Next Article in Journal
Exploratory Analysis of Autophagy–Lysosomal Pathway Proteins in Dermal Fibroblasts as Potential Peripheral Biomarkers for Alzheimer’s Disease: A Pilot Study
Previous Article in Journal
Correction: Belfeki et al. State of the Art of Cardiac Amyloidosis. Biomedicines 2023, 11, 1045
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Perspective

From Monoamines to Systems Psychiatry: Rewiring Depression Science and Care (1960s–2025)

Danube Neuroscience Research Laboratory, HUN-REN-SZTE Neuroscience Research Group, Hungarian Research Network, University of Szeged (HUN-REN-SZTE), Tisza Lajos krt. 113, H-6725 Szeged, Hungary
Biomedicines 2026, 14(1), 35; https://doi.org/10.3390/biomedicines14010035
Submission received: 1 December 2025 / Revised: 16 December 2025 / Accepted: 21 December 2025 / Published: 23 December 2025
(This article belongs to the Section Neurobiology and Clinical Neuroscience)

Abstract

Major depressive disorder (MDD) was long framed as a single clinical entity arising from a linear stress–monoamine–hypothalamic–pituitary–adrenal (HPA) axis cascade. This view was shaped by forced swim and learned helplessness tests in animals and by short-term symptom-based trials using scales such as the Hamilton Depression Rating Scale (HAM-D) and the Montgomery–Åsberg Depression Rating Scale (MADRS). This “unitary cascade” view has been dismantled by advances in neuroimaging, immune–metabolic profiling, sleep phenotyping, and plasticity markers, which reveal divergent circuit-level, inflammatory, and chronobiological patterns across anxiety-linked, pain-burdened, and cognitively weighted depressive presentations, all characterized by high rates of non-response and relapse. Translationally, face-valid rodent assays that equated immobility with despair have yielded limited bedside benefit, whereas cross-species bridges—electroencephalography (EEG) motifs, rapid eye movement (REM) architecture, effort-based reward tasks, and inflammatory/metabolic panels—are beginning to provide mechanistically grounded, clinically actionable readouts. In current practice, depression care is shifting toward systems psychiatry: inflammation-high and metabolic-high archetypes, anhedonia- and circadian-dominant subgroups, formal treatment-resistant depression (TRD) staging, connectivity-guided neuromodulation, esketamine, selected pharmacogenomic panels, and early digital phenotyping, as endpoints broaden to functioning and durability. A central gap is that heterogeneity is acknowledged but rarely built into trial design or implementation. This perspective advances a plasticity-centered systems psychiatry in which a testable prediction is that manipulating defined prefrontal–striatal and prefrontal–limbic circuits in sex-balanced, chronic-stress models will reproduce human network-defined biotypes and treatment response, and proposes hybrid effectiveness–implementation platforms that embed immune–metabolic and sleep panels, circuit-sensitive tasks, and digital monitoring under a shared, preregistered data standard.

1. Framing the Journey—Prompts

Major depressive disorder (MDD) has long been framed through a monoamine-centric lens, a narrative that catalyzed pharmacotherapy yet left substantial non-response and relapse [1]. Historical reassessments of the monoamine hypothesis and contemporary work on clinical, neurobiological, and treatment advances now converge on a simple conclusion: incremental refinements of the same model are no longer sufficient [2,3]. In an era of high-dimensional neuroimaging, immune profiling, and digital phenotyping, revisiting the trajectory from 1960s monoamine theories to contemporary systems psychiatry is therefore not merely historical, but strategically necessary [4,5] (Figure 1).
This perspective views MDD as a nexus condition with dense crosstalk with anxiety, chronic pain, and dementia, where shared and distinct pathways blur categorical boundaries [6]. Large-scale cohort and imaging studies reveal common yet dissociable alterations in prefrontal and limbic circuitry across depression, anxiety, post-traumatic stress disorder, and pain, while mechanistic work highlights bidirectional links with cardiometabolic and neurodegenerative disease [7,8,9]. Within this landscape, anxious, pain-enriched, and cognitively loaded depression exemplify how comorbidity patterns reshape prognosis, treatment response, and trial outcomes [10,11].
Our central thread traces the shift from neurotransmitter-centered framework toward an integrated immune, metabolic, and circuit-based framework that can anchor precision care [12]. This perspective synthesizes evidence on central–peripheral immune interactions, network-level biotypes, and model-driven subtypes of brain organization, and outlines how these advances should inform next-generation therapeutics and trial design [13,14,15]. Rather than providing another unitary model of depression, this perspective advances frameworks that treat heterogeneity as signal, not noise, enabling stratified, personalized interventions across intertwined spectra of mood, anxiety, pain, and cognitive decline [16] (Box 1).
Box 1. Key Terms.
A biomarker is an objective biological signal that indexes state, risk, or treatment response. An endophenotype reflects a heritable, mechanistic intermediate—closer to circuitry than symptoms. Anhedonia captures reduced reward sensitivity or effort, a core transdiagnostic deficit. Treatment-resistant denotes limited response after adequate therapeutic trials. Rapid-acting refers to interventions producing meaningful change within hours to days. A network target is a circuit-defined node guiding drug or device precision. A digital endpoint leverages passive or task-based sensors to quantify behavior in real time.

