• Users Online: 1537
  • Home
  • Print this page
  • Email this page
Home About us Editorial board Ahead of print Current issue Search Archives Submit article Instructions Subscribe Contacts Login 

   Table of Contents      
REVIEW ARTICLE
Year : 2020  |  Volume : 6  |  Issue : 1  |  Page : 1-10

Role of vinpocetine in ischemic stroke and poststroke outcomes: A critical review


Department of Pharmacology, Toxicology and Medicine, College of Medicine, Almustansiriya University, Baghdad, Iraq

Date of Submission17-Oct-2019
Date of Decision06-Nov-2019
Date of Acceptance18-Nov-2019
Date of Web Publication18-Feb-2020

Correspondence Address:
Prof. Hayder M Al-Kuraishy
Department of Pharmacology, Toxicology and Medicine, College of Medicine, Almustansiriya University, P.O. Box: 14132, Baghdad
Iraq
Login to access the Email id

Source of Support: None, Conflict of Interest: None


DOI: 10.4103/bc.bc_46_19

Rights and Permissions
  Abstract 


Vinpocetine (VPN) is a synthetic ethyl-ester derivative of the alkaloid apovincamine from Vinca minor leaves. VPN is a selective inhibitor of phosphodiesterase type 1 (PDE1) that has potential neurological effects through inhibition of voltage-gated sodium channel and reduction of neuronal calcium influx. VPN has noteworthy antioxidant, anti-inflammatory, and anti-apoptotic effects with inhibitory effect on glial and astrocyte cells during and following ischemic stroke (IS). VPN is effective as adjuvant therapy in the management of epilepsy; it reduces seizure frequency by 50% in a dose of 2 mg/kg/day. VPN improves psychomotor performances through modulation of brain monoamine pathway mainly on dopamine and serotonin, which play an integral role in attenuation of depressive symptoms. VPN recover cognitive functions and spatial memory through inhibition of hippocampal and cortical PDE1 with augmentation of cyclic adenosin monophosphate and cyclic guanosin monophosphate ratio, enhancement of cholinergic neurotransmission, and inhibition of neuronal inflammatory mediators. Therefore, VPN is an effective agent in the management of IS and plays an integral role in the prevention and attenuation of poststroke epilepsy, depression, and cognitive deficit through direct cAMP/cGMP-dependent pathway or indirectly through anti-inflammatory and antioxidant effects.

Keywords: Anti-inflammatory, antioxidant, phosphodiestrase type 1, poststroke, stroke, vinpocetine


How to cite this article:
Al-Kuraishy HM, Al-Gareeb AI, Naji MT, Al-Mamorry F. Role of vinpocetine in ischemic stroke and poststroke outcomes: A critical review. Brain Circ 2020;6:1-10

How to cite this URL:
Al-Kuraishy HM, Al-Gareeb AI, Naji MT, Al-Mamorry F. Role of vinpocetine in ischemic stroke and poststroke outcomes: A critical review. Brain Circ [serial online] 2020 [cited 2023 Jun 3];6:1-10. Available from: http://www.braincirculation.org/text.asp?2020/6/1/1/278541




  Introduction Top


Vinpocetine (VPN) is a synthetic ethyl-ester derivative of the alkaloid apovincamine from Vinca minor leaves known as lesser periwinkle. A VPN has a specific chemical structure contains carboxylic acid ethyl ester which is soluble in alcohol, acetone, and sulfoxide [Figure 1].[1]
Figure 1: Chemical structure of vinpocetine

Click here to view


VPN is widely used in the treatment of different cerebrovascular disorders, cognitive dysfunction, memory disorders, tinnitus, macular degeneration, and glaucoma. In addition, VPN is effective in the management of acute kidney injury, renal stone, hair loss, and peptic ulceration.[2]

Nevertheless, this critical review only focused on the potential role of VPN in the management of ischemic stroke (IS).

A multiplicity of search strategies was taken and assumed which included electronic database searches of Medline and PubMed using MeSH terms, keywords, and title words during the search. The terms used for these searches were as follows: (VPN OR apovincamine) AND (cognitive function OR stroke OR brain ischemia OR blood flow OR cerebral circulation OR oxidative stress OR blood viscosity OR cerebral blood flow). (VPN OR apovincamine) AND (cerebral metabolism OR cerebral hypoxia OR ischemic degeneration OR minor stroke). Reference lists of identified and notorious articles were reviewed. In addition, only English articles were considered and case reports were not concerned in the review. The key features of recognized applicable search studies were considered and the conclusions summarized in a critical review.


  Pharmacology of Vinpocetine Top


VPN is a selective inhibitor of phosphodiesterase type 1 (PDE1), which increasing cAMP and cGMP leading to vasodilatation. Furthermore, it inhibits the release of pro-inflammatory cytokines through inhibition of I kappa B kinase/nuclear factor kappa (IKK/NF-κB) activator protein-1 pathway which is involved in the propagation of inflammatory cytokines translocation and release.[3] VPN has potential neurological effects through inhibition of voltage-gated sodium channel, reduction of neuronal calcium influx, and antioxidant effect with augmentation of dopamine metabolism, since it increases 3, 4-dihydroxyphenylacetic acid which is the breakdown metabolites of dopamine.[4]

It has been reported that VPN is a safe drug for long-term use and it well tolerated during management of cerebrovascular disorders. Mild side effects such as headache, flushing, anxiety, dry mouth, and nausea have been accounted during VPN uses. In spite of potent nonselective vasodilator effect, it does not produce stealing effect on the cerebral vasculatures due to the viscosity lowering effect and inhibition of platelet aggregations which together improve cerebral vessels rheological properties. Nevertheless, VPN does not reduce blood pressure and systemic circulation during acute and chronic uses.[5]

VPN is well absorbed from the small intestine, which increased by food. Therefore, fasting bioavailability is 6.7% and nonfasting bioavailability is 60%–100%. Similarly, VPN has no significant drug–drug interactions with different drugs such as oxazepam, imipramine, glibenclamide, and other agents that are used in the management of IS.[6]


  Vinpocetine in Ischemic Stroke Top


IS represents the main leading cause of death in the United States of America and developed countries and regarded it as the main cause of long-term disability. IS represents 11.9% of annual total death and accounts for 90% of all stroke cases.[7] Arboix study discussed briefly the risk factors of IS.[8] These risk factors are divided into nonmodifiable risk factors (sex, age, inherited factors, ethnicity, and low birth weight at birth) and modifiable risk factors (diabetes mellitus, hypertension, smoking, obesity, alcohol abuse, oral contraceptive, and metabolic syndrome).[9] IS is mainly caused by arterial thrombosis on the atherosclerotic plaque of cerebral vessels, causing cerebral ischemia, infarction, and induction of peri-infarct inflammation. Neuroinflammations contribute to tissue repair and neuronal damage as well as retrograde and anterograde axonal degenerations.[10] IS leads to glucose and oxygen deprivation of neuronal cells also which, causes oxidative stress, excitotoxicity, and calcium overload that eventually causing neuronal cell death and development of infarction core.[11] The infracted core and damaged neuronal cells due to induced oxidative stress, release various inflammatory molecules that causing vasculitis and damage of blood–brain barrier (BBB).[12] Moreover, activated microglia and infiltrated macrophage during IS release different neurotransmitters and interact with neurons causing neuroinflammation and neuronal injury. As well, interleukin-8 (IL-8), NF-κB, and tumor necrosis factor (TNF-α) are overexpressed during IS which plays a potential role in the initiation of inflammation and apoptosis.[13] In a similar way, vascular smooth muscle and endothelial cells of cerebral vasculature are activated by NF-κB pathway leading to further obstruction and thrombosis. Therefore, NF-κB pathway is an important pathway in the pathogeneses and development of neurological deficit thus, inhibition of the NF-κB pathway by VPN is regarded as important and main mechanism of VPN neuroprotection.[14]

In addition, activated microglia expresses cholesterol transporter protein, which is overexpressed during brain injury and IS and inhibited by VPN.[15]

During IS, voltage-gated sodium channels are activated causing intracellular accumulation of Na and Ca leading to neuronal cell damage, excitotoxicity, edema, acidosis, and acute cellular dysfunctions. VPN inhibits voltage-gated sodium channels leading to dose-dependent reduction of intracellular concentrations of Na and Ca. Thus, the neuroprotective effect of VPN during IS is chiefly mediated by inhibition of neuronal voltage-gated sensitive Na-channel.[16]

Different studies illustrated that oxidative stress, excitotoxicity, and impaired energy metabolism leading to neuronal death by both apoptosis and necrosis during IS. These events lead to reduction of cAMP system which is important in the expression and regulation of brain-derived neurotrophic factor (BDNF), which improves neuronal survival. PDE1 is mainly localized in striatum and cortex which participating in the regulation of neuronal motor activity.[17],[18]

Indeed, VPN increases neuronal cGMP through inhibition of calmodulin-dependent phosphodiesterase which improves cerebral blood flow and oxygen consumption.[19] VPN improves cerebral metabolism through enhancing glucose and oxygen supply and ATP production by cerebral vasodilation. These effects prevent IS induced-memory and cognitive dysfunctions due to improvement of neurotransmitters such as serotonin, dopamine, and noradrenaline, which are involved in the regulation of cognitive function.[20]

Systemic review by Bereczki and Fekete showed that VPN decreases the size of cerebral infarction after middle cerebral artery occlusion in rats and mice. In controlled human studies, VPN increases cerebral perfusion and oxygen extraction and prevents the worsening of attention in patients with multiple cerebral infarcts so; VPN has been used to treat stroke in several countries in Europe. Therefore, there is no evidence to support the routine use of VPN in all patients with acute IS. Further trials are needed to decide if the routine application of VPN decreases case fatality and the proportion of dependent survivors in acute IS.[21] Besides, a previous clinical study involved 92 patients with acute IS treated with VPN and/or ganglioside illustrated that this combination can promote the neural functional reconstruction and inhibit the occurrence of cerebral hemorrhage and cerebral re-infarction in convalescents with acute cerebral infarction.[22] Therefore, these studies confirm the valuable role of VPN alone or in combination with other neuroprotective agents in acute IS.

Antioxidant effects of vinpocetine in ischemic stroke

In IS, overproduction of free radicals and reactive oxygen and/or nitrogen species leads to neuro-pathological changes through complex interactions with cellular components such as proteins, DNA, and lipids. Free radicals, mainly superoxide and nonradicals such as hydrogen peroxide, may cause further neurological injury through depletion of endogenous antioxidant capacity. Therefore, drug with antioxidant potential may play a role in the prevention of cerebral injury during IS.[23]

A recent study by Al-Kuraishy et al. reported that VPN is a potent antioxidant agent improves antioxidant capacity and reduces oxidative stress.[24] As well, Santos et al. study illustrated that VPN attenuates oxidative stress during IS through inhibition of lipid peroxidation and generation of free radical.[25] In addition, VPN has a neuroprotective effect, through the antioxidant effect since it prevents oxidative stress injury and toxic demyelination in rat brain.[26] The antioxidant neuroprotective effect of VPN is mainly at low-moderate doses since high doses of VPN lead to oxidative stress due to pro-oxidant and pro-inflammatory effects.[27] Deshmukh et al. reported that antioxidant potential of VPN contributes to the prevention of IS induced-neuronal injury through modulation of cholinergic neurons.[28] Therefore, antioxidant mechanisms of VPN are related to direct free radical scavenging effect, potentiating endogenous antioxidant capacity and inhibition the generation of free radicals. The molecular antioxidant effect of VPN is linked to the suppression of ADP stimulated respiration, mitochondrial Na+/Ca+ exchange, mitochondrial swelling, and regulation of mitochondrial membrane potentials.[29],[30]

Anti-inflammatory effects of vinpocetine in ischemic stroke

IS-induced inflammatory changes and neuroinflammations lead to secondary brain damage. Toll-like receptors (TLRs) are overexpressed in IS, leading to the induction of the release of pro-inflammatory mediators through myeloid differentiation factor-88 (MyD88)-dependent pathway and Toll/IL-IR domain-containing adaptor factor protein inducing interferon-beta (TRIF)-dependent pathway.[31] Therefore, inhibition of TLR4/MyD88 and NF-KB pathways lead to noteworthy neuroprotection against IS. It has been noted that VPN inhibits TNF-α induced NF-κB activation, pro-inflammatory releases, and inflammatory biomarkers such as IL-1β and IL-33 in an experimental ischemic model.[32] In intriguing way, Zhang and Yang reported that VPN inhibits the release of chemokines and inflammatory cytokines from microglia, macrophage, and endothelial cells in IS through inhibition of NF-κB pathways in IS and associated atherosclerosis.[33] In a similar way, VNP leads to the significant neuroprotective effect and regulation of neuronal plasticity through the anti-inflammatory effect via suppression of IKK pathway in IS [Figure 2].[34],[35]
Figure 2: Anti-inflammatory effects of vinpocetine, (a) PDEI-dependent

Click here to view


Usually, microglia cells are resident macrophages in the brain and act as an active immune defense against cerebral injury and infection through induction and regulation of neuroinflammatory reactions. Microglia improves brain homeostasis by removal of tissue debris, dead cells, and induction of neurogenesis and preservation of myelin sheath with secretion of neuroprotective factors such as insulin-like growth factor. On the other hand, activated microglia leads to neuronal injury during IS through the release of TNF-α, IL-6, IL1β, and nitric oxide (NO).[36] It has been reported that VPN inhibits neuronal inflammation in IS via suppression of microglia activity.[37] Furthermore, VPN inhibits IS-induced inflammatory changes and reduces brain edema and infarction size mainly through inhibition the expression of NF-κB and TNF-α in the activated microglia which is PDE1-independent pathway.[38] Animal model studies showed that after 4-6 h of ischemia, circulating leukocytes adhere to the vessel walls, leading to migration and accumulation in the ischemic brain lesion, which results in secondary injury. Neutrophils are highly associated with the neuronal damage so, inhibiting the adhesion molecules that facilitate neutrophil entry into the injured brain will improves neurological outcomes. This can be accomplished by VPN's inhibition of IKK/NF-κB in endothelial cells and macrophages. The protective effect seen in lymphocyte-deficient mice, or caused by blocking postischemic trafficking of T-cells into the ischemic brain, occurs 24–48 h after ischemia.[39],[40] As a consequence, VPN acts as an anti-inflammatory agent by ameliorating cerebral ischemia/reperfusion injuryin vitro and in vivo. VPN inhibits inflammatory responses through the TLR4/MyD88/NF-κB signaling pathway that are independent of TRIF-mediated inflammatory responses. As a result, VPN may be an attractive therapeutic candidate for the treatment of cerebral ischemic injuries and inflammatory diseases.

