Skip to main content

MINI REVIEW article

Front. Aging Neurosci., 24 August 2022
Sec. Neurocognitive Aging and Behavior
Volume 14 - 2022 | https://doi.org/10.3389/fnagi.2022.821866

Perspectives on the complex links between depression and dementia

  • 1Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, Canada
  • 2Division of Neurology, University of Ottawa, Ottawa, ON, Canada

This review highlights that depression is a growing health problem for the individual, and because of its high frequency in most societies, a growing burden on health care budgets. The focus of the review is the physiological links between depression and dementia, specifically Alzheimer’s disease. It suggests that depression is a significant risk factor for cognitive decline and explores the pathways that may lead depressed individuals to suffer this outcome. This review shows that depression and a number of its precursors activate pro-inflammatory mediators. These lead to cerebral small vessel disease with the consequent reduction in cerebral blood flow, which is known to precede cognitive decline. Thus, the impact of depression on the physiological events that lead to dementia is identical to the impact of other dementia risk factors recently reviewed. Depression is distinct, however, in being a relatively treatable condition, but the impact of treating depression on later cognitive decline is not always positive, leading to the hypothesis that only the antidepressants that attenuate inflammation alleviate subsequent cognitive decline.

Introduction

Caring for dementia patients is consuming growing portions of the health care budgets of many countries. The recognition that certain risk factors increase the risk for dementia provides the possibility of reducing this burden by intensifying efforts to reduce and control the risk factors. In a recent article (Hakim, 2021), a hypothesis was presented that a sequence of physiological events links dementia risk factors to their cognitive outcomes. It was proposed that in the presence of a risk factor, the sequence that leads to dementia is triggered by inflammation which leads to cerebral small vessel disease. This results in a reduction of cerebral perfusion, which precedes the appearance of any clinical evidence for cognitive impairment. Data were presented to highlight this sequence in three recognized dementia risk factors: obesity, sedentary lifestyle, and insufficient sleep. Depression is recognized as a risk factor for dementia, and the current literature review explores the pathway from depression to dementia and suggests that it follows the same sequence of physiological events that link other risk factors to dementia. A significant difference between depression and the other risk factors is the possibility that some antidepressant medications may alleviate this risk to cognitive decline. This benefit, however, may be limited to the antidepressants that reduce inflammation.

Dementia and depression are growing problems

All published estimates agree that the number of people affected by dementia will substantially increase over time, mainly due to projected trends in population aging and growth. The GBD2019 Dementia Forecasting Collaborators estimate the number of people with dementia globally will increase from 57.4 million in 2019 to 152.8 million in 2050 (GBD 2019 Dementia Forecasting Collaborators, 2022). Wittenberg et al. (2020) in 2020 projected that the number of older people with dementia will more than double in the next 25 years.

When dementia is estimated by age group, a clear correlation is evident between aging and prevalence of the condition. Gautrin et al. (1990) calculated dementia prevalence to be 1% for ages 65–74, 4% for ages 75–84, and 10.5% for 85 and over. In contrast, depression is not more prevalent with age even though its prevalence is also rising in many jurisdictions. A community-based study of American adults found that the 1-year frequency of major depressive disorder (MDD) rose from 3.33 to 7.06% between 1991–92 and 2001–02 (Compton, 2006). An analysis of the Minnesota Multiphasic Personality Inventory (MMPI), which consists of data on 63,706 American college students and 13,870 high school students revealed that younger adults were 6–8 times more likely than older adults to meet the criteria for clinical depression in 2007 compared to peers in 1938 (Twenge et al., 2010). In a Swedish population studied approximately every 10 years, it was shown that the risk for depression in young adults had increased 10-fold from 1957 to 1972 compared to the period from 1947 to 1957 (Hagnell, 1989; Hagnell et al., 1993). A data brief from the US department of Health and Human Services reported in September 2020 that the percentage of adults who experienced any symptoms of depression was highest among those aged 18–29 (21.0%), followed by those aged 45–64 and those older than 65 (18.4%), and lastly by those aged 30–44 (16.8%). For all degrees of depression severity, women were more likely to be affected than men (Villarroel and Terlizzi, 2020). There is thus a concordance of studies showing that the incidence of depression is rising and that the younger adult age groups are more likely to develop depression, with onset at increasingly earlier ages (Hidaka, 2012). We may therefore be in the middle of an epidemic of depression, and its impact on cognitive functions is worthy of further analysis.

Depression increases the risk for dementia

Cognitive decline in later life has been associated with many factors; a review of these factors revealed a link between depression and the onset of dementia (Steffens et al., 2004). In a 14-year longitudinal study which followed 4,922 healthy men aged 71–89 years, 18.3% developed dementia. Interestingly, the men who were older and had a history of depression were at greater risk of developing dementia (Almeida et al., 2017). The authors concluded that the link between depression and cognitive decline was evident during the initial 5 years of follow-up. Zeki Al Hazzouri et al. (2018) reported in the Northern Manhattan Study (2018) that greater depressive symptoms, adjusted for other variables, were significantly associated with worse baseline episodic memory in populations. Greater depressive symptoms were significantly related to poorer baseline episodic memory function (β [95% confidence interval] = −0.21 [−0.33 to −0.10],p = 0.0003) even when the models had been adjusted for socio-demographics, vascular risk factors, and medications for behavioral and mental health issues (Zeki Al Hazzouri et al., 2018). Gatchel et al. (2019), in a longitudinal study of 276 cognitively unimpaired older adults, showed that worsening depressive symptoms were significantly associated with declining cognition. Finally, researchers have concluded that half of the patients with major depressive disorders showed cognitive and memory impairments (Köhler et al., 2010). Norton et al. (2014) predicted that depression accounted for 5–11% of all Alzheimer’s disease cases. There is therefore concordant evidence in the literature that depression is a significant risk factor for cognitive decline.

