potential of peripheral Na(v)1.3 knockdown at a specific segmental level, we constructed adeno-associated viral (AAV) vector expressing small hairpin RNA against rat Na(v)1.3 and injected it into lumbar dorsal root ganglion (DRG) of rats with spared nerve injury (SNI). Our data show that direct DRG injection provides a model that can be used for proof-of-principle studies in chronic pain with respect to peripheral delivery route of gene transfer constructs, high transduction efficiency, flexibility in terms of segmental localization, and limited behavioral effects of the surgical procedure. We show that knockdown of Na(v)1.3 in lumbar 4 (L4) DRG results in an attenuation of nerve injury-induced mechanical allodynia in the SNI model. Taken together, our studies support the contribution of peripheral Na(v)1.3 to pain in adult rats with neuropathic pain, validate Na(v)1.3 as a target, and provide validation for this approach of AAV-mediated peripheral gene therapy.Neuropathic pain is a chronic condition that is often refractory to treatment with available therapies and thus an unmet medical need. We have previously shown that the voltage-gated sodium channel Nav1.3 is upregulated in peripheral and central nervous system (CNS) of rats following nerve injury, and that it contributes to nociceptive neuron hyperexcitability in neuropathic conditions. To evaluate the therapeutic potential of peripheral Nav1.3 knockdown at a specific segmental level, we constructed adeno-associated viral (AAV) vector expressing small hairpin RNA against rat Nav1.3 and injected it into lumbar dorsal root ganglion (DRG) of rats with spared nerve injury (SNI). Our data show that direct DRG injection provides a model that can be used for proof-of-principle studies in chronic pain with respect to peripheral delivery route of gene transfer constructs, high transduction efficiency, flexibility in terms of segmental localization, and limited behavioral effects of the surgical procedure. We show that knockdown of Nav1.3 in lumbar 4 (L4) DRG results in an attenuation of nerve injury-induced mechanical allodynia in the SNI model. Taken together, our studies support the contribution of peripheral Nav1.3 to pain in adult rats with neuropathic pain, validate Nav1.3 as a target, and provide validation for this approach of AAV-mediated peripheral gene therapy.


The Journal of Neuroscience | 2012

Maladaptive Dendritic Spine Remodeling Contributes to Diabetic Neuropathic Pain

Andrew M. Tan; Omar Abdel Samad; Tanya Z. Fischer; Peng Zhao; Anna-Karin Persson; Stephen G. Waxman

Diabetic neuropathic pain imposes a huge burden on individuals and society, and represents a major public health problem. Despite aggressive efforts, diabetic neuropathic pain is generally refractory to available clinical treatments. A structure–function link between maladaptive dendritic spine plasticity and pain has been demonstrated previously in CNS and PNS injury models of neuropathic pain. Here, we reasoned that if dendritic spine remodeling contributes to diabetic neuropathic pain, then (1) the presence of malformed spines should coincide with the development of pain, and (2) disrupting maladaptive spine structure should reduce chronic pain. To determine whether dendritic spine remodeling contributes to neuropathic pain in streptozotocin (STZ)-induced diabetic rats, we analyzed dendritic spine morphology and electrophysiological and behavioral signs of neuropathic pain. Our results show changes in dendritic spine shape, distribution, and shape on wide-dynamic-range (WDR) neurons within lamina IV–V of the dorsal horn in diabetes. These diabetes-induced changes were accompanied by WDR neuron hyperexcitability and decreased pain thresholds at 4 weeks. Treatment with NSC23766 (N6-[2-[[4-(diethylamino)-1-methylbutyl]amino]-6-methyl-4-pyrimidinyl]-2-methyl-4,6-quinolinediamine trihydrochloride), a Rac1-specific inhibitor known to interfere with spine plasticity, decreased the presence of malformed spines in diabetes, attenuated neuronal hyperresponsiveness to peripheral stimuli, reduced spontaneous firing activity from WDR neurons, and improved nociceptive mechanical pain thresholds. At 1 week after STZ injection, animals with hyperglycemia with no evidence of pain had few or no changes in spine morphology. These results demonstrate that diabetes-induced maladaptive dendritic spine remodeling has a mechanistic role in neuropathic pain. Molecular pathways that control spine morphogenesis and plasticity may be promising future targets for treatment.


Experimental Neurology | 2012

Spinal cord injury, dendritic spine remodeling, and spinal memory mechanisms.

Andrew M. Tan; Stephen G. Waxman

Spinal cord injury (SCI) often results in the development of neuropathic pain, which can persist for months and years after injury. Although many aberrant changes to sensory processing contribute to the development of chronic pain, emerging evidence demonstrates that mechanisms similar to those underlying classical learning and memory can contribute to central sensitization, a phenomenon of amplified responsiveness to stimuli in nociceptive dorsal horn neurons. Notably, dendritic spines have emerged as major players in learning and memory, providing a structural substrate for how the nervous system modifies connections to form and store information. Until now, most information regarding dendritic spines has been obtained from studies in the brain. Recent experimental data in the spinal cord, however, demonstrate that Rac1-regulated dendritic spine remodeling occurs on second-order wide dynamic range neurons and accompanies neuropathic pain after SCI. Thus, SCI-induced synaptic potentiation engages a putative spinal memory mechanism. A compelling, novel possibility for pain research is that a synaptic model of long-term memory storage could explain the persistent nature of neuropathic pain. Such a conceptual bridge between pain and memory could guide the development of more effective strategies for treatment of chronic pain after injury to the nervous system.