Effects of vinpocetine on ischemic reperfusion injury in ischemic stroke

Ischemic-reperfusion (I/R) injury in IS leads to activations of perivascular macrophages, which play a role in the progression of neuronal damage through the release of pro-inflammatory biomarkers which also participate in the injury to BBB. Furthermore, activate macrophages, microglia, T-cells, and dendritic cells; infiltrate the infarct site following I/R-injury causing further damage through the release of monocyte chemoattracting protein-1 which attracts circulating neutrophils into the injury site. VPN inhibits TNF-α induced-IKKα/β activation with reduction of target genes activations and reduction of various forms of pro-inflammatory cytokines and mediator following I/R injury in IS [Figure 3].[40],[41]
Figure 3: Effects of vinpocetine on pro-inflammatory mediators during ischemic-reperfusion injury in ischemic stroke

Click here to view


In addition, injured neurons in IS release specific proteins called danger-associated molecular patterns including heat shock protein, high mobility group-box 1 protein, adenosin triphosphate (ATP), and nicotinamide adenine dinucleotide which activate TLR4 receptors on perivascular macrophage, microglia, and endothelial cells. Therefore, TLR4 antagonist reduces the infarct size, attenuates IS-induced inflammatory changes, and I/R injury.[42],[43] Differentin vitro andin vivo studies illustrated that VPN inhibits I/R injury in IS through suppression of TLR4 receptors and NF-κB signaling pathway in animal model studies.[44]

Neuronal mitochondrial reactive oxygen species contribute to the pathogenesis of I/R injury in IS as well as neurodegeneration and glutamate excitotoxicity.[45] VPN activates peripheral benzodiazepine receptors which regulate mitochondrial outer cell membrane and prevent the opening of mitochondrial permeability transition pore (MPTP). Furthermore, VPN prevents mitochondrial dysfunction through prevention of mitochondrial depolarization, inhibition of mitochondrial Na+/Ca2+ exchange, anticipation of mitochondrial Ca2+ release, MPTP opening, and the release of free radicals from outer mitochondrial membrane during neuronal injury.[46] Furthermore, VPN regulates mitochondrial redox homeostasis through induction of ATP hydrolysis, inhibition of mitochondrial respiration and regulation of ATP synthesis. As a result, VPN preserves mitochondrial integrity and attenuates inflammatory and oxidative damage following I/R injury in IS. Moreover, Qiu et al. illustrated that VPN is effective in reducing the volume of cerebral infarct and attenuation I/R injury through downregulation of NF-κB p65 and cyclo-oxygenase 2 with upregulation of neuroprotective mediator called peroxisome proliferator-activator receptor γ during IS.[47]

Vinpocetine and postischemic stroke

Immunological and inflammatory reactions in postischemic stroke

In the brain, there are multiple communications between the glial cell and other immune cells, which together participate in the immune reactions during ischemic events. In the post-IS (PIS), B-cell, T-cell, macrophage, and neutrophils enter the brain to connect and engage glial cells in immune interactions. This interaction maintains homeostasis and prevents further neuronal damage through generation of pro-survival factors such as transforming growth factor-β and IL-10 which promote the resolution of inflammations.[48]

It has been noticed that IS activates neuroinflammations which increase the permeability of BBB due to activation of mast cells and macrophages which release histamine and pro-inflammatory cytokines, respectively. These mediators recruit immune cells to the site of injury leading to the progression of ischemic injury.[49] Therefore, the relationship between immune cells and neurons during IS is such an intricate relationship.

Microglia is regarded as a first-line defense mechanism of innate immunity against ischemic injury which activated within hours following IS. There are two activation pathways for microglia, which are the classical pathway (M1) and alternative pathway (M2). M1 activation leads to induction of inducible NO synthase and TNF-α causing neuronal damage, while M2 activation leads to induction the release of pro-inflammatory cytokines and arginase leading to neuroprotection.[50] Aging is associated with impaired M2 activation and thus; M1 activation overriding M2 causing more inflammatory changes in elderly patients with IS.[51]

Similarly, astrocyte which is another type of glial cell contributes to the formation of BBB and is activated following IS. Reactive astrocyte subdivided into A1 which plays a role in the neuronal damage through upregulation of complement genes, and A2 which plays a role in the neuroprotection through upregulation of neurotrophic factors.[52] One month following IS, astrocyte undergoes morphological and functional changes leading to reactive gliosis and activation of T-cell at ischemic regions.[53]

Therefore, astrocyte and glial cells act as bridge for interaction between neurons and immune system through different pro-inflammatory cytokines [Figure 4].[54] It has been shown that inflammatory changes, glial and astrocyte activations at poststroke period participating together in the induction of different poststroke complications such as depression, epilepsy, dementia, and cognitive dysfunctions.[55] Vardian study illustrated that VPN has noteworthy antioxidant, anti-inflammatory, and antiapoptotic effects with inhibitory effect on glial and astrocyte cells during and following IS. Furthermore, VPN reduces astrocyte edema and excitability through cAMP-dependent PKA pathway.[56]
Figure 4: Microglial and astrocyte activations in postischemic stroke.CCR2: chemokine receptor 2, PAMPs: pathogen-associated molecular patterns, LPS: lipopolysaccharides, PD-1: programmed death-ligand 1, NK: natural killer, CD: cluster of differentiation

Click here to view


As well, experimental studies observed that VPN inhibits lipopolysaccharides-induced NF-κB activation and TNF-α, IL-1β, and IL-33 production by macrophages. When these cytokines activate endothelial cells, they upregulate the expression of adhesion molecules promoting neutrophil recruitment to the infracted tissues. Hence, VPN reduces neutrophil recruitment and the activity of myeloperoxidase in mice.[40] Therefore, VPN is an effective agent in the prevention of CNS inflammation following acute IS and can be used to attenuate poststroke inflammatory reactions.[57]

Vinpocetin for postischemic stroke epilepsy

Kim et al. reported that PIS predisposes for early- and late-onset epilepsy which called poststroke seizure (PSS) due to the disturbances in the neuronal metabolic homeostasis, reactive gliosis, glutamate release, and neuronal hyperexcitability.[58] Recently, Garza-Morales et al. found that VPN is effective as adjuvant therapy in the management of epilepsy, it reduces seizure frequency by 50% in a dose of 2 mg/kg/day as compared with placebo.[59] The antiepileptic mechanisms of VPN are through blockade of presynaptic Na-channel-mediated glutamate release, inhibition of TNF-α and IL-1 β which play a role in the augmentation of presynaptic Ca and Na permeability.[60],[61] The site of stroke lesions has been reported as a chief clinical factor enduring tendency to generate further and additional seizures. Several studies have established that early PSS is an independent risk factor for the development of late and repeated seizures.[62] Early seizure control is important because uncontrolled repetitive seizures may affect patients not only through potential physical injuries but also throughout the harmful effects on the brain with stroke lesion and degenerative changes. It is also probable that recurrent peri-infarct depolarization might be injurious to already susceptible tissues because the additional metabolic stress could cause further neuronal injuries.[63],[64] Even though, seizures after stroke are known to be related to increased resource utilization and length of hospital stay as well as decreased survival at 30-day and 1-year time points.[65] In view of lower volume of cortical gray matter and decreased excitability due to degenerative changes, it is easy to assume that younger patients might develop seizures more often.[66] Herein, VPN may be used to control PSS alone or in combination with other antiseizure medications through improvement of the neuronal plasticity and reduction of poststroke inflammation and oxidative stress-induced neuronal excitability.[67]

Vinpocetin for poststroke depression

Poststroke depression (PSD) is a critical psychiatric complication of IS characterized by psychomotor disturbances, fatigue, and sleep disorders with the prevalence of 33% following IS.[68] PSD is developed due to inflammatory reaction-induced neuroplasticity and imbalance of pro-inflammatory/anti-inflammatory ratio which causing glutamate excitotoxicity and intracellular Ca dysregulation.[69] Different studies illustrated that inflammatory cytokines induced-PSD lead to a reduction in the synthesis of serotonin, BDNF, and fibroblast growth factor-2 (FGF-2), which are important in the regulation of mood and neurotransmission.[70],[71]