The risk factors for depression

It is now widely appreciated that depression can have a number of precursors and social determinants (Slavich and Irwin, 2014). Exposure to early life stressors such as social stressors, social isolation, and the inability to form attachments are all possible risk factors for depression (Panksepp, 2003; Watt and Panksepp, 2009). Early maltreatment has been associated with late-life depression and suicide risk (Comijs et al., 2013). Watt and Panksepp (2009) conceptualize depression as arising from an evolutionarily preserved “shutdown mechanism” resulting from protracted separation distress in early life. These types of stressors have been linked to inflammation and changes in immune function which may lead to both depression and dementia.

In 2003, Eisenberger et al. (2003), demonstrated that with increasing social distress greater activity could be observed in the anterior cingulate cortex, an area implicated in generating the aversive experience of physical pain. A study done by Slavich and Irwin (2014), examined young adults who were exposed to social stressors while monitoring markers of inflammatory activity and brain activity using fMRI. Social stress exposure resulted in significant increases in a soluble receptor for tumor necrosis factor alpha and interleukin-6. The TNF-alpha receptor increases were associated with greater activity in the dorsal anterior cingulate cortex and anterior insula. These regions have been previously associated with processing rejection-related distress. A second study by Muscatell et al. (2015) showed that higher levels of neuronal activity in the amygdala in response to stress was associated with greater increases in inflammation.

Overall, these studies suggest that proinflammatory cytokines are the crucial mediators between the risk factors and their depression consequences often evident as sad mood, chronic feeling of fatigue, social withdrawal, and anhedonia. Therefore, targeting inflammation may offer new opportunities for preventing and treating depression.

The pathogenesis of cognitive decline. Alzheimer’s disease is the major contributor to dementia

Crous-Bou et al. (2017) state that Alzheimer’s disease accounts for 60–80% of dementias, making it the most common precursor of cognitive decline. In that condition the hallmark pathological criteria have included elevated levels of amyloid-beta peptide and hyperphosphorylated tau which accumulates intracellularly and becomes microscopically evident as neurofibrillary tangles. Recent evidence, however, suggests that a decline in cerebral blood flow precedes these pathological hallmarks of Alzheimer’s disease, potentially by many years. In an extensive study by Iturria-Medina et al. (2016), where multiple simultaneous measurements of regional cerebral perfusion and other biomarkers of Alzheimer’s disease were made, a decline in cerebral perfusion preceded all other pathological hallmarks of Alzheimer’s disease. Bangen et al. (2018) have subsequently confirmed that reduced regional cerebral blood flow relates to poorer cognition in adults with type 2 diabetes. More recently, Bracko et al. (2021) confirmed the crucial role that a reduction in cerebral blood flow plays in Alzheimer’s disease.

The pathways from depression to dementia

Depression is associated with a reduction in cerebral blood flow

Multiple studies have shown that cerebral blood flow is reduced in the setting of depression. Popa-Wagner et al. (2015) eloquently described in 2015 how dysfunction of cerebral autoregulation in aging can impair CBF and increase susceptibility to hypoxia and ischemia. Using arterial spin labeling, Cooper et al. (2020) compared cerebral blood flow (CBF) between 164 individuals suffering from major depression and 94 healthy controls. They reported reduced CBF in the right parahippocampus, thalamus, fusiform, and middle temporal gyri along with bilateral insula regions in depressed patients compared to controls. This confirmed the results obtained by Meyer et al. (1973) who showed that in severe depression there is bilateral hemispheric reduction of CBF. Takano et al. (2006) reported that the regional CBF (rCBF) in depressed patients was decreased compared to normal controls in widespread areas including the frontal lobe and limbic regions such as the cingulate cortex and parahippocampal gyrus. Oda et al. (2003) showed in depressed patients that rCBF in the frontal lobe, temporal lobe, and anterior cingulate gyrus were reduced regardless of the presence of subcortical hyperintensities. Where there was MRI hyperintensity, however, patients displayed reduced rCBF in the thalamus, basal ganglia, and brainstem a long with the cortical areas. In addition, the white matter hyperintensity scale was negatively correlated with rCBF in subcortical brain regions, such as the thalamus and right basal ganglia (Oda et al., 2003).

Depression also modifies vascular risk factors (Hakim, 2011). The effect of emotions on heart and blood vessel function was investigated. It was found that sadness created a distinctive pattern, showing slight increases in blood pressure and vascular resistance, and a reduction in the pumping capacity of the heart (Sinha et al., 1992). Thus, the reduction in CBF seen in depressed individuals may partially be the result of the impact of sadness on vascular risk factors.

Cerebral small vessel disease is evident in depressed individuals

Depression is as immense a risk factor for small vessel disease as high blood pressure (Wang et al., 2014). People who are depressed have abnormalities in the same brain regions known to be at risk for the development of small covert white matter strokes (Aizenstein et al., 2011). Greater depressive symptoms, after adjusting for sociodemographic, behavioral, and vascular risk factor variables, are correlated with smaller cerebral parenchymal fraction (β [95% confidence interval] = −0.56 [−1.05 to −0.07], p = 0.02) and increased odds of subclinical brain infarcts (odds ratio [95% confidence interval] = 1.55 [1.00–2.42], p = 0.05) (Zeki Al Hazzouri et al., 2018). Although this publication concludes that increased symptoms of depression were not significantly linked to white matter hyper-intensity volume, numerous magnetic resonance imaging (MRI) investigations have demonstrated that late life depression is related to the increased prevalence of white matter hyper-intensities on MRI (Krishnan et al., 2004; Taylor et al., 2005, 2013; Chen et al., 2006; Herrmann et al., 2008; Firbank et al., 2012).

The role of inflammation in depression

Possibly the most important mechanism associating depression with cognitive decline is how the immune system responds to persistent depression. This topic was extensively covered recently in the excellent article by Dafsari and Jessen (2020), where it was concluded that depressed patients exhibit chronic inflammation. Elevations in the interleukin system and tissue necrosis factor (TNFα) and C-reactive protein (CRP) have been reported in depressed patients, frequently associated with a simultaneous decrease in anti-inflammatory regulation (Dowlati et al., 2010; Felger and Lotrich, 2013). Miller and Raison (2016) concluded from meta-analyses that the most consistent biomarkers of inflammation in patients with depression were the peripheral blood interleukins, such as interleukin (IL)-1β, IL-6, TNFα, and CRP. Polymorphisms in inflammatory cytokine genes have been linked to depression and the individual’s response to therapy. These polymorphisms are for genes such as IL-Iβ, TNFα, and CRP (Bufalino et al., 2013). Other genes that have been linked to depression come from meta-analyses of genome-wide association studies and are associated with the immune system’s response to pathogens (Raison and Miller, 2013). It has been shown that if non-depressed persons are given inflammatory cytokines such as IFNα the onset of signs of depression occurs (Reichenberg et al., 2001; Bonaccorso et al., 2002; Capuron et al., 2002; Harrison et al., 2009). In addition, if cytokines like TNFα, or components of the inflammatory signaling pathway like cyclooxygenase 2, can be reduced, then the signs of depression can be reduced in individuals with various medical conditions such as rheumatoid arthritis, psoriasis and cancer, and major depressive disorder (Tyring et al., 2006; Köhler et al., 2014; Abbott et al., 2015). These findings highlight that depression has a large influence on the inflammatory pathways.

The impacts of treating depression

Depression can be managed by a variety of means, including psychotherapy, electroconvulsive therapy, and anti-depressant medication. This review will focus on the more prevalent therapy, namely oral antidepressants.

There is considerable debate on the impact of treating depression on the subsequent development of dementia. Coupland et al. (2019) presented evidence from a case-control study in which they reported that anticholinergic antidepressants such as Paxil and other tricyclic antidepressants may actually increase rather than decrease the risk of subsequent Alzheimer’s disease. Similar negative impact on cognition is attributed to SSRI’s (Wang et al., 2016). Consequently, when faced with a patient suffering from depression, the possibility of offering psychotherapy rather than medical therapy should be considered, and in the latter case, the impact of the specific drug being considered on dementia risk should be reviewed before it is prescribed.

The literature describes a number of physiological impacts attributed to the use of antidepressants.

On inflammation

Findings suggest that some antidepressants possess significant anti-inflammatory properties (Tynan et al., 2012; Walker, 2013; Jeon and Kim, 2017). Along with their impact on the cells of the peripheral immune system, selective serotonin reuptake inhibitors (SSRIs) can limit microglial and astroglial inflammatory processes (Dafsari and Jessen, 2020). As an example, fluoxetine causes the downregulation of genes involved in the pro-inflammatory response pathways such as the activation of IL-6 signaling and nuclear factor kappa b (NF-κb) signaling, and of TNFα signaling-related molecules (Patrício et al., 2015). Further, the dopamine enhancer bupropion inhibits pro-inflammatory cytokine production and lowers production of TNFα and interferon γ in mice (Brustolim et al., 2006). Researchers have shown that SSRIs (e.g., sertraline, fluoxetine, and paroxetine) likely inhibit microglial TNFα and nitrous oxide production. In mixed glial cell cultures, serotonin, and norepinephrine reuptake inhibitors (SNRIs) such as the MAO inhibitor moclobemide and selective noradrenaline reuptake inhibitors are anti-inflammatory (Vollmar et al., 2008; Bielecka et al., 2010). The reduction in neuroinflammation resulting from the noradrenaline reuptake inhibitor was also able to partially restore microglial function (Heneka et al., 2015). Popa-Wagner et al. (2014) suggested in 2014 that inflammation may be one pathophysiologic mechanism that contributes to treatment resistance in depression.

The effects of antidepressant treatment on microglial activation in patients with MDD were studied by using 18F-FEPPA PET. It revealed that the longer patients went without treatment, the greater the microglial activation was. However, if the patients were given antidepressants, the increase in microglial activation was no longer observed (Setiawan et al., 2018). The anticholinergic effects of some tricyclic antidepressant drugs have been shown to raise the risk of dementia possibly through accelerated glial transition to a neurodegenerative phenotype (Gamage et al., 2020).

Thus, there is a clear link for both depression and dementia to the inflammatory process, and the ability of any antidepressant approach or treatment to moderate the inflammatory load may be key to its success in reducing dementia.

Other physiological impacts of antidepressants that have been reported to date include.

On cerebral hemodynamics

Bench et al. (1995) performed early scans of patients suffering from depression and then rescanned the same patients following treatment with an antidepressant medication. They found that recovery from depression was linked to increases in rCBF flow in the same areas in which focal decreases in this parameter were described in the depressed state compared with normal subjects. Similar findings in another study described patients with depression having reduced blood flow to the left frontal brain region. However, with the antidepressant medication venlafaxine, the blood flow was restored (Navarro et al., 2004). Ishizaki et al. (2008) showed that following pharmacotherapy rCBF improved remarkably in the left dorsolateral medial prefrontal cortex (PFC) and the right parietooccipital regions while decreased CBF in some other regions of the PFC did not significantly improve. In a sample of older patients, Wei et al. (2018) reported that rCBF increases were linked to reductions in depressive symptoms. This led the authors to state that their observations were consistent with the vascular depression hypothesis in late-life depression.

On incidence of dementia

Antidepressant treatment may reduce cognitive decline (Mossello et al., 2008). It is estimated that the incidence of dementia would decline by 4% in the population if antidepressant treatment is applied (Mossello et al., 2008). Bartels et al. (2020) sought to determine the result of antidepressant drug classes on the risk for developing dementia using multiple treatment intervals. The researchers analyzed data of 62,317 individuals with an incident dementia diagnosis who were included in the German Disease Analyzer database and compared outcomes to those of controls matched by age and sex. They conducted logistic regression analyses, which were adjusted for health insurance status and comorbid diseases linked to dementia or antidepressant treatment, to evaluate the links between dementia incidence and treatment with four major classes of antidepressant drug, as well as 14 of the most commonly prescribed individual antidepressants. Results showed an association between treatment for 2 years or longer with any antidepressant and a lower risk for dementia among 17 of 18 comparisons. Particularly for long-term treatment, tricyclic antidepressants were linked to a reduction in the incidence of dementia. Long-term treatment with escitalopram (OR = 0.66; 95% CI, 0.5–0.89) was associated with the lowest risk for dementia on an individual antidepressant basis.

However, it is important to emphasize that antidepressant medications have a host of risks, contradictions and side effects that must be considered for each individual prior to starting treatment. As has been stated, some antidepressants may be ineffective or even have negative effects on cognition (Lu and Tune, 2003; Wang et al., 2016; Moraros et al., 2017).

Summary and conclusion

This review highlights that depression is a risk factor for dementia and details the physiological steps that link depression to its negative cognitive function. These steps begin with activation of inflammatory mediators, followed by a decline in the density of cerebral small vessels, which then leads to a drop in cerebral blood flow. This sequence is evident in the brains of depressed individuals when they are still cognitively normal but predicts the eventual decline in memory function.

The multifaceted physiological consequences of depression described here conform to the already outlined pattern for other recognized risks for dementia such as obesity, sedentary lifestyle and inadequate sleep (Hakim, 2021). The major difference between depression and the other cognitive risk factors is the possibility of offering various therapeutic modalities to the affected individuals.

This review cautions, however, that some antidepressant medications may worsen the cognitive impact of depression and recommends a careful evaluation of the proposed therapy on the subsequent decline in cognitive function. In addition, the use of social supports to reduce cognitive decline and depression should also be considered. Reducing social isolation has been shown to potentially delay the onset of dementia (Xiang et al., 2021). Positive social support was shown to reduce the risk of dementia whereas negative support increased the risk among persons aged 50 years and over (Khondoker et al., 2017). Overall, high quality social relationships appear to be important for overall cognitive health and can also reduce depression in older people (Murata et al., 2017).

With the identification of the proposed intermediary steps to link depression and other risk factors to cognitive decline, research can focus on identifying and neutralizing the inflammatory mediators, with the goal of interrupting the negative impact they have on cognitive function.

Author contributions

AH reviewed the literature, wrote the manuscript, and approved the submitted version.

Acknowledgments

The author would like to acknowledge his gratitude to the University of Ottawa Brain and Mind Research Institute and to Sarah Schock, for their support and contributions in the production of this manuscript.

Conflict of interest

The author declares that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

Abbott, R., Whear, R., Nikolaou, V., Bethel, A., Coon, J. T., Stein, K., et al. (2015). Tumour necrosis factor-α inhibitor therapy in chronic physical illness: a systematic review and meta-analysis of the effect on depression and anxiety. J. Psychosom. Res. 79, 175–184. doi: 10.1016/j.jpsychores.2015.04.008

PubMed Abstract | CrossRef Full Text | Google Scholar

Aizenstein, H. J., Andreescu, C., Edelman, K. L., Cochran, J. L., Price, J., Butters, M. A., et al. (2011). fMRI correlates of white matter hyperintensities in late-life depression. Am. J. Psychiatry 168, 1075–1082. doi: 10.1176/appi.ajp.2011.10060853

PubMed Abstract | CrossRef Full Text | Google Scholar

Almeida, O. P., Hankey, G. J., Yeap, B. B., Golledge, J., and Flicker, L. (2017). Depression as a modifiable factor to decrease the risk of dementia. Transl. Psychiatry 7:e1117. doi: 10.1038/tp.2017.90

PubMed Abstract | CrossRef Full Text | Google Scholar

Bangen, K. J., Werhane, M. L., Weigand, A. J., Edmonds, E. C., Delano-Wood, L., Thomas, K. R., et al. (2018). Reduced regional cerebral blood flow relates to poorer cognition in older adults with type 2 diabetes. Front. Aging Neurosci. 10:270. doi: 10.3389/fnagi.2018.00270

PubMed Abstract | CrossRef Full Text | Google Scholar

Bartels, C., Belz, M., Vogelgsang, J., Hessmann, P., Bohlken, J., Wiltfang, J., et al. (2020). To be continued? Long-term treatment effects of antidepressant drug classes and individual antidepressants on the risk of developing dementia: a german case-control study. J. Clin. Psychiatry 81:19m13205. doi: 10.4088/JCP.19m13205

PubMed Abstract | CrossRef Full Text | Google Scholar

Bench, C. J., Frackowiak, R. S. J., and Dolan, R. J. (1995). Changes in regional cerebral blood flow on recovery from depression. Psychol. Med. 25, 247–261. doi: 10.1017/S0033291700036151

PubMed Abstract | CrossRef Full Text | Google Scholar

Bielecka, A. M., Paul-Samojedny, M., and Obuchowicz, E. (2010). Moclobemide exerts anti-inflammatory effect in lipopolysaccharide-activated primary mixed glial cell culture. Naunyn Schmiedebergs Arch. Pharmacol. 382, 409–417. doi: 10.1007/s00210-010-0535-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Bonaccorso, S., Marino, V., Puzella, A., Pasquini, M., Biondi, M., Artini, M., et al. (2002). Increased depressive ratings in patients with hepatitis C receiving interferon-α-based immunotherapy are related to interferon-α-induced changes in the serotonergic system. J. Clin. Psychopharmacol. 22, 86–90. doi: 10.1097/00004714-200202000-00014

PubMed Abstract | CrossRef Full Text | Google Scholar

Bracko, O., Cruz Hernández, J. C., Park, L., Nishimura, N., and Schaffer, C. B. (2021). Causes and consequences of baseline cerebral blood flow reductions in Alzheimer’s disease. J. Cereb. Blood Flow Metab. 41, 1501–1516. doi: 10.1177/0271678X20982383

PubMed Abstract | CrossRef Full Text | Google Scholar

Brustolim, D., Ribeiro-dos-Santos, R., Kast, R. E., Altschuler, E. L., and Soares, M. B. P. (2006). A new chapter opens in anti-inflammatory treatments: the antidepressant bupropion lowers production of tumor necrosis factor-alpha and interferon-gamma in mice. Int. Immunopharmacol. 6, 903–907. doi: 10.1016/j.intimp.2005.12.007

PubMed Abstract | CrossRef Full Text | Google Scholar

Bufalino, C., Hepgul, N., Aguglia, E., and Pariante, C. M. (2013). The role of immune genes in the association between depression and inflammation: a review of recent clinical studies. Brain. Behav. Immun. 31, 31–47. doi: 10.1016/j.bbi.2012.04.009

PubMed Abstract | CrossRef Full Text | Google Scholar

Capuron, L., Gumnick, J. F., Musselman, D. L., Lawson, D. H., Reemsnyder, A., Nemeroff, C. B., et al. (2002). Neurobehavioral effects of interferon-α in cancer patients: phenomenology and paroxetine responsiveness of symptom dimensions. Neuropsychopharmacology 26, 643–652. doi: 10.1016/S0893-133X(01)00407-9

CrossRef Full Text | Google Scholar

Chen, P. S., McQuoid, D. R., Payne, M. E., and Steffens, D. C. (2006). White matter and subcortical gray matter lesion volume changes and late-life depression outcome: a 4-year magnetic resonance imaging study. Int. Psychogeriatr. 18, 445–456. doi: 10.1017/S1041610205002796

PubMed Abstract | CrossRef Full Text | Google Scholar

Comijs, H. C., Van Exel, E., Van Der Mast, R. C., Paauw, A., Oude Voshaar, R., and Stek, M. L. (2013). Childhood abuse in late-life depression. J. Affect Disord. 147, 241–246. doi: 10.1016/j.jad.2012.11.010

PubMed Abstract | CrossRef Full Text | Google Scholar

Compton, W. (2006). Changes in the prevalence of major depression and comorbid substance use disorders in the United States between 1991–1992 and 2001–2002. Am. J. Psychiatry 163, 2141–2147. doi: 10.1176/appi.ajp.163.12.2141

CrossRef Full Text | Google Scholar

Cooper, C. M., Chin Fatt, C. R., Liu, P., Grannemann, B., Carmody, T., Almeida, J. R. C., et al. (2020). Discovery and replication of cerebral blood flow differences in major depressive disorder. Mol. Psychiatry 25, 1500–1510. doi: 10.1038/s41380-019-0464-7

PubMed Abstract | CrossRef Full Text | Google Scholar

Coupland, C. A. C., Hill, T., Dening, T., Morriss, R., Moore, M., and Hippisley-Cox, J. (2019). Anticholinergic drug exposure and the risk of dementia. JAMA Intern. Med. 179, 1084–1093. doi: 10.1001/jamainternmed.2019.0677

PubMed Abstract | CrossRef Full Text | Google Scholar

Crous-Bou, M., Minguillón, C., Gramunt, N., and Molinuevo, J. L. (2017). Alzheimer’s disease prevention: from risk factors to early intervention. Alzheimers Res. Ther. 9:71. doi: 10.1186/s13195-017-0297-z

PubMed Abstract | CrossRef Full Text | Google Scholar

Dafsari, F. S., and Jessen, F. (2020). Depression—an underrecognized target for prevention of dementia in Alzheimer’s disease. Transl. Psychiatry 10:160. doi: 10.1038/s41398-020-0839-1

PubMed Abstract | CrossRef Full Text | Google Scholar

Dowlati, Y., Herrmann, N., Swardfager, W., Liu, H., Sham, L., Reim, E. K., et al. (2010). A meta-analysis of cytokines in major depression. Biol. Psychiatry 67, 446–457. doi: 10.1016/j.biopsych.2009.09.033

PubMed Abstract | CrossRef Full Text | Google Scholar

Eisenberger, N. I., Lieberman, M. D., and Williams, K. D. (2003). Does rejection hurt? An fMRI study of social exclusion. Science 302, 290–292. doi: 10.1126/science.1089134

PubMed Abstract | CrossRef Full Text | Google Scholar

Felger, J. C., and Lotrich, F. E. (2013). Inflammatory cytokines in depression: neurobiological mechanisms and therapeutic implications. Neuroscience 246, 199–229. doi: 10.1016/j.neuroscience.2013.04.060

PubMed Abstract | CrossRef Full Text | Google Scholar

Firbank, M. J., Teodorczuk, A., Van Der Flier, W. M., Gouw, A. A., Wallin, A., Erkinjuntti, T., et al. (2012). Relationship between progression of brain white matter changes and late-life depression: 3-Year results from the LADIS study. Br. J. Psychiatry 201, 40–45. doi: 10.1192/bjp.bp.111.098897

PubMed Abstract | CrossRef Full Text | Google Scholar

Gamage, R., Wagnon, I., Rossetti, I., Childs, R., Niedermayer, G., Chesworth, R., et al. (2020). Cholinergic modulation of glial function during aging and chronic neuroinflammation. Front. Cell Neurosci. 14:577912. doi: 10.3389/fncel.2020.577912

PubMed Abstract | CrossRef Full Text | Google Scholar

Gatchel, J. R., Rabin, J. S., Buckley, R. F., Locascio, J. J., Quiroz, Y. T., Yang, H. S., et al. (2019). Longitudinal association of depression symptoms with cognition and cortical amyloid among community-dwelling older adults. JAMA Netw. Open 2:e198964. doi: 10.1001/jamanetworkopen.2019.8964

PubMed Abstract | CrossRef Full Text | Google Scholar

Gautrin, D., Froda, S., Tetreault, H., and Gauvreau, D. (1990). Canadian projections of cases suffering from Alzheimer’s disease and senile dementia of Alzheimer type oer the period 1986-2031. Can. J. Psychiatry 35, 162–165. doi: 10.1177/070674379003500211

PubMed Abstract | CrossRef Full Text | Google Scholar

GBD 2019 Dementia Forecasting Collaborators (2022). Estimation of the global prevalence of dementia in 2019 and forecasted prevalence in 2050: an analysis for the Global Burden of Disease Study 2019. Lancet Public Health 7, e105–e125. doi: 10.1016/S2468-2667(21)00249-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Hagnell, O. (1989). Repeated incidence and prevalence studies of mental disorders in a total population followed during 25 years the Lundby Study, Sweden. Acta Psychiatr. Scand. 79, 61–77. doi: 10.1111/j.1600-0447.1989.tb05216.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Hagnell, O., Ojesjo, L., Otterbeck, L., and Rorsman, B. (1993). Prevalence of mental disorders, personality traits and mental complaints in the Lundby Study: a point prevalence study of the 1957 Lundby cohort of 2612 inhabitants of a geographically defined area who were re-examined in 1972 regardless of domicile. Scand. J. Soc. Med. Suppl. 21, 1–77.

PubMed Abstract | Google Scholar

Hakim, A. M. (2011). Depression, strokes and dementia: new biological insights into an unfortunate pathway. Cardiovasc. Psychiatry Neurol. 2011:649629. doi: 10.1155/2011/649629

PubMed Abstract | CrossRef Full Text | Google Scholar

Hakim, A. M. (2021). A Proposed hypothesis on dementia: inflammation, small vessel disease, and hypoperfusion is the sequence that links all harmful lifestyles to cognitive impairment. Front. Aging Neurosci. 13:679837. doi: 10.3389/fnagi.2021.679837

PubMed Abstract | CrossRef Full Text | Google Scholar

Harrison, N. A., Brydon, L., Walker, C., Gray, M. A., Steptoe, A., and Critchley, H. D. (2009). Inflammation causes mood changes through alterations in subgenual cingulate activity and mesolimbic connectivity. Biol. Psychiatry 66, 407–414. doi: 10.1016/j.biopsych.2009.03.015

PubMed Abstract | CrossRef Full Text | Google Scholar

Heneka, M. T., Carson, M. J., El Khoury, J., Landreth, G. E., Brosseron, F., Feinstein, D. L., et al. (2015). Neuroinflammation in Alzheimer’s disease. Lancet Neurol. 14, 388–405. doi: 10.1016/S1474-4422(15)70016-5

CrossRef Full Text | Google Scholar

Herrmann, L. L., Le Masurier, M., and Ebmeier, K. P. (2008). White matter hyperintensities in late life depression: a systematic review. J. Neurol. Neurosurg. Psychiatry 79, 619–624. doi: 10.1136/jnnp.2007.124651

PubMed Abstract | CrossRef Full Text | Google Scholar

Hidaka, B. H. (2012). Depression as a disease of modernity: explanations for increasing prevalence. J. Affect. Disord. 140, 205–214. doi: 10.1016/j.jad.2011.12.036

PubMed Abstract | CrossRef Full Text | Google Scholar

Ishizaki, J., Yamamoto, H., Takahashi, T., Takeda, M., Yano, M., and Mimura, M. (2008). Changes in regional cerebral blood flow following antidepressant treatment in late-life depression. Int. J. Geriatr. Psychiatry 23, 805–811. doi: 10.1002/gps.1980

PubMed Abstract | CrossRef Full Text | Google Scholar

Iturria-Medina, Y., Sotero, R. C., Toussaint, P. J., Mateos-Pérez, J. M., Evans, A. C., and The Alzheimer’s Disease Neuroimaging Initiative (2016). Early role of vascular dysregulation on late-onset Alzheimer’s disease based on multifactorial data-driven analysis. Nat. Commun. 7:11934. doi: 10.1038/ncomms11934

PubMed Abstract | CrossRef Full Text | Google Scholar

Jeon, S. W., and Kim, Y. K. (2017). Inflammation-induced depression: its pathophysiology and therapeutic implications. J. Neuroimmunol. 313, 92–98. doi: 10.1016/j.jneuroim.2017.10.016

PubMed Abstract | CrossRef Full Text | Google Scholar

Khondoker, M., Rafnsson, S. B., Morris, S., Orrell, M., and Steptoe, A. (2017). Positive and negative experiences of social support and risk of dementia in later life: an investigation using the english longitudinal study of ageing. J. Alzheimers Dis. 58, 99–108. doi: 10.3233/JAD-161160

PubMed Abstract | CrossRef Full Text | Google Scholar

Köhler, O. E., Benros, M., Nordentoft, M., Farkouh, M. E., Iyengar, R. L., Mors, O., et al. (2014). Effect of anti-inflammatory treatment on depression, depressive symptoms, and adverse effects a systematic review and meta-analysis of randomized clinical trials. JAMA Psychiatry 71, 1381–1391. doi: 10.1001/jamapsychiatry.2014.1611

PubMed Abstract | CrossRef Full Text | Google Scholar

Köhler, S., Van Boxtel, M. P. J., Van Os, J., Thomas, A. J., O’Brien, J. T., Jolles, J., et al. (2010). Depressive symptoms and cognitive decline in community-dwelling older adults. J. Am. Geriatr. Soc. 58, 873–879. doi: 10.1111/j.1532-5415.2010.02807.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Krishnan, K. R. R., Taylor, W. D., McQuoid, D. R., MacFall, J. R., Payne, M. E., Provenzale, J. M., et al. (2004). Clinical characteristics of magnetic resonance imaging-defined subcortical ischemic depression. Biol. Psychiatry 55, 390–397. doi: 10.1016/j.biopsych.2003.08.014

PubMed Abstract | CrossRef Full Text | Google Scholar

Lu, C. J., and Tune, L. E. (2003). Chronic exposure to anticholinergic medications adversely affects the course of Alzheimer disease. Am. J. Geriatr. Psychiatry 11, 458–461. doi: 10.1097/00019442-200307000-00009

CrossRef Full Text | Google Scholar

Meyer, J. S., Shimazu, K., Fukuuchi, Y., Ouchi, T., Okamoto, S., Koto, A., et al. (1973). Impaired neurogenic cerebrovascular control and dysautoregulation after stroke. Stroke 4, 169–186. doi: 10.1161/01.STR.4.2.169

CrossRef Full Text | Google Scholar

Miller, A. H., and Raison, C. L. (2016). The role of inflammation in depression: from evolutionary imperative to modern treatment target. Nat. Rev. Immunol. 16, 22–34. doi: 10.1038/nri.2015.5

PubMed Abstract | CrossRef Full Text | Google Scholar

Moraros, J., Nwankwo, C., Patten, S. B., and Mousseau, D. D. (2017). The association of antidepressant drug usage with cognitive impairment or dementia, including Alzheimer disease: a systematic review and meta-analysis. Depress. Anxiety 34, 217–226. doi: 10.1002/da.22584

PubMed Abstract | CrossRef Full Text | Google Scholar

Mossello, E., Boncinelli, M., Caleri, V., Cavallini, M. C., Palermo, E., Di Bari, M., et al. (2008). Is antidepressant treatment associated with reduced cognitive decline in Alzheimer’s disease? Dement. Geriatr. Cogn. Disord. 25, 372–379. doi: 10.1159/000121334

PubMed Abstract | CrossRef Full Text | Google Scholar

Murata, C., Saito, T., Tsuji, T., Saito, M., and Kondo, K. (2017). A 10-year follow-up study of social ties and functional health among the old: the AGES project. Int. J. Environ. Res. Public Health 14:717. doi: 10.3390/ijerph14070717

PubMed Abstract | CrossRef Full Text | Google Scholar

Muscatell, K. A., Dedovic, K., Slavich, G. M., Jarcho, M. R., Breen, E. C., Bower, J. E., et al. (2015). Greater amygdala activity and dorsomedial prefrontal-amygdala coupling are associated with enhanced inflammatory responses to stress. Brain Behav. Immun. 43, 46–53. doi: 10.1016/j.bbi.2014.06.201

PubMed Abstract | CrossRef Full Text | Google Scholar

Navarro, V., Gastó, C., Lomeña, F., Torres, X., Mateos, J. J., Portella, M. J., et al. (2004). Prognostic value of frontal functional neuroimaging in late-onset severe major depression. Br. J. Psychiatry 184, 306–311. doi: 10.1192/bjp.184.4.306

PubMed Abstract | CrossRef Full Text | Google Scholar

Norton, S., Matthews, F. E., Barnes, D. E., Yaffe, K., and Brayne, C. (2014). Potential for primary prevention of Alzheimer’s disease: an analysis of population-based data. Lancet Neurol. 13, 788–794. doi: 10.1016/S1474-4422(14)70136-X

CrossRef Full Text | Google Scholar

Oda, K., Okubo, Y., Ishida, R., Murata, Y., Ohta, K., Matsuda, T., et al. (2003). Regional cerebral blood flow in depressed patients with white matter magnetic resonance hyperintensity. Biol. Psychiatry 53, 150–156. doi: 10.1016/S0006-3223(02)01548-2

CrossRef Full Text | Google Scholar

Panksepp, J. (2003). Feeling the pain of social loss. Science 302, 237–239. doi: 10.1126/science.1091062

PubMed Abstract | CrossRef Full Text | Google Scholar

Patrício, P., Mateus-Pinheiro, A., Irmler, M., Alves, N. D., Machado-Santos, A. R., Morais, M., et al. (2015). Differential and converging molecular mechanisms of antidepressants’ action in the hippocampal dentate gyrus. Neuropsychopharmacology 40, 338–349. doi: 10.1038/npp.2014.176

PubMed Abstract | CrossRef Full Text | Google Scholar

Popa-Wagner, A., Buga, A. M., Popescu, B., and Muresanu, D. (2015). Vascular cognitive impairment, dementia, aging and energy demand. A vicious cycle. J. Neural Transm. 122, S47–S54. doi: 10.1007/s00702-013-1129-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Popa-Wagner, A., Buga, A. M., Tica, A. A., and Albu, C. V. (2014). Perfusion deficits, inflammation and aging precipitate depressive behaviour. Biogerontology 15, 439–448. doi: 10.1007/s10522-014-9516-1

PubMed Abstract | CrossRef Full Text | Google Scholar

Raison, C. L., and Miller, A. H. (2013). The evolutionary significance of depression in Pathogen Host Defense (PATHOS-D). Mol. Psychiatry 18, 15–37. doi: 10.1038/mp.2012.2

PubMed Abstract | CrossRef Full Text | Google Scholar

Reichenberg, A., Yirmiya, R., Schuld, A., Kraus, T., Haack, M., Morag, A., et al. (2001). Cytokine-associated emotional and cognitive disturbances in humans. Arch. Gen. Psychiatry 58, 445–452. doi: 10.1001/archpsyc.58.5.445

PubMed Abstract | CrossRef Full Text | Google Scholar

Setiawan, E., Attwells, S., Wilson, A. A., Mizrahi, R., Rusjan, P. M., Miler, L., et al. (2018). Association of translocator protein total distribution volume with duration of untreated major depressive disorder: a cross-sectional study. Lancet Psychiatry 5, 339–347. doi: 10.1016/S2215-0366(18)30048-8

CrossRef Full Text | Google Scholar

Sinha, R., Lovallo, W. R., and Parsons, O. A. (1992). Cardiovascular differentiation of emotions. Psychosom. Med. 54, 422–435. doi: 10.1097/00006842-199207000-00005

PubMed Abstract | CrossRef Full Text | Google Scholar

Slavich, G. M., and Irwin, M. R. (2014). From stress to inflammation and major depressive disorder: a social signal transduction theory of depression. Psychol. Bull. 140, 774–815. doi: 10.1037/a0035302

PubMed Abstract | CrossRef Full Text | Google Scholar

Steffens, D. C., Welsh-Bohmer, K. A., Burke, J. R., Plassman, B. L., Beyer, J. L., Gersing, K. R., et al. (2004). Methodology and preliminary results from the neurocognitive outcomes of depression in the elderly study. J. Geriatr. Psychiatry Neurol. 17, 202–211. doi: 10.1177/0891988704269819

PubMed Abstract | CrossRef Full Text | Google Scholar

Takano, H., Kato, M., Inagaki, A., Watanabe, K., and Kashima, H. (2006). Time course of cerebral blood flow changes following electroconvulsive therapy in depressive patients - Measured at 3 time points using single photon emission computed tomography. Keio J. Med. 55, 153–160. doi: 10.2302/kjm.55.153