Experimental Neurology | 2011

Rac1-regulated dendritic spine remodeling contributes to neuropathic pain after peripheral nerve injury

Andrew M. Tan; Yu-Wen Chang; Peng Zhao; Bryan C. Hains; Stephen G. Waxman

Although prior studies have implicated maladaptive remodeling of dendritic spines on wide-dynamic range dorsal horn neurons as a contributor to pain after spinal cord injury, there have been no studies on dendritic spines after peripheral nerve injury. To determine whether dendritic spine remodeling contributes to neuronal hyperexcitability and neuropathic pain after peripheral nerve injury, we analyzed dendritic spine morphology and functional influence in lamina IV-V dorsal horn neurons after sham, chronic constriction injury (CCI) of the sciatic nerve, and CCI treatment with NSC23766, a selective inhibitor of Rac1, which has been implicated in dendritic spine development. 10 days after CCI, spine density increased with mature, mushroom-shaped spines preferentially distributed along dendritic branch regions closer to the cell body. Because spine morphology is strongly correlated with synaptic function and transmission, we recorded the response of single units to innocuous and noxious peripheral stimuli and performed behavioral assays for tactile allodynia and thermal hyperalgesia. Wide dynamic range dorsal horn neurons of CCI animals exhibited hyperexcitable responses to a range of stimuli. They also showed reduced nociceptive thresholds in the ipsilateral hind paw. 3-day treatment with NSC23766 significantly reduced post-CCI spine dimensions and densities, and attenuated injury-induced hyperexcitability. Drug treatment reduced behavioral measures of tactile allodynia, but not for thermal hyperalgesia. Together, our results demonstrate that peripheral nerve injury induces Rac1-regulated remodeling of dendritic spines on dorsal horn neurons, and suggest that this spine remodeling contributes to neuropathic pain.


Molecular Medicine | 2015

Virus-Mediated Knockdown of Nav1.3 in Dorsal Root Ganglia of STZ-Induced Diabetic Rats Alleviates Tactile Allodynia

Andrew M. Tan; Omar Abdel Samad; Sulayman D. Dib-Hajj; Stephen G. Waxman

Diabetic neuropathic pain affects a substantial number of people and represents a major public health problem. Available clinical treatments for diabetic neuropathic pain remain only partially effective and many of these treatments carry the burden of side effects or the risk of dependence. The misexpression of sodium channels within nociceptive neurons contributes to abnormal electrical activity associated with neuropathic pain. Voltage-gated sodium channel Nav1.3 produces tetrodotoxin-sensitive sodium currents with rapid repriming kinetics and has been shown to contribute to neuronal hyperexcitability and ectopic firing in injured neurons. Suppression of Nav1.3 activity can attenuate neuropathic pain induced by peripheral nerve injury. Previous studies have shown that expression of Nav1.3 is upregulated in dorsal root ganglion (DRG) neurons of diabetic rats that exhibit neuropathic pain. Here, we hypothesized that viral-mediated knockdown of Nav1.3 in painful diabetic neuropathy would reduce neuropathic pain. We used a validated recombinant adeno-associated virus (AAV)-shRNA-Nav1.3 vector to knockdown expression of Nav1.3, via a clinically applicable intrathecal injection method. Three weeks following vector administration, we observed a significant rate of transduction in DRGs of diabetic rats that concomitantly reduced neuronal excitability of dorsal horn neurons and reduced behavioral evidence of tactile allodynia. Taken together, these findings offer a novel gene therapy approach for addressing chronic diabetic neuropathic pain.


Journal of Neurophysiology | 2009

Dendritic spine remodeling after spinal cord injury alters neuronal signal processing.

Andrew M. Tan; Jin-Sung Choi; Stephen G. Waxman; Bryan C. Hains

Central sensitization, a prolonged hyperexcitability of dorsal horn nociceptive neurons, is a major contributor to abnormal pain processing after spinal cord injury (SCI). Dendritic spines are micron-sized dendrite protrusions that can regulate the efficacy of synaptic transmission. Here we used a computational approach to study whether changes in dendritic spine shape, density, and distribution can individually, or in combination, adversely modify the input-output function of a postsynaptic neuron to create a hyperexcitable neuronal state. The results demonstrate that a conversion from thin-shaped to more mature, mushroom-shaped spine structures results in enhanced synaptic transmission and fidelity, improved frequency-following ability, and reduced inhibitory gating effectiveness. Increasing the density and redistributing spines toward the soma results in a greater probability of action potential activation. Our results demonstrate that changes in dendritic spine morphology, documented in previous studies on spinal cord injury, contribute to the generation of pain following SCI.

Collaboration


Dive into the Andrew M. Tan's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jeffery D. Kocsis

United States Department of Veterans Affairs

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge