Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 1847 2023-03-07 05:31:36 |
2 format correct Meta information modification 1847 2023-03-07 06:30:54 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Sinha, J.K.; Sachdeva, P.; Ghosh, S.; Ghosh, S.; Bhaskar, R.; Raghunath, M. Stem Cell Types for Treating Depression. Encyclopedia. Available online: https://encyclopedia.pub/entry/41920 (accessed on 26 June 2024).
Sinha JK, Sachdeva P, Ghosh S, Ghosh S, Bhaskar R, Raghunath M. Stem Cell Types for Treating Depression. Encyclopedia. Available at: https://encyclopedia.pub/entry/41920. Accessed June 26, 2024.
Sinha, Jitendra Kumar, Punya Sachdeva, Shampa Ghosh, Soumya Ghosh, Rakesh Bhaskar, Manchala Raghunath. "Stem Cell Types for Treating Depression" Encyclopedia, https://encyclopedia.pub/entry/41920 (accessed June 26, 2024).
Sinha, J.K., Sachdeva, P., Ghosh, S., Ghosh, S., Bhaskar, R., & Raghunath, M. (2023, March 07). Stem Cell Types for Treating Depression. In Encyclopedia. https://encyclopedia.pub/entry/41920
Sinha, Jitendra Kumar, et al. "Stem Cell Types for Treating Depression." Encyclopedia. Web. 07 March, 2023.
Stem Cell Types for Treating Depression
Edit

Major Depressive Disorder (MDD), colloquially known as depression, is a debilitating condition affecting an estimated 3.8% of the population globally, of which 5.0% are adults and 5.7% are above the age of 60. MDD is differentiated from common mood changes and short-lived emotional responses due to subtle alterations in gray and white matter, including the frontal lobe, hippocampus, temporal lobe, thalamus, striatum, and amygdala. It can be detrimental to a person’s overall health if it occurs with moderate or severe intensity. It can render a person suffering terribly to perform inadequately in their personal, professional, and social lives. Depression, at its peak, can lead to suicidal thoughts and ideation. Antidepressants manage clinical depression and function by modulating the serotonin, norepinephrine, and dopamine neurotransmitter levels in the brain. 

depression stem cells molecular pathways neurogenesis

1. Introduction

Major Depressive Disorder (MDD) is considered to be the most frequent psychiatric disorder [1] and, according to the World Health Organization (WHO), the leading cause of disability [2]. Low mood, diminished interest in daily activities, guilt, loss of pleasure, difficulty concentrating, low self-esteem, trouble sleeping, and altered appetite are some of the MDD symptoms. These issues can become chronic or recurrent, with severe consequences on a person’s ability to carry out daily activities. At its worst, depression can lead to suicidal thoughts [3]. Depression has been related to an increased chance of suffering from other severe illnesses, such as cardiovascular disease [4], stroke [3], Alzheimer’s disease [5], epilepsy [6], diabetes [7], and cancer [8]. Depressive symptoms are more commonly seen in older people, but this is due to factors linked with ageing, including physical disability [9], cognitive deficits, socioeconomic drawbacks, and other factors [10]. Treatment-resistant depression (TRD) can be caused by continuous exposure to environmental stressors during development [11].
Almost all antidepressants work the same way and effectively treat severe MDD across the lifespan [12][13]. However, antidepressant therapy has a number of adverse side effects, including sedation, headaches, decreased blood pressure, insomnia, weight gain, indigestion, feeling agitated, dry mouth, diarrhea, and sexual dysfunction [14]. This frequently leads to poor patient compliance, resulting in a recurrence of depressive symptoms and a higher risk of suicide [14].

2. Neurochemistry of Depression: The Monoamine Hypothesis

Norepinephrine (NE), serotonin (5-hydroxytryptamine, 5HT), and dopamine (DA) dysregulation are linked to the pathological changes seen in depression [15] (Figure 1). According to the monoamine hypothesis of depression, NE, 5HT, and DA work in synchrony to regulate emotions and mood [15]. In the depressed mood, the dysregulation of these three monoamines is observed, along with extracellular 5HT levels being lower than average. It has been reported that monoamines and metabolites are found to be lower in the urine, blood, and cerebrospinal fluid (CSF) of patients with depression as compared to age-matched controls [16].
Figure 1. The monoamine hypothesis of depression. The reduced levels of serotonin, norepinephrine, and dopamine have been observed and are understood as one of the main factors responsible for the generation of depressive symptoms.
The sympathetic nervous system’s primary neurotransmitter, NE, is produced in the locus coeruleus (LC) and is capable of sending projections all over the CNS. Research on depression began with a focus on the noradrenergic system, starting with introducing the “motor activation deficit” hypothesis [16]. Patients with depression and victims of suicidal behavior have been seen to show deficits in the noradrenergic system in the LC compared with healthy individuals [17]. Furthermore, some genetic changes in norepinephrine transporters (NETs) are highly plausible to be associated with psychiatric diseases [18][19]. It has been discovered that heterogeneously projecting neurons from the LC can separately lead to modulation of fear and learning, emphasizing that an imbalance in the activation of these neuronal groups may be linked with post-traumatic stress disorder (PTSD) and depression in rats [15]. Another monoamine neurotransmitter, DA, is well known to play an essential role in motivation, concentration, reward, psychomotor speed, and emotional response [15]. Notably, changes in DA functioning have also been linked to depression-related fatigue symptoms [20]. Depressed patients have lower DA metabolites in their CSF, which supports this hypothesis [21]. Pharmacologically, increased dopamine binding and availability of the dopamine transporter (DAT) in the striatum have been observed to improve depression symptoms in humans [22]. Furthermore, ropinirole, a dopaminergic targeting drug, has improved antidepressant treatment responsiveness in refractory patients [23].
Serotonin is synthesized in the dorsal raphe nucleus from tryptophan (via a 5-OH-tryptophan intermediate). The dorsal raphe nucleus sends projections to the whole CNS including, remarkably, the brain areas that are vulnerable to stress, such as the hippocampus. Serotonin is a widely distributed neurotransmitter that also acts as a neuromodulator [24][25][26]. Ever since its discovery, 5HT in the brain has been linked to circadian rhythms, sleep, cognitive abilities, appetite, motor activities, and many more biological functions [27]. Furthermore, 5HT involvement in response to stress [28] and psychiatric disease [29] has received a lot of attention. A decrease in serum 5HT [30] and plasma tryptophan [31] levels and alterations in metabolite levels of 5HIAA (5-hydroxyindoleacetic acid) in CSF have been observed in patients with a depressive illness [31]. Serotonin and its metabolites however have not been found to be a persistent biomarker of depression [26]. The function of 5HT receptors (particularly autoreceptors) and 5HT transporters is also being studied in patients with depression [26].

3. Growth Factors Involved in Depression

There are several biological factors associated with depressive symptoms, as shown in Figure 2. It has been investigated that stress-induced epigenetic changes can lead to depression [32][33]. Two meta-analyses of studies examining temporal lobe structures in MDD indicated that patients with recurrent depression have a smaller hippocampus [34][35]. Synaptic plasticity in neural circuits associated with depressive behaviors is regulated by the brain-derived neurotrophic factor (BDNF) [36][37][38]. Interestingly, stress-induced impairments in the brain structure and synaptic plasticity may be reversed by BDNF upregulation, leading to flexibility in cognition and an elevated capacity to acclimatize to environmental changes that might stimulate depressive episodes. According to current research, in depressed subjects, BDNF levels in the blood are lower, and they increase with antidepressant treatment [39]. Additionally, elevated BDNF plasma levels have been linked to better treatment outcomes regardless of the medication used [40].
Figure 2. Biological factors and clinical manifestations associated with depression. There are multiple biological causes at molecular, genetic, epigenetic, cellular, and systems levels. These causes result in clinical depression and can have a plethora of symptoms that may vary in different individuals.
Furthermore, anxiety, depression risk, neuroticism, and serotonergic neurotransmission have all been linked to altered serum BDNF levels and BDNF gene polymorphism [2]. Lithium augmentation refers to the addition of lithium to an antidepressant in the acute treatment phase of depression [41][42]. Antidepressant augmentation with lithium is a well-studied augmentation therapy for patients with depression who have not been responding well to antidepressant therapy [41]. Moreover, lithium augmentation has the ability to increase BDNF concentrations in the serum [40].
The reduced levels of nerve growth factor (NGF) are involved in the pathophysiology of depression. Fluoxetine and lithium are known to treat depression by upregulating NGF protein levels in the hippocampus [43]. Neurotrophin-3 (NT-3) is responsible for neuron proliferation, differentiation, and survival. NT-3 also promotes the growth of axons and dendrites. Clinical data from post-mortem research showed that people with depressive disorders have lower NT-3 levels in their parietal brain [44][45]. The decreased glial cell line-derived neurotrophic factor (GDNF) in the serum is correlated with the development of depressive symptoms. GDNF has a crucial role in the survival and maintenance of monoaminergic neurons. It has been reported that reduced GDNF levels abnormally regulate serotonergic neurons and reduce post and pre-synaptic serotonin receptors in patients with depression [46]. Fibroblast growth factor-2 (FGF-2) is another growth factor associated with depression [47]. Recent research has suggested that BDNF may signal via the Akt and GSK3 pathways [48].
Further, it has been seen that erythropoietin impacts neuroplasticity and could be used to treat depression in the future. A study by Miskowiak et al. recruited 40 patients with a bipolar mood disorder and 40 patients with TRD. The patients received either a weekly intravenous infusion of erythropoietin or saline. It was observed that a single dose of erythropoietin (Eprex; 40,000IU) could improve cognitive functioning and reduce the neurocognitive processing involved in negative emotional information in normal versus people with depression in a way that is similar to antidepressants’ effects [49]. In the limited research conducted on unipolar depression, inositol, a component of the intracellular phosphatidyl-inositol second-messenger system, has also been shown to be effective [50]. Finally, peripheral VGF (nonacronymic) showed a reduced expression in MDD patients, and recombinant VGF administration causes antidepressant effects in rats [50].

4. Neural Stem Cells and Depression

NSCs have garnered interest in recent years with the extensive published literature elucidating that the adult brain maintains multipotent NSCs in contrast to the old dogma of the brain being a generally invariable and quiescent organ that lacks the flexibility to regenerate. With their most generally accepted distinguishing traits, NSCs are also ascribed to the so-called tissue stem cells [51], featuring the power to stay undifferentiated without an outlined phenotype under specific conditions, the power of dividing and proliferating (self-renewal), and also the ability to be differentiated into a progeny like neurons, oligodendroglia, and astroglia upon neurogenesis initiation. They are the unique types of competent cells found within the adult mammalian brain’s “neurogenic” regions, such as the hippocampus [52], subventricular zone [53], and neural structures [54], and might create neurons both spontaneously and in response to local signals induction. Neurogenesis (NG) is assumed to need an explicit set of signaling cues to be delivered to cells that are neurogenic in a very spatially and temporally coordinated manner by their surroundings so as to activate stem cells or progenitors to develop new neurons and, in addition to the well-known modulators [55], injury is considered to be sufficient to activate neurogenesis. Neurogenesis is also stimulated by the expression of BDNF [56]. NSCs are often extracted from adult brain tissues, including post-mortem brain tissue [57], and become significant candidates for increasing or restoring the quality and function of brain tissue affected with CNS-related illnesses. The NSCs are clonally expanded in vitro, genetically manipulated, or stimulated to transform CNS cell lineages [57]. Understanding how adult neurogenesis is regulated has required significant work. 
Growth factors, transmitters, enzymes, tissue hormones, neuromodulators, and antibodies are predicted to be secreted into the local tissue environment by activated cells, eliciting desirable tissue responses. In damaged neuronal and glial networks, the newly empowered cells and their progeny can operate as functional enhancers and scaffold “healing agents.” These properties have led to substantial advancements in the invention of therapies for trauma and perfusion issues such as stroke [58], ischemia, or neurodegeneration-related conditions [58][59][60]. Not unexpectedly, the prospects of NSCs in mental health care are being hotly debated. Numerous psychiatric illnesses are likely to possess genetic variants and specific cellular and anatomical correlations that are mostly unknown [58].
In depression, a reduction of neurogenesis is often seen in the hippocampus [61]. This further implies that neurogenesis deficiencies might cause the symptoms associated with depression, while enhanced neurogenesis can mediate antidepressant action and ease symptoms. However, various conflicting reports regarding the role of neurogenesis in alleviating depression must be first reconciled before this bidirectional concept’s complete legitimacy is established [62]. The activation of adult hippocampal neurogenesis leads to the transformation of neural somatic cell progeny to mature CNS neurons. These CNS neurons then acquire functional and morphological qualities to integrate into existing neural networks or replace various other brain cells that have died [63][64].

References

  1. Maj, M. When Does Depression Become a Mental Disorder? Br. J. Psychiatry 2011, 199, 85–86.
  2. Lang, U.E.; Borgwardt, S. Molecular Mechanisms of Depression: Perspectives on New Treatment Strategies. Cell Physiol. Biochem. 2013, 31, 761–777.
  3. Ramasubbu, R.; Patten, S.B. Effect of Depression on Stroke Morbidity and Mortality. Can. J. Psychiatry 2003, 48, 250–257.
  4. Van der Kooy, K.; van Hout, H.; Marwijk, H.; Marten, H.; Stehouwer, C.; Beekman, A. Depression and the Risk for Cardiovascular Diseases: Systematic Review and Meta Analysis. Int. J. Geriat. Psychiatry 2007, 22, 613–626.
  5. Green, R.C.; Cupples, L.A.; Kurz, A.; Auerbach, S.; Go, R.; Sadovnick, D.; Duara, R.; Kukull, W.A.; Chui, H.; Edeki, T.; et al. Depression as a Risk Factor for Alzheimer Disease: The MIRAGE Study. Arch. Neurol. 2003, 60, 753.
  6. Hesdorffer, D.C.; Hauser, W.A.; Annegers, J.F.; Cascino, G. Major Depression Is a Risk Factor for Seizures in Older Adults. Ann. Neurol. 2000, 47, 246–249.
  7. Nouwen, A.; Lloyd, C.E.; Pouwer, F. Depression and Type 2 Diabetes Over the Lifespan: A Meta-Analysis. Diabetes Care 2009, 32, e56.
  8. Penninx, B.W.J.H.; Guralnik, J.M.; Havlik, R.J.; Pahor, M.; Ferrucci, L.; Cerhan, J.R.; Wallace, R.B. Chronically Depressed Mood and Cancer Risk in Older Persons. JNCI J. Natl. Cancer Inst. 1998, 90, 1888–1893.
  9. Kaur, J.; Ghosh, S.; Singh, P.; Dwivedi, A.K.; Sahani, A.K.; Sinha, J.K. Cervical Spinal Lesion, Completeness of Injury, Stress, and Depression Reduce the Efficiency of Mental Imagery in People With Spinal Cord Injury. Am. J. Phys. Med. Rehabil. 2022, 101, 513–519.
  10. Maniam, J.; Antoniadis, C.P.; Youngson, N.A.; Sinha, J.K.; Morris, M.J. Sugar Consumption Produces Effects Similar to Early Life Stress Exposure on Hippocampal Markers of Neurogenesis and Stress Response. Front. Mol. Neurosci. 2015, 8, 86.
  11. Rush, A.J.; Trivedi, M.H.; Wisniewski, S.R.; Nierenberg, A.A.; Stewart, J.W.; Warden, D.; Niederehe, G.; Thase, M.E.; Lavori, P.W.; Lebowitz, B.D.; et al. Acute and Longer-Term Outcomes in Depressed Outpatients Requiring One or Several Treatment Steps: A STAR*D Report. Am. J. Psychiatry 2006, 163, 1905–1917.
  12. Akil, H.; Gordon, J.; Hen, R.; Javitch, J.; Mayberg, H.; McEwen, B.; Meaney, M.J.; Nestler, E.J. Treatment Resistant Depression: A Multi-Scale, Systems Biology Approach. Neurosci. Biobehav. Rev. 2018, 84, 272–288.
  13. Salzman, C.; Wong, E.; Wright, B.C. Drug and ECT Treatment of Depression in the Elderly, 1996–2001: A Literature Review. Biol. Psychiatry 2002, 52, 265–284.
  14. Keller, M.B.; Hirschfeld, R.M.A.; Demyttenaere, K.; Baldwin, D.S. Optimizing Outcomes in Depression: Focus on Antidepressant Compliance. Int. Clin. Psychopharmacol. 2002, 17, 265–271.
  15. Nestler, E.J.; Carlezon, W.A. The Mesolimbic Dopamine Reward Circuit in Depression. Biol. Psychiatry 2006, 59, 1151–1159.
  16. Roy, A. Cerebrospinal Fluid Monoamine Metabolites and Suicidal Behavior in Depressed Patients: A 5-Year Follow-up Study. Arch. Gen. Psychiatry 1989, 46, 609.
  17. Klimek, V.; Stockmeier, C.; Overholser, J.; Meltzer, H.Y.; Kalka, S.; Dilley, G.; Ordway, G.A. Reduced Levels of Norepinephrine Transporters in the Locus Coeruleus in Major Depression. J. Neurosci. 1997, 17, 8451–8458.