2. Early Paradigms and Assumptions (1960s–1990s)

Early paradigms of depression research became largely organized around stress, monoamines, and the hypothalamic–pituitary–adrenal axis as the primary translators of adversity into “behavioral despair” [2,17]. Forced swim and learned helplessness tests, later complemented by chronic stress paradigms in rodents and primates, operationalized immobility, withdrawal, and subdued exploration as proxies for hopelessness [18,19]. These models were highly sensitive to monoaminergic and hypothalamic–pituitary–adrenal (HPA) axis-modulating drugs and, for a time, appeared to offer a direct bridge between synaptic mechanisms and clinical symptoms [20,21]. Parallel work on glucocorticoid feedback, neurosteroids, and early immune and gut–brain signals refined this picture, yet still treated depression largely as a unitary endpoint of a relatively linear stress–monoamine–hypothalamic–pituitary–adrenal (HPA) axis cascade [22,23].
On the clinical side, the introduction of DSM III and DSM III R reconfigured nosology around symptom checklists, while the Hamilton Depression Rating Scale (HAM-D), and later the Montgomery–Åsberg Depression Rating Scale (MADRS), scores became the dominant trial endpoints [24,25]. Response and remission were quantified by cutoffs on these scales, and short, fixed-duration trials with single primary mood outcomes became the gold standard for regulatory sensitive evidence [26,27]. Placebo trials in phases, designed to screen out early improvers and sharpen drug signal, further shaped effect sizes and sample composition, often at the cost of ecological validity [26,28]. Translation across trials relied on scale equivalence rather than deeper phenotyping or mechanistic anchors [29,30].
Within this framework, crucial sources of heterogeneity were marginalized [31]. Sex- and age-dependent profiles of mood, anxiety, and somatic symptoms were rarely modeled explicitly in either preclinical or clinical research, despite emerging data on sex-specific monoamine and HPA signatures [32,33,34,35]. Ancestry, social context, chronic pain, and subthreshold anxiety were treated as noise, not structure [36]. An overreliance on narrow, short-term symptom endpoints, focused on short-term mood changes in narrowly defined samples, left longer-term trajectories, functional outcomes, and comorbidity patterns largely uncharted, planting many of the seeds for the replication, generalization, and treatment-resistance challenges that later decades would confront [37,38,39].

3. Key Mechanistic Pivots in Systems Psychiatry

This section outlines ten mechanistic domains that collectively illustrate the transition from monoamine-centered models of depression to a multiscale systems psychiatry framework, with each subsection integrating a defined biological mechanism with its translational implications. Each section focuses on a specific mechanistic targe—plasticity and circuit dynamics, reward and stress alignment, immune–metabolic and genomic milieu, or multi-point intervention logic—and links it to concrete translational readouts. Together, these pivots outline how synapses, circuits, body-wide signals, and trial architectures can be jointly tuned to redesign treatment-resistant depression (TRD) as a tractable, stratified systems problem.

3.1. Plasticity and Circuit Control of Depressive States

Across these cards, depression science shifts from monoamine scarcity toward dynamic systems. Each mini-anchor foregrounds mechanisms, actionable readouts, and clinically tractable targets that scaffold next-generation translational trial designs.

3.1.1. Synaptic Plasticity and Intrinsic Excitability

At the most proximal level, synaptic plasticity and intrinsic excitability reframe depression as a disorder of experience-dependent circuit wiring rather than as a simple transmitter shortage [40,41]. Long-term potentiation and depression, AMPA receptor throughput, and evoked excitatory postsynaptic potentials index rapid synaptic remodeling, while neurogenesis and ERK-sensitive priming, including ketamine-induced effects, extend plasticity across longer timescales [42,43,44,45]. Importantly, early electroencephalography, local field potentials, resting-state functional magnetic resonance imaging, and AMPA-forward assays provide convergent translational readouts that anchor plasticity as a measurable and clinically actionable target [46,47].

3.1.2. Glutamate/γ–Aminobutyric Acid (GABA) Microcircuit Control

Building on synaptic plasticity, glutamate and γ-aminobutyric acid microcircuits define how excitation–inhibition balance is locally implemented within cortical networks. Somatostatin- and parvalbumin-expressing interneurons tune affective set points by shaping oscillatory dynamics and information flow across layers [48,49,50,51]. Within this framework, N-methyl-D-aspartate (NMDA) and metabotropic glutamate receptor modulation, inhibitory postsynaptic potentials, and layer-specific oscillations emerge as key mechanistic levers [52,53,54]. Translationally, oscillation-anchored endpoints, task-locked transcranial magnetic stimulation–electroencephalography, and interneuron-specific computational or animal models link microcircuit dysfunction to scalable EEG and behavioral signatures [55,56].