Inflammatory cytokines are implicated in the induction of PSD through activation of indolamine-2,3-dioxygenase at the marginal zone of the infracted area leading to depletion of serotonin and initiation of depression.[72] Furthermore, different clinical studies revealed a relationship between different cytokines and molecules with PSD, Wiener et al. found that nerve growth factor (NGF) which important secretory protein inhibits apoptosis and improves neuronal differentiations was low in PSD.[73] On the other hand, calcitonin gene-related peptide (CGRP) which is a neuroprotective peptide is elevated in patients with PSD and thus CGRP antagonist could improve depressive symptoms.[66] Similarly, reduced dopamine concentrations in ischemic striatum have been demonstrated in a mouse model of chronic PSD.[74] In addition to monoamine neurotransmitters, a low plasma glutamate has also been reported to be associated with early-onset PSD. This study found that the levels of IL-6 and TNF-α significantly higher in the PSD group than in the non-PSD group. Thus, inflammatory cytokines are implicated in the pathogenesis of PSD.[75]

Therefore, anti-inflammatory drugs with rehabilitation therapy enhance neuronal plasticity and functional recovery after IS.[76] VPN reduces the inflammatory processes and improves neuronal plasticity through inhibition of the releases of inflammatory cytokines and chemokines from macrophage, microglia, and vascular smooth and endothelial cells with restoration of synaptic neurotransmissions.[77] As well, VPN improves psychomotor performances through modulation of brain monoamine pathway mainly on dopamine and serotonin, which play an integral role in attenuation of depressive symptoms.[78] Chen et al. reported that VPN improves neuronal functions and neurotransmission through modulation of NGF levels following IS.[22] Similarly, VPN improves neuronal transmission and inhibits induced pain pathways in PSD through downregulation of CGRP.[79] Herewith, VPN attenuates PSD through different pathways either directly by activation of neuronal cAMP/cGMP pathway or indirectly through antioxidant, anti-inflammatory, and modulation of brain peptides and neurotransmitters. Since hippocampal cAMP-PKA response element of BDNF signaling pathway is decreased in patients with PSD. Hence, improvement of neuronal cAMP could interestingly prevent PSD.[80]

Vinpocetin for poststroke cognitive deficit

Poststroke cognitive deficit (PSCD) is defined as a global cognitive disability within 6 months after stroke regardless of presumptive causes according to the American Psychiatric Associations Diagnostic and Statistical Manual of Mental Disorder. As well, 30% of stroke survivors found to have a noteworthy degree of cognitive decline within the 1st month after the stroke.[81] It has been noticed that some cognitive disorders may also develop subsequent to transient ischemic attack suggesting that PSCD used in this way does not propose underlying neuropathological changes. Therefore, PSCD seems to be suitable for dementia, which associated with vascular insult and neurodegenerative processes.[82] Various cross-sectional and longitudinal studies illustrated a link between high levels of inflammatory biomarkers in stroke survivors and risk of PSCD. Erythrocyte sedimentation rate (ESR), C-reactive protein, IL-12, and IL-6 sera levels are elevated in patients with PSCD and regarded as predictor factors.[83],[84]

The inflammatory mechanism of PSCD is related to the dysregulation in inflammatory and immune factors since reduction of IL-8 and IL-6 is associated with changes in both white and gray matters, suggesting a role in the pathogenesis of PSCD. As well, IL-1, IL-10, TNF-α, and α-synuclein are increased in PSCD.[85] Shen and Gao study reported that high somatostatin and low neuron-specific enolase in patients with PSCD compared to the healthy controls.[86]

Other mechanisms of PSCD are cerebral hypoperfusion, reduction in the cerebrovascular reserve capacity, impairment of cerebral vasoreactivity and autoregulatory ability, which together initiate abnormal neuronal cell membrane phosphorylation and amyloid-beta formation.[87] In addition, irreversibly injured astrocytes are converted to clasmatodendrosis which leads to disruption of gliovascular association at BBB in the white matter. Clasmatodendrosis is associated with cognitive disorders in patients with PSCD.[88] From these points, the mechanisms of PSCD remain obscure due to overlapping between neuropathological data and findings of PSCD and Alzheimer's disease.[89] VPN improves cognitive functions and spatial memory through inhibition of hippocampal and cortical PDE-1 with augmentation of cAMP/cGMP ratio, enhancement of cholinergic neurotransmission, and inhibition of neuronal IKK/NF-κB.[90],[91] It has been perceived by Bitner study that both cAMP and cGMP activate PKA-response element-binding protein (CREP) improves synaptic plasticity and neurogenesis through upregulation of BDNF. cAMP/cGMP/CREP pathway increases early and late long-term potentiation of memory.[92] Besides, other PDE inhibitors such as sildenafil (PDE-5 inhibitors) and cilotazol (PDE-3 inhibitor) also improve cognitive function and PSCD.[93] Recently, McQuown et al. illustrated that VPN improves memory function mainly through inhibition of PDE-1B isoform, as it mainly located in regions with high dopaminergic neurotransmission such as the prefrontal cortex, striatum, and dentate gyrus.[90] Therefore, VPN is an effective therapy in rehabilitation of cognitive, memory deficit, and PSCD through modulation of inflammatory changes and enhancement of neuronal cAMP/cGMP in poststroke survivors.[94]


  Conclusions Top


Animal model, preclinical, and clinical studies confirmed that VPN is an effective agent in the management of IS and early poststroke complications. VPN plays an integral role in the prevention and attenuation of late poststroke complications such as epilepsy, depression, and cognitive deficit through direct cAMP/cGMP-dependent pathway or indirectly through anti-inflammatory and antioxidant effects. Further studies are recommended to observe and confirm the specific effect of VPN on stroke outcomes and complications.

Acknowledgments

We would like to acknowledge all staff members in the Department of clinical pharmacology and Therapeutics for their great support.

Financial support and sponsorship

Nil.

Conflicts of interest

There are no conflicts of interest.



 
  References Top

1.
Alkuraishy HM, Al-Gareeb AI, Albuhadilly AK. Vinpocetine and pyritinol: A new model for blood rheological modulation in cerebrovascular disorders – A randomized controlled clinical study. Biomed Res Int 2014;2014:324307.  Back to cited text no. 1
    
2.
El-Laithy HM, Shoukry O, Mahran LG. Novel sugar esters proniosomes for transdermal delivery of vinpocetine: Preclinical and clinical studies. Eur J Pharm Biopharm 2011;77:43-55.  Back to cited text no. 2
    
3.
Cao B, Ding Q, Liu X, Liu C, Songhua HU. Clinical observation of soften injection combined with vinpocetine injection in the treatment of acute massive cerebral infarction. China Pharm 2017;28:4527-9.  Back to cited text no. 3
    
4.
Nadeem RI. Evaluation of the Possible Neurobehavioral Effects of Vinpocetine in Parkinsonian-Like Models in Rats CU These; 2018.  Back to cited text no. 4
    
5.
Zhang F, Yan C, Wei C, Yao Y, Ma X, Gong Z, et al. Vinpocetine inhibits NF-κB-dependent inflammation in acute ischemic stroke patients. Transl Stroke Res 2018;9:174-84.  Back to cited text no. 5
    
6.
Manda VK, Avula B, Dale OR, Chittiboyina AG, Khan IA, Walker LA, et al. Studies on pharmacokinetic drug interaction potential of vinpocetine. Medicines (Basel) 2015;2:93-105.  Back to cited text no. 6
    
7.
Cerami C, Perani D. Imaging neuroinflammation in ischemic stroke and in the atherosclerotic vascular disease. Curr Vasc Pharmacol 2015;13:218-22.  Back to cited text no. 7
    