  18. Marshe, V.S.; Maciukiewicz, M.; Rej, S.; Tiwari, A.K.; Sibille, E.; Blumberger, D.M.; Karp, J.F.; Lenze, E.J.; Reynolds, C.F.; Kennedy, J.L.; et al. Norepinephrine Transporter Gene Variants and Remission From Depression With Venlafaxine Treatment in Older Adults. Am. J. Psychiatry 2017, 174, 468–475.
  19. Dunn, A.J. Effects of Cytokines and Infections on Brain Neurochemistry. Clin. Neurosci. Res. 2006, 6, 52–68.
  20. Anacker, C.; Cattaneo, A.; Musaelyan, K.; Zunszain, P.A.; Horowitz, M.; Molteni, R.; Luoni, A.; Calabrese, F.; Tansey, K.; Gennarelli, M.; et al. Role for the Kinase SGK1 in Stress, Depression, and Glucocorticoid Effects on Hippocampal Neurogenesis. Proc. Natl. Acad. Sci. USA 2013, 110, 8708–8713.
  21. Jokinen, J.; Nordström, A.-L.; Nordström, P. The Relationship Between CSF HVA/5-HIAA Ratio and Suicide Intent in Suicide Attempters. Arch. Suicide Res. 2007, 11, 187–192.
  22. Pizzagalli, D.A.; Berretta, S.; Wooten, D.; Goer, F.; Pilobello, K.T.; Kumar, P.; Murray, L.; Beltzer, M.; Boyer-Boiteau, A.; Alpert, N.; et al. Assessment of Striatal Dopamine Transporter Binding in Individuals With Major Depressive Disorder: In Vivo Positron Emission Tomography and Postmortem Evidence. JAMA Psychiatry 2019, 76, 854.
  23. Cassano, P.; Lattanzi, L.; Fava, M.; Navari, S.; Battistini, G.; Abelli, M.; Cassano, G.B. Ropinirole in Treatment-Resistant Depression: A 16-Week Pilot Study. Can. J. Psychiatry 2005, 50, 357–360.
  24. Descarries, L.; Watkins, K.C.; Garcia, S.; Beaudet, A. The Serotonin Neurons in Nucleus Raphe Dorsalis of Adult Rat: A Light and Electron Microscope Radioautographic Study. J. Comp. Neurol. 1982, 207, 239–254.
  25. Bunin, M.A.; Wightman, R.M. Quantitative Evaluation of 5-Hydroxytryptamine (Serotonin) Neuronal Release and Uptake: An Investigation of Extrasynaptic Transmission. J. Neurosci. 1998, 18, 4854–4860.
  26. Steinbusch, H.W.M. Distribution of Serotonin-Immunoreactivity in the Central Nervous System of the Rat—Cell Bodies and Terminals. Neuroscience 1981, 6, 557–618.
  27. Mann, J. Role of the Serotonergic System in the Pathogenesis of Major Depression and Suicidal Behavior. Neuropsychopharmacology 1999, 21, 99S–105S.
  28. Chaouloff, F. Serotonin and Stress. Neuropsychopharmacology 1999, 21, 28S–32S.
  29. Andrews, P.W.; Bharwani, A.; Lee, K.R.; Fox, M.; Thomson, J.A. Is Serotonin an Upper or a Downer? The Evolution of the Serotonergic System and Its Role in Depression and the Antidepressant Response. Neurosci. Biobehav. Rev. 2015, 51, 164–188.
  30. Bot, M.; Chan, M.K.; Jansen, R.; Lamers, F.; Vogelzangs, N.; Steiner, J.; Leweke, F.M.; Rothermundt, M.; Cooper, J.; Bahn, S.; et al. Serum Proteomic Profiling of Major Depressive Disorder. Transl. Psychiatry 2015, 5, e599.
  31. Quintana, J. Platelet Serotonin and Plasma Tryptophan Decreases in Endogenous Depression. Clinical, Therapeutic, and Biological Correlations. J. Affect. Disord. 1992, 24, 55–62.
  32. Park, C.; Rosenblat, J.D.; Brietzke, E.; Pan, Z.; Lee, Y.; Cao, B.; Zuckerman, H.; Kalantarova, A.; McIntyre, R.S. Stress, Epigenetics and Depression: A Systematic Review. Neurosci. Biobehav. Rev. 2019, 102, 139–152.