3.1.3. Circuit-Level Nodes (Drugs and Devices)

At the network scale, microcircuit and plasticity deficits converge onto identifiable circuit-level nodes that are now directly targeted by drugs and devices [57,58]. Networks linking the habenula, subcallosal cingulate, and ventromedial prefrontal cortex serve as intervention points for deep brain stimulation, vagus nerve stimulation, and transcranial magnetic stimulation, including accelerated theta-burst protocols guided by connectivity-informed targeting and TMS-EEG physiology [59,60]. Importantly, proof of target engagement, optimized dosing schedules, and responder enrichment based on baseline network topology redefine how device parameters translate into durable clinical outcomes, moving neuromodulation toward precision deployment [61,62].

3.2. Reward, Motivation, and Stress Systems

Where plasticity and microcircuits define how affective states are encoded, reward and stress systems determine why motivation collapses and fatigue persists in many depressive phenotypes. Stress exposure reshapes reward valuation and effort allocation, biasing behavior toward withdrawal and anergia rather than adaptive pursuit. This pivot integrates mesolimbic reward processing with stress-responsive neuromodulatory and endocrine systems, positioning anhedonia and fatigue as system-level failures rather than isolated symptoms.

3.2.1. Reward, Motivation, and Stress–Opioid Tone

At the core of motivational dysfunction, anhedonia reflects disrupted mesolimbic signaling in which dopamine-driven reward processing is gated by dynorphin-mediated κ-opioid stress tone [63,64]. Stress-induced activation of κ-opioid receptors suppresses phasic dopamine release, shifting behavior away from effortful reward-seeking and toward passive coping [65,66,67,68]. Translationally, effort-based choice tasks, ventral striatal activity, and inflammatory status provide convergent readouts of motivational collapse, while selective κ-opioid antagonists and κ-opioid-modulating antidepressants offer mechanistically targeted strategies to dissociate hedonic capacity from stress-induced motivational inhibition [69,70].

3.2.2. HPA–Circadian–Stress Axis

Motivational and reward deficits are further shaped by coupling between the hypothalamic–pituitary–adrenal axis and circadian timekeeping systems. Glucocorticoid receptor sensitivity, cortisol rhythmicity, and sleep architecture jointly tune emotional regulation and energy allocation across the day [71,72,73,74]. Disrupted dim-light melatonin onset, altered actigraphy, and a dysregulated cortisol awakening response signal internal misalignment that amplifies fatigue, negative affect, and relapse vulnerability [75,76,77]. In this context, chronotype-aware protocols, sleep-linked endpoints, and glucocorticoid receptor assays operationalize stress–circadian dysfunction as a modifiable treatment dimension rather than background noise [78,79].

3.3. Immune–Metabolic–Genomic Modifiers of Risk and Treatment Response

If synaptic and circuit mechanisms define how depressive states are encoded and reward–stress systems explain their functional expression, immune, metabolic, and genomic modifiers determine the biological context in which these processes unfold. Peripheral inflammation, metabolic load, and transcriptional regulation quietly bias neural plasticity, reward sensitivity, and treatment durability, transforming background physiology into a primary determinant of symptom profile and therapeutic response. This pivot reframes systemic signals not as epiphenomena, but as modulators that shape both risk and responsiveness across depressive subtypes.

3.3.1. Tryptophan (Trp)–Kynurenine (KYN) Steering

Immune activation redirects tryptophan metabolism toward the KYN pathway, coupling peripheral inflammatory states to central neurotransmission and plasticity disruption [80,81]. Indoleamine 2,3-dioxygenase (IDO) and tryptophan 2,3-dioxygenase (TDO) shift kynurenine balance toward neuroactive metabolites, altering glutamatergic tone, microglial activation, and stress sensitivity [77,78,79,80]. Clinically, C-reactive protein (CRP) levels, KYN/Trp ratios, and quinolinic (QA)-to-kynurenic acid (KYNA) balance align with anhedonia, cognitive slowing, and fatigue-dominant symptom clusters, supporting inflammation-stratified trial designs and mechanistically guided adjunctive interventions in treatment-resistant depression [81,82,83,84,85,86,87].

3.3.2. Neuroimmune and Glia

Beyond metabolic steering, neuroimmune signaling positions microglia and astrocytes as active regulators of synaptic strength and network tone. Complement pathways, colony-stimulating factor 1 receptor, and purinergic P2X7 signaling sculpt synaptic pruning, excitability, and circuit stability under inflammatory conditions [82,83,84]. Translationally, positron emission tomography ligands, cerebrospinal cytokine panels, and electroencephalographic complexity measures provide convergent markers of central immune engagement [85,86,87,88]. Importantly, incorporating sex, baseline inflammatory state, and stimulation-induced immune shifts as design variables reframes immune modulation as a tractable dimension for precision psychiatry [89].