8.
Arboix A. Cardiovascular risk factors for acute stroke: Risk profiles in the different subtypes of ischemic stroke. World J Clin Cases 2015;3:418-29.  Back to cited text no. 8
    
9.
Go AS, Mozaffarian D, Roger VL, Benjamin EJ, Berry JD, Borden WB, et al. Executive summary: Heart disease and stroke statistics-2013 update: A report from the American Heart Association. Circulation 2013;127:143-52.  Back to cited text no. 9
    
10.
Macrez R, Ali C, Toutirais O, Le Mauff B, Defer G, Dirnagl U, et al. Stroke and the immune system: from pathophysiology to new therapeutic strategies. Lancet Neurol 2011;10:471-80.  Back to cited text no. 10
    
11.
Sidorov E, Sanghera DK, Vanamala JKP. Biomarker for ischemic stroke using metabolome: A clinician perspective. J Stroke 2019;21:31-41.  Back to cited text no. 11
    
12.
Yang C, Hawkins KE, Doré S, Candelario-Jalil E. Neuroinflammatory mechanisms of blood-brain barrier damage in ischemic stroke. Am J Physiol Cell Physiol 2019;316:C135-C153.  Back to cited text no. 12
    
13.
Zarruk JG, Greenhalgh AD, David S. Microglia and macrophages differ in their inflammatory profile after permanent brain ischemia. Exp Neurol 2018;301:120-32.  Back to cited text no. 13
    
14.
Wu LR, Liu L, Xiong XY, Zhang Q, Wang FX, Gong CX, et al. Vinpocetine alleviate cerebral ischemia/reperfusion injury by down-regulating TLR4/MyD88/NF-κB signaling. Oncotarget 2017;8:80315-24.  Back to cited text no. 14
    
15.
Fujita M, Imaizumi M, Zoghbi SS, Fujimura Y, Farris AG, Suhara T, et al. Kinetic analysis in healthy humans of a novel positron emission tomography radioligand to image the peripheral benzodiazepine receptor, a potential biomarker for inflammation. Neuroimage 2008;40:43-52.  Back to cited text no. 15
    
16.
Svab G, Doczi J, Gerencser AA, Ambrus A, Gallyas F, Sümegi B, et al. The mitochondrial targets of neuroprotective drug vinpocetine on primary neuron cultures, brain capillary endothelial cells, synaptosomes, and brain mitochondria. Neurochem Res 2019;44:2435-47.  Back to cited text no. 16
    
17.
Patyar S, Prakash A, Modi M, Medhi B. Role of vinpocetine in cerebrovascular diseases. Pharmacol Rep 2011;63:618-28.  Back to cited text no. 17
    
18.
Ahmed HI, Abdel-Sattar SA, Zaky HS. Vinpocetine halts ketamine-induced schizophrenia-like deficits in rats: Impact on BDNF and GSK-3β/β-catenin pathway. Naunyn Schmiedebergs Arch Pharmacol 2018;391:1327-38.  Back to cited text no. 18
    
19.
Zhang W, Huang Y, Li Y, Tan L, Nao J, Hu H, et al. efficacy and safety of vinpocetine as part of treatment for acute cerebral infarction: A randomized, open-label, controlled, multicenter CAVIN (Chinese assessment for vinpocetine in neurology) Trial. Clin Drug Investig 2016;36:697-704.  Back to cited text no. 19
    
20.
Jack C. Interventions that may increase cerebral blood flow. In: Alzheimer's Turning Point. Cham: Springer; 2016. p. 217-28.  Back to cited text no. 20
    
21.
Bereczki D, Fekete I. Vinpocetine for acute ischemic stroke. Stroke. 2008;39:2404-5.  Back to cited text no. 21
    
22.
Chen Q, Li GQ, Li JT. Effect of ganglioside combined with vinpocetine therapy on neural functional reconstruction in convalescents with acute cerebral infarction. J Hainan Med Univ 2016;22:27-30.  Back to cited text no. 22
    
23.
Slemmer JE, Shacka JJ, Sweeney MI, Weber JT. Antioxidants and free radical scavengers for the treatment of stroke, traumatic brain injury and aging. Curr Med Chem 2008;15:404-14.  Back to cited text no. 23
    
24.
Al-Kuraishy HM, Al-Gareeb AI, Al-Nami MS. Vinpocetine improves oxidative stress and pro-inflammatory mediators in acute kidney injury. Int J Prev Med 2019;10:142.  Back to cited text no. 24
[PUBMED]  [Full text]  
25.
Santos MS, Duarte AI, Moreira PI, Oliveira CR. Synaptosomal response to oxidative stress: effect of vinpocetine. Free Radic Res 2000;32:57-66.  Back to cited text no. 25
    
26.
Abdel-Salam OM, Khadrawy YA, Salem NA, Sleem AA. Oxidative stress in a model of toxic demyelination in rat brain: The effect of piracetam and vinpocetine. Neurochem Res 2011;36:1062-72.  Back to cited text no. 26
    
27.
Abdel-Salam OM, Hamdy SM, Seadawy SA, Galal AF, Abouelfadl DM, Atrees SS. Effect of piracetam, vincamine, vinpocetine, and donepezil on oxidative stress and neurodegeneration induced by aluminum chloride in rats. Comp Clin Pathol 2016;25:305-18.  Back to cited text no. 27
    
28.
Deshmukh R, Sharma V, Mehan S, Sharma N, Bedi KL. Amelioration of intracerebroventricular streptozotocin induced cognitive dysfunction and oxidative stress by vinpocetine – A PDE1 inhibitor. Eur J Pharmacol 2009;620:49-56.  Back to cited text no. 28
    
29.
Ishola IO, Akinyede AA, Adeluwa TP, Micah C. Novel action of vinpocetine in the prevention of paraquat-induced parkinsonism in mice: Involvement of oxidative stress and neuroinflammation. Metab Brain Dis 2018;33:1493-500.  Back to cited text no. 29
    
30.
Svab G, Doczi J, Gerencser AA, Ambrus A, Gallyas F, Sümegi B, et al. The mitochondrial targets of neuroprotective drug vinpocetine on primary neuron cultures, brain capillary endothelial cells, synaptosomes, and brain mitochondria. Neurochem Res 2019;44:2435-47.  Back to cited text no. 30
    
31.
Gao W, Xiong Y, Li Q, Yang H. Inhibition of toll-like receptor signaling as a promising therapy for inflammatory diseases: A journey from molecular to nano therapeutics. Front Physiol 2017;8:508.  Back to cited text no. 31
    
32.
Zhang F, Yan C, Wei C, Yao Y, Ma X, Gong Z, et al. Vinpocetine inhibits NF-κB-dependent inflammation in acute ischemic stroke patients. Transl Stroke Res 2018;9:174-84.  Back to cited text no. 32
    
33.
Zhang L, Yang L. Anti-inflammatory effects of vinpocetine in atherosclerosis and ischemic stroke: A review of the literature. Molecules 2014;20:335-47.  Back to cited text no. 33
    
34.
Cohen PA. Vinpocetine: An unapproved drug sold as a dietary supplement. Mayo Clin Proc 2015;90:1455.  Back to cited text no. 34
    
35.
Medina AE. Vinpocetine as a potent antiinflammatory agent. Proc Natl Acad Sci U S A 2010;107:9921-2.  Back to cited text no. 35
    
36.
Ma Y, Wang J, Wang Y, Yang GY. The biphasic function of microglia in ischemic stroke. Prog Neurobiol 2017;157:247-72.  Back to cited text no. 36
    
37.
Zhao YY, Yu JZ, Li QY, Ma CG, Lu CZ, Xiao BG. TSPO-specific ligand vinpocetine exerts a neuroprotective effect by suppressing microglial inflammation. Neuron Glia Biol 2011;7:187-97.  Back to cited text no. 37
    