  33. Ghosh, S.; Sinha, J.K.; Raghunath, M. “Obesageing”: Linking Obesity & Ageing. Indian J. Med. Res. 2019, 149, 610–615.
  34. Campbell, S.; MacQueen, G. An Update on Regional Brain Volume Differences Associated with Mood Disorders. Curr. Opin. Psychiatry 2006, 19, 25–33.
  35. Videbech, P. Hippocampal Volume and Depression: A Meta-Analysis of MRI Studies. Am. J. Psychiatry 2004, 161, 1957–1966.
  36. Mishra, P.; Mittal, A.K.; Kalonia, H.; Madan, S.; Ghosh, S.; Sinha, J.K.; Rajput, S.K. SIRT1 Promotes Neuronal Fortification in Neurodegenerative Diseases through Attenuation of Pathological Hallmarks and Enhancement of Cellular Lifespan. Curr. Neuropharmacol. 2021, 19, 1019–1037.
  37. Moroi, K.; Sato, T. Comparison between Procaine and Isocarboxazid Metabolism in Vitro by a Liver Microsomal Amidase-Esterase. Biochem. Pharmacol. 1975, 24, 1517–1521.
  38. Pittenger, C.; Duman, R.S. Stress, Depression, and Neuroplasticity: A Convergence of Mechanisms. Neuropsychopharmacology 2008, 33, 88–109.
  39. Aydemir, O.; Deveci, A.; Taneli, F. The Effect of Chronic Antidepressant Treatment on Serum Brain-Derived Neurotrophic Factor Levels in Depressed Patients: A Preliminary Study. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2005, 29, 261–265.
  40. Ricken, R.; Adli, M.; Lange, C.; Krusche, E.; Stamm, T.J.; Gaus, S.; Koehler, S.; Nase, S.; Bschor, T.; Richter, C.; et al. Brain-Derived Neurotrophic Factor Serum Concentrations in Acute Depressive Patients Increase During Lithium Augmentation of Antidepressants. J. Clin. Psychopharmacol. 2013, 33, 806–809.
  41. Bauer, M.; Adli, M.; Bschor, T.; Pilhatsch, M.; Pfennig, A.; Sasse, J.; Schmid, R.; Lewitzka, U. Lithium’s Emerging Role in the Treatment of Refractory Major Depressive Episodes: Augmentation of Antidepressants. Neuropsychobiology 2010, 62, 36–42.
  42. Coradduzza, D.; Garroni, G.; Congiargiu, A.; Balzano, F.; Cruciani, S.; Sedda, S.; Nivoli, A.; Maioli, M. MicroRNAs, Stem Cells in Bipolar Disorder, and Lithium Therapeutic Approach. Int. J. Mol. Sci. 2022, 23, 10489.
  43. Mondal, A.C.; Fatima, M. Direct and Indirect Evidences of BDNF and NGF as Key Modulators in Depression: Role of Antidepressants Treatment. Int. J. Neurosci. 2019, 129, 283–296.
  44. de Miranda, A.S.; de Barros, J.L.V.M.; Teixeira, A.L. Is Neurotrophin-3 (NT-3): A Potential Therapeutic Target for Depression and Anxiety? Expert Opin. Ther. Targets 2020, 24, 1225–1238.
  45. Diniz, B.S.; Teixeira, A.L.; Miranda, A.S.; Talib, L.L.; Gattaz, W.F.; Forlenza, O.V. Circulating Glial-Derived Neurotrophic Factor Is Reduced in Late-Life Depression. J. Psychiatr. Res. 2012, 46, 135–139.
  46. Evans, S.J.; Choudary, P.V.; Neal, C.R.; Li, J.Z.; Vawter, M.P.; Tomita, H.; Lopez, J.F.; Thompson, R.C.; Meng, F.; Stead, J.D.; et al. Dysregulation of the Fibroblast Growth Factor System in Major Depression. Proc. Natl. Acad. Sci. USA 2004, 101, 15506–15511.
  47. Beaulieu, J.-M. A Role for Akt and Glycogen Synthase Kinase-3 as Integrators of Dopamine and Serotonin Neurotransmission in Mental Health. J. Psychiatry Neurosci. 2012, 37, 7–16.
  48. Miskowiak, K.W.; Vinberg, M.; Harmer, C.J.; Ehrenreich, H.; Knudsen, G.M.; Macoveanu, J.; Hansen, A.R.; Paulson, O.B.; Siebner, H.R.; Kessing, L.V. Effects of Erythropoietin on Depressive Symptoms and Neurocognitive Deficits in Depression and Bipolar Disorder. Trials 2010, 11, 97.