3.3.3. Metabolic–Endocrine Crosstalk

Metabolic–endocrine factors further constrain neural resilience by coupling energy regulation to mood and cognition. Insulin resistance, adiposity, and glucagon-like peptide-1 (GLP-1) signaling alter brain insulin sensitivity, reward processing, and response to antidepressants and neuromodulation [90,91,92]. Metabolic syndrome and type 2 diabetes (T2D) thus identify patient subgroups in whom standard treatments show reduced efficacy and durability [93,94]. Pragmatic trial designs that embed metabolic stratification and functional endpoints reposition metabolic load as a modifiable determinant rather than a comorbidity to be controlled away [90,95].

3.3.4. Epigenetic/Transcriptional Gating

At the slowest timescale, epigenetic and transcriptional mechanisms stabilize depressive states and constrain recovery trajectories. Histone deacetylases, bromodomain proteins, lysine-specific demethylase 1, and DNA methyltransferases regulate chromatin accessibility and gene expression programs linked to stress sensitivity and plasticity [96,97]. Cell-type-resolved ATAC and RNA sequencing, paired with peripheral chromatin markers, tracks medication- and neuromodulation-induced state transitions [98,99,100]. Incorporating these measures into longitudinal trials supports durability-focused designs that distinguish transient symptom relief from sustained circuit reconfiguration [97,99,100].

3.4. Multi-Point Precision Strategies and Emerging Targets

As plasticity, reward–stress dynamics, and immune–metabolic context converge, treatment-resistant depression emerges not as a failure of single targets, but as a systems-level mismatch between intervention and patient-specific biology. This perspective reframes precision psychiatry as the strategic coordination of multiple levers—pharmacological, neuromodulatory, psychotherapeutic, and digital—aligned with defined biotypes rather than syndromic averages.

Multi-Point Strategies and Next-Wave Targets

Multi-point strategies treat treatment-resistant depression as overlapping, partially independent biotypes that require coordinated intervention across synaptic, circuit, immune, and behavioral domains [101,102,103,104]. Biomarker-informed sequencing, pharmacogenomic guidance, and biotype-specific combinations replace linear, trial-and-error escalation with adaptive logic that exploits mechanistic complementarity [101,105,106,107]. Translationally, adaptive platform trials, N-of-1 designs, and digitally enriched endpoints allow for rapid iteration while preserving individual-level inference, shifting evaluation from short-term symptom reduction toward durability, functioning, and real-world effectiveness [102,103,104,107].

4. Divergence → Reconnection

As monoamine and stress-based models began to fray, a new translational strategy sought bridges that could genuinely scale across species rather than relying on face validity alone [108]. Electrophysiological motifs, such as frontal alpha asymmetry and reward related event-related potentials, emerged as candidate intermediate phenotypes that can be measured in rodents and humans using homologous paradigms [109,110,111]. Sleep architecture, particularly REM density and slow wave fragmentation, offered another shared metric, mirrored by alterations in stress-exposed animals and patients with melancholic or TRD [112,113,114]. In parallel, probabilistic reward tasks and cognitive control batteries were adapted across rodents, non-human primates, and humans, while longitudinal inflammatory and endocrine panels were integrated into both preclinical and clinical protocols, enabling richer cross-species modeling of reward inflammation coupling [115,116].
However, the field also accumulated clear evidence of repeated failure to incorporate lessons from earlier methodological missteps [117]. The assumption that immobility equals despair persisted long after it became evident that forced swim behavior reflects a narrow coping style, not the multidimensional construct of depression [118,119]. Short-duration trials and brief stress paradigms continued to dominate despite growing evidence that synaptic, circuit, and immune remodeling unfold over weeks to months [120,121]. Single-endpoint thinking, usually centered on global depression scores or single behavioral tests, repeatedly obscured domain-specific gains and masked heterogeneity in trajectories [122,123].
The most promising advances have come from deliberate back-translation [124,125,126]. Symptom clusters and rating scale factors are first decomposed into Research Domain Criteria (RdoC)-like domains of negative valence, reward processing, arousal, and cognition, and are then re-instantiated as tractable tasks and physiological readouts in animals [125,127]. Multimodal biomarker programs now use human-defined phenotypes to guide the design of cross-species EEG batteries, reward and cognitive paradigms, and peripheral immune signatures [128,129]. In doing so, they begin to reconnect bench and bedside around shared dimensions rather than diagnostic labels alone [130,131] (Table 1).