38.
Wang H, Zhang K, Zhao L, Tang J, Gao L, Wei Z. Anti-inflammatory effects of vinpocetine on the functional expression of nuclear factor-kappa B and tumor necrosis factor-alpha in a rat model of cerebral ischemia-reperfusion injury. Neurosci Lett 2014;566:247-51.  Back to cited text no. 38
    
39.
Zheng Z, Yenari MA. Post-ischemic inflammation: Molecular mechanisms and therapeutic implications. Neurol Res 2004;26:884-92.  Back to cited text no. 39
    
40.
Jeon KI, Xu X, Aizawa T, Lim JH, Jono H, Kwon DS, et al. Vinpocetine inhibits NF-kappaB-dependent inflammation via an IKK-dependent but PDE-independent mechanism. Proc Natl Acad Sci U S A 2010;107:9795-800.  Back to cited text no. 40
    
41.
Nivison-Smith L, Khoo P, Acosta ML, Kalloniatis M. Pre-treatment with vinpocetine protects against retinal ischemia. Exp Eye Res 2017;154:126-38.  Back to cited text no. 41
    
42.
Abdel-Rahman EA, Mahmoud AM, Aaliya A, Radwan Y, Yasseen B, Al-Okda A, et al. Resolving contributions of oxygen-consuming and ROS-generating enzymes at the synapse. Oxid Med Cell Longev 2016;2016:1089364.  Back to cited text no. 42
    
43.
Valencia A, Sapp E, Kimm JS, McClory H, Reeves PB, Alexander J, et al. Elevated NADPH oxidase activity contributes to oxidative stress and cell death in Huntington's disease. Hum Mol Genet 2013;22:1112-31.  Back to cited text no. 43
    
44.
Essam RM, Ahmed LA, Abdelsalam RM, El-Khatib AS. Phosphodiestrase-1 and 4 inhibitors ameliorate liver fibrosis in rats: Modulation of cAMP/CREB/TLR4 inflammatory and fibrogenic pathways. Life Sci 2019;222:245-54.  Back to cited text no. 44
    
45.
Colombo BB, Fattori V, Guazelli CFS, Zaninelli TH, Carvalho TT, Ferraz CR, et al. Vinpocetine ameliorates acetic acid-induced colitis by inhibiting NF-κB activation in mice. Inflammation 2018;41:1276-89.  Back to cited text no. 45
    
46.
Nag S, Krasikova R, Airaksinen AJ, Arakawa R, Petukhovd M, Gulyas B. Synthesis and biological evaluation of [18F] fluorovinpocetine, a potential PET radioligand for TSPO imaging. Bioorg Med Chem Lett 2019;29:2270-4.  Back to cited text no. 46
    
47.
Qiu X, Wang J, Lanying HE, Luo Y. Vinpocetine alleviates cerebral ischemia-reperfusion injury in rats by regulation of the expressions of nuclear factor κB p65, peroxisome proliferator-activated receptor γ and cyclooxygenase-2. Int J Cerebrovasc Dis 2015;23:517-21.  Back to cited text no. 47
    
48.
Yan J, Greer JM, Etherington K, Cadigan GP, Cavanagh H, Henderson RD, et al. Immune activation in the peripheral blood of patients with acute ischemic stroke. J Neuroimmunol 2009;206:112-7.  Back to cited text no. 48
    
49.
Lambertsen KL, Finsen B, Clausen BH. Post-stroke inflammation-target or tool for therapy? Acta Neuropathol 2019;137:693-714.  Back to cited text no. 49
    
50.
Xu L, He D, Bai Y. Microglia-mediated inflammation and neurodegenerative disease. Mol Neurobiol 2016;53:6709-15.  Back to cited text no. 50
    
51.
Lee DC, Ruiz CR, Lebson L, Selenica ML, Rizer J, Hunt JB Jr., et al. Aging enhances classical activation but mitigates alternative activation in the central nervous system. Neurobiol Aging 2013;34:1610-20.  Back to cited text no. 51
    
52.
Liu Z, Chopp M. Astrocytes, therapeutic targets for neuroprotection and neurorestoration in ischemic stroke. Prog Neurobiol 2016;144:103-20.  Back to cited text no. 52
    
53.
Pekny M, Wilhelmsson U, Tatlisumak T, Pekna M. Astrocyte activation and reactive gliosis-A new target in stroke? Neurosci Lett 2019;689:45-55.  Back to cited text no. 53
    
54.
Hersh J, Yang SH. Glia-immune interactions post-ischemic stroke and potential therapies. Exp Biol Med (Maywood) 2018;243:1302-12.  Back to cited text no. 54
    
55.
Ahangar AA, Saadat P, Alijanpour S, Galeshi M, Hosseinalipour S. Post ischemic stroke complication: How much nursing diagnosis are confirms by neurologist. J Patient Care 2018;4:2.  Back to cited text no. 55
    
56.
Vardjan N. Celica Biomedical, Assignee. Mechanism and Drug Targets for Reducing Cell Edema (Neuroprotection) and Cytoplasmic Excitability in Astrocytes in Normal and pathological States. United States Patent US 9,970,924; 2018. p. 15.  Back to cited text no. 56
    
57.
Zhuang J, Peng W, Li H, Lu Y, Wang K, Fan F, et al. Inhibitory effects of vinpocetine on the progression of atherosclerosis are mediated by Akt/NF-κB dependent mechanisms in apoE-/- mice. PLoS One 2013;8:e82509.  Back to cited text no. 57
    
58.
Kim HJ, Park KD, Choi KG, Lee HW. Clinical predictors of seizure recurrence after the first post-ischemic stroke seizure. BMC Neurol 2016;16:212.  Back to cited text no. 58
    
59.
Garza-Morales S, Briceño-González E, Ceja-Moreno H, Ruiz-Sandoval JL, Góngora-Rivera F, Rodríguez-Leyva I, et al. Extended-release vinpocetine: A possible adjuvant treatment for focal onset epileptic seizures. Bol Med Hosp Infant Me×2019;76:215-24.  Back to cited text no. 59
    
60.
Sitges M, Sanchez-Tafolla BM, Chiu LM, Aldana BI, Guarneros A. Vinpocetine inhibits glutamate release induced by the convulsive agent 4-aminopyridine more potently than several antiepileptic drugs. Epilepsy Res 2011;96:257-66.  Back to cited text no. 60
    
61.
Pande SD, Lwin MT, Kyaw KM, Khine AA, Thant AA, Win MM, et al. Post-stroke seizure-Do the locations, types and managements of stroke matter? Epilepsia Open 2018;3:392-8.  Back to cited text no. 61
    
62.
Villa RF, Ferrari F, Moretti A. Post-stroke depression: Mechanisms and pharmacological treatment. Pharmacol Ther 2018;184:131-44.  Back to cited text no. 62
    
63.
Cai W, Stewart R, Mueller C, Li YJ, Shen WD. Poststroke depression and risk of stroke recurrence and mortality: Protocol of a meta-analysis and systematic review. BMJ Open 2018;8:e026316.  Back to cited text no. 63
    
64.
Huang CW, Saposnik G, Fang J, Steven DA, Burneo JG. Influence of seizures on stroke outcomes: A large multicenter study. Neurology 2014;82:768-76.  Back to cited text no. 64
    
65.
Petrides G, Braga RJ, Fink M, Mueller M, Knapp R, Husain M, et al. Seizure threshold in a large sample: Implications for stimulus dosing strategies in bilateral electroconvulsive therapy: A report from CORE. J ECT 2009;25:232-7.  Back to cited text no. 65
    