  49. Eden Evins, A.; Demopulos, C.; Yovel, I.; Culhane, M.; Ogutha, J.; Grandin, L.D.; Nierenberg, A.A.; Sachs, G.S. Inositol Augmentation of Lithium or Valproate for Bipolar Depression. Bipolar Disord. 2006, 8, 168–174.
  50. Cattaneo, A.; Sesta, A.; Calabrese, F.; Nielsen, G.; Riva, M.A.; Gennarelli, M. The Expression of VGF Is Reduced in Leukocytes of Depressed Patients and It Is Restored by Effective Antidepressant Treatment. Neuropsychopharmacology 2010, 35, 1423–1428.
  51. Urbán, N.; Blomfield, I.M.; Guillemot, F. Quiescence of Adult Mammalian Neural Stem Cells: A Highly Regulated Rest. Neuron 2019, 104, 834–848.
  52. Kukekov, V.G.; Laywell, E.D.; Suslov, O.; Davies, K.; Scheffler, B.; Thomas, L.B.; O’Brien, T.F.; Kusakabe, M.; Steindler, D.A. Multipotent Stem/Progenitor Cells with Similar Properties Arise from Two Neurogenic Regions of Adult Human Brain. Exp. Neurol. 1999, 156, 333–344.
  53. Alvarez-Buylla, A.; García-Verdugo, J.M. Neurogenesis in Adult Subventricular Zone. J. Neurosci. 2002, 22, 629–634.
  54. Liu, Z.; Martin, L.J. Olfactory Bulb Core Is a Rich Source of Neural Progenitor and Stem Cells in Adult Rodent and Human. J. Comp. Neurol. 2003, 459, 368–391.
  55. Kempermann, G. Regulation of Adult Hippocampal Neurogenesis—Implications for Novel Theories of Major Depression 1: Regulation of Adult Hippocampal Neurogenesis. Bipolar Disord. 2002, 4, 17–33.
  56. Mansoor, A.K.; Thomas, S.; Sinha, J.K.; Alladi, P.A.; Ravi, V.; Raju, T.R. Olfactory tract transection reveals robust tissue-level plasticity by cellular numbers and neurotrophic factor expression in olfactory bulb. Indian J. Exp. Biol. 2012, 50, 765–770.
  57. Feldmann, R.E.; Mattern, R. The Human Brain and Its Neural Stem Cells Postmortem: From Dead Brains to Live Therapy. Int. J. Leg. Med. 2006, 120, 201–211.
  58. Lindvall, O.; Kokaia, Z. Recovery and Rehabilitation in Stroke: Stem Cells. Stroke 2004, 35, 2691–2694.
  59. Sachdeva, P.; Ghosh, S.; Ghosh, S.; Han, S.; Banerjee, J.; Bhaskar, R.; Sinha, J.K. Childhood Obesity: A Potential Key Factor in the Development of Glioblastoma Multiforme. Life 2022, 12, 1673.
  60. Ghosh, S.; Manchala, S.; Raghunath, M.; Sharma, G.; Singh, A.K.; Sinha, J.K. Role of Phytomolecules in the Treatment of Obesity: Targets, Mechanisms and Limitations. Curr. Top. Med. Chem. 2021, 21, 863–877.
  61. Goldman, S. Stem and Progenitor Cell–Based Therapy of the Human Central Nervous System. Nat. Biotechnol. 2005, 23, 862–871.
  62. Lipska, B.K. Using Animal Models to Test a Neurodevelopmental Hypothesis of Schizophrenia. J. Psychiatry Neurosci. 2004, 29, 282–286.
  63. Feldmann, R.E.; Sawa, A.; Seidler, G.H. Causality of Stem Cell Based Neurogenesis and Depression—To Be or Not to Be, Is That the Question? J. Psychiatr. Res. 2007, 41, 713–723.
  64. Zhao, C. Distinct Morphological Stages of Dentate Granule Neuron Maturation in the Adult Mouse Hippocampus. J. Neurosci. 2006, 26, 3–11.
More
Information
Subjects: Neurosciences
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , , , ,
View Times: 918
Entry Collection: Neurodegeneration
Revisions: 2 times (View History)
Update Date: 07 Mar 2023
1000/1000
Video Production Service