5. Clinical Applications Today

Clinical applications of systems psychiatry are increasingly shifting beyond one-size-fits-all algorithms toward stratified care grounded in measurable biological and behavioral dimensions [132]. Depression is increasingly parsed into high-inflammation, metabolically burdened, anhedonia-dominant, and disrupted sleep or circadian profiles, overlaid on the formal staging of treatment resistance [133,134]. Elevated CRP altered neutrophil to lymphocyte ratios, and immune metabolic gene signatures delineate patients whose symptoms cluster around anergia and anhedonia, while sleep fragmentation and circadian misalignment identify another modifiable axis that cuts across stages of non-response [135]. Parallel work on pharmacogenomics, circuit-based biotypes, and age-specific EEG signatures is entering specialist clinics and early-phase trials [13,136,137].
A clear distinction now separates tools that are clinically ready from those that are almost ready [138]. Structured TRD staging, esketamine, neuromodulation devices, and several pharmacogenomic panels have achieved regulatory approval and are supported by comparative effectiveness data [106,139,140]. By contrast, multiplex inflammatory and metabolic panels, task-based cognitive emotional biomarkers, and digital phenotyping batteries remain largely in the candidate domain, often validated in carefully selected cohorts but not yet embedded in stepped care pathways [129,141].
Translating these advances into everyday practice faces substantial hurdles [142]. Access to neuromodulation, ketamine-based interventions, and genomic testing is uneven, constrained by cost, infrastructure, and workforce training [143]. Regulatory frameworks still privilege drug over device and algorithm, and reimbursement rarely rewards stratified assessment [144]. Without deliberate attention to equity, biologically enriched care risks deepening disparities, as populations with higher inflammatory burden, multimorbidity, and limited digital access may be the least likely to receive precision-guided interventions [145,146,147] (Figure 2).

6. What We Got Wrong/Right

Many of the field’s “failures” now look like solvable design problems rather than fatal errors [148,149]. Trial-and-error prescribing, short-term symptom-focused trials, and inflated remission estimates exposed what happens when heterogeneity is ignored and protocols drift [148,150,151]. The corrective principles are clearer now: implement decision support and data-driven personalization, redesign trials to prioritize long-term functioning and real-world effectiveness, and adhere strictly to pre-specified protocols with transparent reporting [152,153,154]. The concept of difficult-to-treat depression and consensus definitions of TRD are direct products of these lessons [104,155,156].
A second cluster of mistakes involved what and whom we chose to measure [157,158]. Overbroad diagnostic criteria without biomarkers, unclear psychotherapy “active ingredients,” late recognition of comorbidity, and neglect of patient preference and digital engagement all narrowed impact [148,159,160,161]. The responses are already reshaping practice [162]. Biomarker-informed and endophenotype-based frameworks, component-focused psychotherapy trials, integrated care models, shared decision tools, guided digital and blended interventions, and Delphi-based TRD guidelines collectively represent durable gains and a more realistic architecture for precision psychiatry [104,156,160,163].

7. Outlook

The next decade will be judged by whether systems psychiatry can be changed from an attractive narrative into falsifiable science [164,165]. Two preclinical predictions are within reach. First, that manipulating specific prefrontal–striatal and prefrontal–limbic circuits in sex-balanced, chronic stress models will yield biotypes that map onto human imaging, defined network patterns, and differential treatment response [166,167,168]. Second, that patient-derived cellular systems combined with multi-omic profiling will prospectively predict which immune and metabolic perturbations in animals reproduce the anhedonia-dominant, high-inflammation phenotypes observed in humans [135,169].
On the clinical side, trial design must shift in two concrete ways. Large, multivariate, cross-trial prediction studies should be used not only to detect average effects but to learn stable individual-level treatment rules [170,171,172]. In parallel, factorial and adaptive designs need to dissect psychotherapy and combined “active ingredients” for treatment, rather than testing monolithic packages [173,174,175,176].
A viable implementation pathway is hybrid effectiveness implementation research embedded in integrated care, where stratification tools, targeted pharmacotherapy, and digital monitoring are co-designed with health systems [126,177]. Underpinning all of this should be a shared data standard that harmonizes symptom networks, EEG and imaging markers, and trial metadata, with preregistered analytic pipelines and mandatory external validation [178,179].

8. Conclusions

In this perspective review, I propose plasticity-centered systems psychiatry as the key lens through which both model-building and bedside decisions can be restructured. Synaptic remodeling, circuit dynamics, immune–metabolic context, and sleep-dependent homeostasis are repositioned as explicit, testable levers for intervention rather than diffuse background processes. A concrete practice change is to implement structured, plasticity-informed care pathways that link symptom profiles to simple circuit-sensitive indices such as reward tasks, sleep and circadian metrics, and low-burden immune or metabolic panels. A parallel research change is to develop adaptive, multimodal platforms in which the ten subsequent papers systematically interrogate promising synaptic, microcircuit, network, immune, and chronobiological targets for depression treatment.

Funding

This work was supported by the HUN-REN Hungarian Research Network.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Acknowledgments

The graphical abstract and figures are created using biorender.com (accessed on 1 December 2025).