PubMed Abstract | CrossRef Full Text | Google Scholar

Taylor, W. D., MacFall, J. R., Payne, M. E., McQuoid, D. R., Steffens, D. C., Provenzale, J. M., et al. (2005). Greater MRI lesion volumes in elderly depressed subjects than in control subjects. Psychiatry Res. Neuroimaging 139, 1–7. doi: 10.1016/j.pscychresns.2004.08.004

PubMed Abstract | CrossRef Full Text | Google Scholar

Taylor, W. D., Zhao, Z., Ashley-Koch, A., Payne, M. E., Steffens, D. C., Krishnan, R. R., et al. (2013). Fiber tract-specific white matter lesion severity Findings in late-life depression and by AGTR1 A1166C genotype. Hum. Brain Mapp. 34, 295–303. doi: 10.1002/hbm.21445

PubMed Abstract | CrossRef Full Text | Google Scholar

Twenge, J. M., Gentile, B., DeWall, C. N., Ma, D., Lacefield, K., and Schurtz, D. R. (2010). Birth cohort increases in psychopathology among young Americans, 1938-2007: a cross-temporal meta-analysis of the MMPI. Clin. Psychol. Rev. 30, 145–154. doi: 10.1016/j.cpr.2009.10.005

PubMed Abstract | CrossRef Full Text | Google Scholar

Tynan, R. J., Weidenhofer, J., Hinwood, M., Cairns, M. J., Day, T. A., and Walker, F. R. (2012). A comparative examination of the anti-inflammatory effects of SSRI and SNRI antidepressants on LPS stimulated microglia. Brain Behav. Immun. 26, 469–479. doi: 10.1016/j.bbi.2011.12.011

PubMed Abstract | CrossRef Full Text | Google Scholar

Tyring, S., Gottlieb, A., Papp, K., Gordon, K., Leonardi, C., Wang, A., et al. (2006). Etanercept and clinical outcomes, fatigue, and depression in psoriasis: double-blind placebo-controlled randomised phase III trial. Lancet 367, 29–35. doi: 10.1016/S0140-6736(05)67763-X

PubMed Abstract | CrossRef Full Text | Google Scholar

Villarroel, M. A., and Terlizzi, E. P. (2020). Symptoms of Depression Among Adults: United States, 2019. Hyattsville: NCHS Data Brief.

Google Scholar

Vollmar, P., Haghikia, A., Dermietzel, R., and Faustmann, P. M. (2008). Venlafaxine exhibits an anti-inflammatory effect in an inflammatory co-culture model. Int. J. Neuropsychopharmacol. 11, 111–117. doi: 10.1017/S1461145707007729

PubMed Abstract | CrossRef Full Text | Google Scholar

Walker, F. R. (2013). A critical review of the mechanism of action for the selective serotonin reuptake inhibitors: do these drugs possess anti-inflammatory properties and how relevant is this in the treatment of depression? Neuropharmacology 67, 304–317. doi: 10.1016/j.neuropharm.2012.10.002

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, C., Gao, S., Hendrie, H. C., Kesterson, J., Campbell, N. L., Shekhar, A., et al. (2016). Antidepressant use in the elderly is associated with an increased risk of dementia. Alzheimer Dis. Assoc. Disord. 30, 99–104. doi: 10.1097/WAD.0000000000000103

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, L., Leonards, C. O., Sterzer, P., and Ebinger, M. (2014). White matter lesions and depression: a systematic review and meta-analysis. J. Psychiatr. Res. 56, 56–64. doi: 10.1016/j.jpsychires.2014.05.005

PubMed Abstract | CrossRef Full Text | Google Scholar

Watt, D. F., and Panksepp, J. (2009). Depression: an evolutionarily conserved mechanism to terminate separation distress? a review of aminergic, peptidergic, and neural network perspectives. Neuropsychoanalysis 11, 7–15. doi: 10.1080/15294145.2009.10773593

CrossRef Full Text | Google Scholar

Wei, W., Karim, H. T., Lin, C., Mizuno, A., Andreescu, C., Karp, J. F., et al. (2018). Trajectories in cerebral blood flow following antidepressant treatment in late-life depression: support for the vascular depression hypothesis. J. Clin. Psychiatry 79:18m12106. doi: 10.4088/JCP.18m12106

PubMed Abstract | CrossRef Full Text | Google Scholar

Wittenberg, R., Hu, B., Jagger, C., Kingston, A., Knapp, M., Comas-Herrera, A., et al. (2020). Projections of care for older people with dementia in England: 2015 to 2040. Age Ageing 49, 264–269. doi: 10.1093/ageing/afz154

PubMed Abstract | CrossRef Full Text | Google Scholar

Xiang, X., Lai, P. H. L., Bao, L., Sun, Y., Chen, J., Dunkle, R. E., et al. (2021). Dual trajectories of social isolation and dementia in older adults: a population-based longitudinal study. J. Aging Health 33, 63–74. doi: 10.1177/0898264320953693

PubMed Abstract | CrossRef Full Text | Google Scholar

Zeki Al Hazzouri, A., Caunca, M. R., Nobrega, J. C., Nobrega, J. C., Elfassy, T., Cheung, Y. K., et al. (2018). Greater depressive symptoms, cognition, and markers of brain aging: Northern Manhattan Study. Neurology 90, e2077–e2085. doi: 10.1212/WNL.0000000000005639

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: depression, dementia, inflammation, cerebral small vessel disease, treatment, risk factors

Citation: Hakim A (2022) Perspectives on the complex links between depression and dementia. Front. Aging Neurosci. 14:821866. doi: 10.3389/fnagi.2022.821866

Received: 24 November 2021; Accepted: 05 August 2022;
Published: 24 August 2022.

Edited by:

Kristy A. Nielson, Marquette University, United States

Reviewed by:

Aurel Popa-Wagner, University of Medicine and Pharmacy of Craiova, Romania

Copyright © 2022 Hakim. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Antoine Hakim, ahakim@toh.ca

Download