66.
Chan L, Hu CJ, Fan YC, Li FY, Hu HH, Hong CT, et al. Incidence of poststroke seizures: A meta-analysis. J Clin Neurosci 2018;47:347-51.  Back to cited text no. 66
    
67.
Gómez CD, Buijs RM, Sitges M. The anti-seizure drugs vinpocetine and carbamazepine, but not valproic acid, reduce inflammatory IL-1β and TNF-α expression in rat hippocampus. J Neurochem 2014;130:770-9.  Back to cited text no. 67
    
68.
Espárrago Llorca G, Castilla-Guerra L, Fernández Moreno MC, Ruiz Doblado S, Jiménez Hernández MD. Post-stroke depression: An update. Neurologia 2015;30:23-31.  Back to cited text no. 68
    
69.
Levada OA, Troyan AS. Poststroke depression biomarkers: A narrative review. Front Neurol 2018;9:577.  Back to cited text no. 69
    
70.
Pascoe MC, Crewther SG, Carey LM, Crewther DP. Inflammation and depression: Why poststroke depression may be the norm and not the exception. Int J Stroke 2011;6:128-35.  Back to cited text no. 70
    
71.
Anisman H, Hayley S. Inflammatory factors contribute to depression and its comorbid conditions. Sci Signal 2012;5:pe45.  Back to cited text no. 71
    
72.
Spalletta G, Bossù P, Ciaramella A, Bria P, Caltagirone C, Robinson RG. The etiology of poststroke depression: A review of the literature and a new hypothesis involving inflammatory cytokines. Mol Psychiatry 2006;11:984-91.  Back to cited text no. 72
    
73.
Wiener CD, de Mello Ferreira S, Pedrotti Moreira F, Bittencourt G, de Oliveira JF, Lopez Molina M, et al. Serum levels of nerve growth factor (NGF) in patients with major depression disorder and suicide risk. J Affect Disord 2015;184:245-8.  Back to cited text no. 73
    
74.
Geng LY, Qian FY, Qian JF, Zhang ZJ. The combination of plasma glutamate and physical impairment after acute stroke as a potential indicator for the early-onset post-stroke depression. J Psychosom Res 2017;96:35-41.  Back to cited text no. 74
    
75.
Shao B, Zhou YL, Wang H, Lin YS. The role of calcitonin gene-related peptide in post-stroke depression in chronic mild stress-treated ischemic rats. Physiol Behav 2015;139:224-30.  Back to cited text no. 75
    
76.
Greifzu F, Schmidt S, Schmidt KF, Kreikemeier K, Witte OW, Löwel S. Global impairment and therapeutic restoration of visual plasticity mechanisms after a localized cortical stroke. Proc Natl Acad Sci U S A 2011;108:15450-5.  Back to cited text no. 76
    
77.
Lourenco-Gonzalez Y, Fattori V, Domiciano TP, Rossaneis AC, Borghi SM, Zaninelli TH, et al. Repurposing of the nootropic drug vinpocetine as an analgesic and anti-inflammatory agent: evidence in a mouse model of superoxide anion-triggered inflammation. Mediators Inflamm 2019;2019:6481812.  Back to cited text no. 77
    
78.
Al-Gareeb AI, Al-Windy S, Al-Kuraishy H. The effects of vinpocetine on the psychomotor performances: Randomized clinical trial, single blind random clinical study. J Al-Nahrain Univ Sci 2012;15:129-33.  Back to cited text no. 78
    
79.
Csillik B, Mihály A, Knyihár-Csillik E. Antinociceptive effect of vinpocetine-a comprehensive survey. Ideggyogy Sz 2010;63:185-92.  Back to cited text no. 79
    
80.
Wang C, Guo J, Guo R. Effect of XingPiJieYu decoction on spatial learning and memory and cAMP-PKA-CREB-BDNF pathway in rat model of depression through chronic unpredictable stress. BMC Complement Altern Med 2017;17:73.  Back to cited text no. 80
    
81.
Hénon H, Durieu I, Guerouaou D, Lebert F, Pasquier F, Leys D. Poststroke dementia: incidence and relationship to prestroke cognitive decline. Neurology 2001;57:1216-22.  Back to cited text no. 81
    
82.
Justin BN, Turek M, Hakim AM. Heart disease as a risk factor for dementia. Clin Epidemiol 2013;5:135-45.  Back to cited text no. 82
    
83.
Rothenburg LS, Herrmann N, Swardfager W, Black SE, Tennen G, Kiss A, et al. The relationship between inflammatory markers and post stroke cognitive impairment. J Geriatr Psychiatry Neurol 2010;23:199-205.  Back to cited text no. 83
    
84.
Narasimhalu K, Lee J, Leong YL, Ma L, De Silva DA, Wong MC, et al. Inflammatory markers and their association with post stroke cognitive decline. Int J Stroke 2015;10:513-8.  Back to cited text no. 84
    
85.
Jokinen H, Melkas S, Ylikoski R, Pohjasvaara T, Kaste M, Erkinjuntti T, et al. Post-stroke cognitive impairment is common even after successful clinical recovery. Eur J Neurol 2015;22:1288-94.  Back to cited text no. 85
    
86.
Shen Y, Gao HM. Serum somatostatin and neuron-specific enolase might be biochemical markers of vascular dementia in the early stage. Int J Clin Exp Med 2015;8:19471-5.  Back to cited text no. 86
    
87.
Hagberg G, Fure B, Thommessen B, Ihle-Hansen H, Øksengård AR, Nygård S, et al. Predictors for favorable cognitive outcome post-stroke: A-seven-year follow-up study. Dement Geriatr Cogn Disord 2019;28:1-11.  Back to cited text no. 87
    
88.
Hase Y, Horsburgh K, Ihara M, Kalaria RN. White matter degeneration in vascular and other ageing-related dementias. J Neurochem 2018;144:617-33.  Back to cited text no. 88
    
89.
Akinyemi RO, Allan LM, Oakley A, Kalaria RN. Hippocampal neurodegenerative pathology in post-stroke dementia compared to other dementias and aging controls. Front Neurosci 2017;11:717.  Back to cited text no. 89
    
90.
McQuown S, Xia S, Baumgärtel K, Barido R, Anderson G, Dyck B, et al. Phosphodiesterase 1b (PDE1B) regulates spatial and contextual memory in hippocampus. Front Mol Neurosci 2019;12:21.  Back to cited text no. 90
    
91.
Ali AA, Ahmed HI, Khaleel SA, Abu-Elfotuh K. Vinpocetine mitigates aluminum-induced cognitive impairment in socially isolated rats. Physiol Behav 2019;208:112571-79.  Back to cited text no. 91
    
92.
Scott Bitner R. Cyclic AMP response element-binding protein (CREB) phosphorylation: A mechanistic marker in the development of memory enhancing Alzheimer's disease therapeutics. Biochem Pharmacol 2012;83:705-14.  Back to cited text no. 92
    
93.
Reneerkens OA, Rutten K, Steinbusch HW, Blokland A, Prickaerts J. Selective phosphodiesterase inhibitors: A promising target for cognition enhancement. Psychopharmacology (Berl) 2009;202:419-43.  Back to cited text no. 93
    
94.
Robert Teasell MD, Macaluso S. Post-Stroke Cognitive Disorders; 2018;12:1-86.  Back to cited text no. 94
    


    Figures

  [Figure 1], [Figure 2], [Figure 3], [Figure 4]