Conflicts of Interest

The author declares no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
5-CSRTTfive-choice serial reaction time task
AhRaryl hydrocarbon receptor
AMPAα-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid
ATACassay for transposase-accessible chromatin
BMIbody mass index
BOLDblood-oxygen-level–dependent
CANTABCambridge Neuropsychological Test Automated Battery
CRPC-reactive protein
CSF1Rcolony-stimulating factor 1 receptor
Dex/CRHdexamethasone/corticotropin-releasing hormone
DLMOdim light melatonin onset
DLPFCdorsolateral prefrontal cortex
DNMTsDNA methyltransferases
DSMDiagnostic and Statistical Manual of Mental Disorders
EEGelectroencephalography
EefRTEffort Expenditure for Rewards Task
eEPSPsevoked excitatory postsynaptic potentials
ERKextracellular signal-regulated kinase
EMGelectromyography
fMRIfunctional magnetic resonance imaging
GABAγ-aminobutyric acid
GLP-1glucagon-like peptide-1
GRglucocorticoid receptor
HAM-DHamilton Depression Rating Scale
HbA1cglycated hemoglobin
HDACshistone deacetylases
HOMA-IRhomeostatic model assessment of insulin resistance
HPAhypothalamic–pituitary–adrenal axis
IDOindoleamine 2,3-dioxygenase
IFN-αinterferon-alpha
IL-6interleukin-6
IPSPsinhibitory postsynaptic potentials
κkappa
KYNkynurenine
KYNAkynurenic acid
LFPlocal field potential
LPSlipopolysaccharide
LSD1lysine-specific demethylase 1
LTDlong-term depression
LTPlong-term potentiation
MADRSMontgomery–Åsberg Depression Rating Scale
MDDmajor depressive disorder
MEGmagnetoencephalography
NMDAN-methyl-D-aspartate
PETpositron emission tomography
PFCprefrontal cortex
QAquinolinic acid
RDoCResearch Domain Criteria
REMrapid eye movement
RNAribonucleic acid
rsfMRIresting-state functional magnetic resonance imaging
SCCsubcallosal cingulate cortex
T2Dtype 2 diabetes
TDOtryptophan 2,3-dioxygenase
TMStranscranial magnetic stimulation
TMS-EEGtranscranial magnetic stimulation–electroencephalography
TNFtumor necrosis factor
TRDtreatment-resistant depression
Trptryptophan
vmPFCventromedial prefrontal cortex
VNSvagus nerve stimulation

References

  1. Hirschfeld, R.M. History and evolution of the monoamine hypothesis of depression. J. Clin. Psychiatry 2000, 61, 4–6. [Google Scholar] [PubMed]
  2. Boku, S.; Nakagawa, S.; Toda, H.; Hishimoto, A. Neural basis of major depressive disorder: Beyond monoamine hypothesis. Psychiatry Clin. Neurosci. 2018, 72, 3–12. [Google Scholar] [CrossRef]
  3. Liu, B.; Liu, J.; Wang, M.; Zhang, Y.; Li, L. From Serotonin to Neuroplasticity: Evolvement of Theories for Major Depressive Disorder. Front. Cell Neurosci. 2017, 11, 305. [Google Scholar] [CrossRef]
  4. Spellman, T.; Liston, C. Toward Circuit Mechanisms of Pathophysiology in Depression. Am. J. Psychiatry 2020, 177, 381–390. [Google Scholar] [CrossRef]
  5. Fries, G.R.; Saldana, V.A.; Finnstein, J.; Rein, T. Molecular pathways of major depressive disorder converge on the synapse. Mol. Psychiatry 2023, 28, 284–297. [Google Scholar] [CrossRef] [PubMed]
  6. Brandl, F.; Weise, B.; Mulej Bratec, S.; Jassim, N.; Hoffmann Ayala, D.; Bertram, T.; Ploner, M.; Sorg, C. Common and specific large-scale brain changes in major depressive disorder, anxiety disorders, and chronic pain: A transdiagnostic multimodal meta-analysis of structural and functional MRI studies. Neuropsychopharmacology 2022, 47, 1071–1080. [Google Scholar] [CrossRef]
  7. Sindermann, L.; Redlich, R.; Opel, N.; Böhnlein, J.; Dannlowski, U.; Leehr, E.J. Systematic transdiagnostic review of magnetic-resonance imaging results: Depression, anxiety disorders and their co-occurrence. J. Psychiatr. Res. 2021, 142, 226–239. [Google Scholar] [CrossRef]
  8. Arnaud, A.M.; Brister, T.S.; Duckworth, K.; Foxworth, P.; Fulwider, T.; Suthoff, E.D.; Werneburg, B.; Aleksanderek, I.; Reinhart, M.L. Impact of Major Depressive Disorder on Comorbidities: A Systematic Literature Review. J. Clin. Psychiatry 2022, 83, 43390. [Google Scholar] [CrossRef]
  9. Tanaka, M.; Battaglia, S.; Liloia, D. Navigating Neurodegeneration: Integrating Biomarkers, Neuroinflammation, and Imaging in Parkinson’s, Alzheimer’s, and Motor Neuron Disorders. Biomedicines 2025, 13, 1045. [Google Scholar] [CrossRef]
  10. Berk, M.; Köhler-Forsberg, O.; Turner, M.; Penninx, B.; Wrobel, A.; Firth, J.; Loughman, A.; Reavley, N.J.; McGrath, J.J.; Momen, N.C.; et al. Comorbidity between major depressive disorder and physical diseases: A comprehensive review of epidemiology, mechanisms and management. World Psychiatry 2023, 22, 366–387. [Google Scholar] [CrossRef]
  11. Tanaka, M.; Battaglia, S. From Biomarkers to Behavior: Mapping the Neuroimmune Web of Pain, Mood, and Memory. Biomedicines 2025, 13, 2226. [Google Scholar] [CrossRef]
  12. Bottaccioli, A.G.; Bologna, M.; Bottaccioli, F. Rethinking Depression-Beyond Neurotransmitters: An Integrated Psychoneuroendocrineimmunology Framework for Depression’s Pathophysiology and Tailored Treatment. Int. J. Mol. Sci. 2025, 26, 2759. [Google Scholar] [CrossRef] [PubMed]
  13. Tozzi, L.; Zhang, X.; Pines, A.; Olmsted, A.M.; Zhai, E.S.; Anene, E.T.; Chesnut, M.; Holt-Gosselin, B.; Chang, S.; Stetz, P.C.; et al. Personalized brain circuit scores identify clinically distinct biotypes in depression and anxiety. Nat. Med. 2024, 30, 2076–2087. [Google Scholar] [CrossRef] [PubMed]
  14. Jiao, W.; Lin, J.; Deng, Y.; Ji, Y.; Liang, C.; Wei, S.; Jing, X.; Yan, F. The immunological perspective of major depressive disorder: Unveiling the interactions between central and peripheral immune mechanisms. J. Neuroinflamm. 2025, 22, 10. [Google Scholar] [CrossRef]
  15. Tanaka, M.; Battaglia, S. Dualistic Dynamics in Neuropsychiatry: From Monoaminergic Modulators to Multiscale Biomarker Maps. Biomedicines 2025, 13, 1456. [Google Scholar] [CrossRef]
  16. Drevets, W.C.; Wittenberg, G.M.; Bullmore, E.T.; Manji, H.K. Immune targets for therapeutic development in depression: Towards precision medicine. Nat. Rev. Drug Discov. 2022, 21, 224–244. [Google Scholar] [CrossRef]
  17. Tafet, G.E.; Nemeroff, C.B. The Links Between Stress and Depression: Psychoneuroendocrinological, Genetic, and Environmental Interactions. J. Neuropsychiatry Clin. Neurosci. 2016, 28, 77–88. [Google Scholar] [CrossRef]
  18. Yankelevitch-Yahav, R.; Franko, M.; Huly, A.; Doron, R. The forced swim test as a model of depressive-like behavior. J. Vis. Exp. 2015, 97, 52587. [Google Scholar] [CrossRef]
  19. Yan, H.C.; Cao, X.; Das, M.; Zhu, X.H.; Gao, T.M. Behavioral animal models of depression. Neurosci. Bull. 2010, 26, 327–337. [Google Scholar] [CrossRef]
  20. Dale, E.; Bang-Andersen, B.; Sánchez, C. Emerging mechanisms and treatments for depression beyond SSRIs and SNRIs. Biochem. Pharmacol. 2015, 95, 81–97. [Google Scholar] [CrossRef] [PubMed]