This article has been cited by
1 SARS-COV-2 infection and Parkinson's disease: Possible links and perspectives
Hayder M. Al-kuraishy, Ali I. Al-Gareeb, Ajeet Kaushik, Malgorzata Kujawska, Eman A. Ahmed, Gaber El-Saber Batiha
Journal of Neuroscience Research. 2023;
[Pubmed] | [DOI]
2 A story of the potential effect of non-steroidal anti-inflammatory drugs (NSAIDs) in Parkinson’s disease: beneficial or detrimental effects
Mohammed Alrouji, Hayder M. Al-Kuraishy, Ali I. Al-Gareeb, Hebatallah M. Saad, Gaber El-Saber Batiha
Inflammopharmacology. 2023;
[Pubmed] | [DOI]
3 The Role of Iron Metabolism, Lipid Metabolism, and Redox Homeostasis in Alzheimer’s Disease: from the Perspective of Ferroptosis
Linyu Wu, Xiaohui Xian, Zixuan Tan, Fang Dong, Guangyu Xu, Min Zhang, Feng Zhang
Molecular Neurobiology. 2023;
[Pubmed] | [DOI]
4 Safety and Efficacy of Vinpocetine as a Neuroprotective Agent in Acute Ischemic Stroke: A Systematic Review and Meta-Analysis
Prateek Kumar Panda, Aparna Ramachandran, Pragnya Panda, Indar Kumar Sharawat
Neurocritical Care. 2022;
[Pubmed] | [DOI]
5 Citicoline and COVID-19: vis-à-vis conjectured
Hayder M. Al-kuraishy, Ali K. Al-Buhadily, Ali I. Al-Gareeb, Mohammed Alorabi, Nasser A. Hadi Al-Harcan, Maisra M. El-Bouseary, Gaber El-Saber Batiha
Naunyn-Schmiedeberg's Archives of Pharmacology. 2022;
[Pubmed] | [DOI]
6 Screening potential anaphylactoid components in vinpocetine injection using a high expression Mas -related G-protein-coupled receptor X2 cell membrane chromatography
Jiayu Lu, Xin Jia, Changhe Wang, Huaizhen He
Journal of Applied Toxicology. 2022;
[Pubmed] | [DOI]
7 Vinpocetine is the forthcoming adjuvant agent in the management of COVID-19
Hayder M Al-kuraishy, Ali I Al-Gareeb, Muyiwa Samuel Fageyinbo, Gaber El-Saber Batiha
Future Science OA. 2022;
[Pubmed] | [DOI]
8 Hypoxia and Alpha-Synuclein: Inextricable Link Underlying the Pathologic Progression of Parkinson's Disease
Mengyuan Guo, Xunming Ji, Jia Liu
Frontiers in Aging Neuroscience. 2022; 14
[Pubmed] | [DOI]
9 The role of Phosphodiesterase-1 and its natural product inhibitors in Alzheimer’s disease: A review
Nazir Ahmad, Kaisun Nesa Lesa, Ari Sudarmanto, Nanang Fakhrudin, Zullies Ikawati
Frontiers in Pharmacology. 2022; 13
[Pubmed] | [DOI]
10 Role of Neuropilin 1 in COVID-19 Patients with Acute Ischemic Stroke
Asma W. Al-Thomali, Hayder M. Al-kuraishy, Ali I. Al-Gareeb, Ali K. Al-buhadiliy, Michel De Waard, Jean-Marc Sabatier, Atif Ali Khan Khalil, Hebatallah M. Saad, Gaber El-Saber Batiha
Biomedicines. 2022; 10(8): 2032
[Pubmed] | [DOI]
11 Ki20227 aggravates apoptosis, inflammatory response, and oxidative stress after focal cerebral ischemia injury
Cheng Jiang,Ze-Ning Wang,Yu-Chen Kang,Yi Chen,Wei-Xin Lu,Hai-Jun Ren,Bo-Ru Hou
Neural Regeneration Research. 2022; 17(1): 137
[Pubmed] | [DOI]
12 Neuroprotective approach in acute ischemic stroke: A systematic review of clinical and experimental studies
Fettah Eren, SuedaEcem Yilmaz
Brain Circulation. 2022; 8(4): 172
[Pubmed] | [DOI]
13 Statin therapy associated with decreased neuronal injury measured by serum S100ß levels in patients with acute ischemic stroke
HayderM Al-Kuraishy, AliI Al-Gareeb, MarwaThaier Naji
International Journal of Critical Illness and Injury Science. 2021; 11(4): 246
[Pubmed] | [DOI]
14 Emerging neuroprotective strategies for the treatment of ischemic stroke: An overview of clinical and preclinical studies
Surojit Paul,Eduardo Candelario-Jalil
Experimental Neurology. 2021; 335: 113518
[Pubmed] | [DOI]
15 Advances in Cyclic Nucleotide Phosphodiesterase-Targeted PET Imaging and Drug Discovery
Jiyun Sun,Zhiwei Xiao,Ahmed Haider,Catherine Gebhard,Hao Xu,Hai-Bin Luo,Han-Ting Zhang,Lee Josephson,Lu Wang,Steven H. Liang
Journal of Medicinal Chemistry. 2021;
[Pubmed] | [DOI]
16 Cardiac myxoma as a potential trigger of takotsubo cardiomyopathy: A brief review on mechanistic and clinical perspectives
Kenan Yalta,Ugur Ozkan,Tülin Yalta,Ertan Yetkin
Monaldi Archives for Chest Disease. 2021;
[Pubmed] | [DOI]
17 Statins Therapy Improves Acute Ischemic Stroke in Patients with Cardio-metabolic Disorders Measured by Lipoprotein-Associated Phospholipase A2 (Lp-PLA2): New Focal Point
HayderM Al-kuraishy, NawarR Hussien, MarwaS Al-naimi, AliI Al-Gareeb, Claire Lugnier
Neurology India. 2021; 69(6): 1637
[Pubmed] | [DOI]
18 A systematic review of molecular approaches that link mitochondrial dysfunction and neuroinflammation in Parkinson’s disease
Sugumar Mani,Murugan Sevanan,Alagudurai Krishnamoorthy,Sathiya Sekar
Neurological Sciences. 2021;
[Pubmed] | [DOI]
19 Takotsubo cardiomyopathy: an obscure cause of emerging cardiovascular manifestations in the setting of Bickerstaff’s brainstem encephalitis
Kenan Yalta,Ertan Yetkin,Tulin Yalta
Neurological Sciences. 2020;
[Pubmed] | [DOI]
20 Clinical plasma concentration of vinpocetine does not affect osteogenic differentiation of mesenchymal stem cells
Esma Yildirim,Gulay Sezer
Pharmacological Reports. 2020;
[Pubmed] | [DOI]
21 ALSUntangled 57: Vinpocetine
Amyotrophic Lateral Sclerosis and Frontotemporal Degeneration. 2020; : 1
[Pubmed] | [DOI]
22 Brain natriuretic peptide in patients with acute ischemic stroke: Role of statins
HayderM Al-Kuraishy,AliI Al-Gareeb,MarwaThaier Naji
Biomedical and Biotechnology Research Journal (BBRJ). 2020; 4(3): 239
[Pubmed] | [DOI]
23 Involvement of orexinergic system in psychiatric and neurodegenerative disorders: A scoping review
HayderM Al-Kuraishy,MayH Abdulhadi,NawarR Hussien,MarwaS Al-Niemi,HudaA Rasheed,AliI Al-Gareeb
Brain Circulation. 2020; 6(2): 70
[Pubmed] | [DOI]



 

Top
 
 
  Search
 
    Similar in PUBMED
   Search Pubmed for
   Search in Google Scholar for
 Related articles
    Access Statistics
    Email Alert *
    Add to My List *
* Registration required (free)  

 
  In this article
Abstract
Introduction
Pharmacology of ...
Vinpocetine in I...
Conclusions
References
Article Figures

 Article Access Statistics
    Viewed39980    
    Printed232    
    Emailed0    
    PDF Downloaded4384    
    Comments [Add]    
    Cited by others 23    

Recommend this journal