  21. Abelaira, H.M.; Réus, G.Z.; Quevedo, J. Animal models as tools to study the pathophysiology of depression. Braz. J. Psychiatry 2013, 35, S112–S120. [Google Scholar] [CrossRef]
  22. Sharma, S.; Chawla, S.; Kumar, P.; Ahmad, R.; Kumar Verma, P. The chronic unpredictable mild stress (CUMS) Paradigm: Bridging the gap in depression research from bench to bedside. Brain Res. 2024, 1843, 149123. [Google Scholar] [CrossRef]
  23. Ji, N.; Lei, M.; Chen, Y.; Tian, S.; Li, C.; Zhang, B. How Oxidative Stress Induces Depression? ASN Neuro 2023, 15, 17590914231181037. [Google Scholar] [CrossRef]
  24. Bech, P. Modern psychometrics in clinimetrics: Impact on clinical trials of antidepressants. Psychother. Psychosom. 2004, 73, 134–138. [Google Scholar] [CrossRef]
  25. Regier, D.A.; Narrow, W.E.; Kuhl, E.A.; Kupfer, D.J. The conceptual development of DSM-V. Am. J. Psychiatry 2009, 166, 645–650. [Google Scholar] [CrossRef] [PubMed]
  26. Hengartner, M.P.; Jakobsen, J.C.; Sørensen, A.; Plöderl, M. Efficacy of new-generation antidepressants assessed with the Montgomery-Asberg Depression Rating Scale, the gold standard clinician rating scale: A meta-analysis of randomised placebo-controlled trials. PLoS ONE 2020, 15, e0229381. [Google Scholar] [CrossRef] [PubMed]
  27. Khan, A.; Khan, S.R.; Shankles, E.B.; Polissar, N.L. Relative sensitivity of the Montgomery-Asberg Depression Rating Scale, the Hamilton Depression rating scale and the Clinical Global Impressions rating scale in antidepressant clinical trials. Int. Clin. Psychopharmacol. 2002, 17, 281–285. [Google Scholar] [CrossRef]
  28. Guizzaro, L.; Morgan, D.D.V.; Falco, A.; Gallo, C. Hamilton scale and MADRS are interchangeable in meta-analyses but can disagree at trial level. J. Clin. Epidemiol. 2020, 124, 106–117. [Google Scholar] [CrossRef] [PubMed]
  29. Leucht, S.; Fennema, H.; Engel, R.R.; Kaspers-Janssen, M.; Szegedi, A. Translating the HAM-D into the MADRS and vice versa with equipercentile linking. J. Affect. Disord. 2018, 226, 326–331. [Google Scholar] [CrossRef]
  30. Santi, N.S.; Biswal, S.B.; Naik, B.N.; Sahoo, J.P.; Rath, B. Comparison of Hamilton Depression Rating Scale and Montgomery-Åsberg Depression Rating Scale: Baked Straight from a Randomized Study. Cureus 2023, 15, e45098. [Google Scholar] [CrossRef]
  31. Ratheesh, A.; Berk, M.; Schmaal, L. Can we overcome the heterogeneity of mood disorders in clinical trials? Aust. N. Z. J. Psychiatry 2023, 57, 309–311. [Google Scholar] [CrossRef]
  32. Su, Z.; Yang, X.; Hou, J.; Liu, S.; Wang, Y.; Chen, Z. Gender differences in the co-occurrence of anxiety and depressive symptoms among early adolescents: A network approach. J. Psychiatr. Res. 2024, 179, 300–305. [Google Scholar] [CrossRef]
  33. Vasiliadis, H.M.; Desjardins, F.; Roberge, P.; Grenier, S. Sex Differences in Anxiety Disorders in Older Adults. Curr. Psychiatry Rep. 2020, 22, 75. [Google Scholar] [CrossRef]
  34. Thompson, K.N.; Hübel, C.; Cheesman, R.; Adey, B.N.; Armour, C.; Davies, M.R.; Hotopf, M.; Jones, I.R.; Kalsi, G.; McIntosh, A.M.; et al. Age and sex-related variability in the presentation of generalized anxiety and depression symptoms. Depress. Anxiety 2021, 38, 1054–1065. [Google Scholar] [CrossRef]
  35. Pavlidi, P.; Kokras, N.; Dalla, C. Sex Differences in Depression and Anxiety. Curr. Top. Behav. Neurosci. 2023, 62, 103–132. [Google Scholar] [CrossRef]
  36. Kuper, P.; Miguel, C.; Cuijpers, P.; Apfelbacher, C.; Buntrock, C.; Karyotaki, E.; Sprenger, A.A.; Harrer, M. Sample size and geographical region predict effect heterogeneity in psychotherapy research for depression: A meta-epidemiological study. J. Clin. Epidemiol. 2025, 183, 111779. [Google Scholar] [CrossRef] [PubMed]
  37. Cai, N.; Choi, K.W.; Fried, E.I. Reviewing the genetics of heterogeneity in depression: Operationalizations, manifestations and etiologies. Hum. Mol. Genet. 2020, 29, R10–R18. [Google Scholar] [CrossRef] [PubMed]
  38. Hickie, I.B.; Berk, M.; Scott, J.; Crouse, J.; Scott, E.; Wray, N.; Iorfino, F. What are the best strategies for stratification of clinical cohorts with depression and other mood disorders? Res. Dir. Depress. 2024, 1, e18. [Google Scholar] [CrossRef]
  39. Dunlop, K.; Grosenick, L.; Downar, J.; Vila-Rodriguez, F.; Gunning, F.M.; Daskalakis, Z.J.; Blumberger, D.M.; Liston, C. Dimensional and categorical solutions to parsing depression heterogeneity in a large single-site sample. Biol. Psychiatry 2024, 96, 422–434. [Google Scholar] [CrossRef]
  40. Marsden, W.N. Synaptic plasticity in depression: Molecular, cellular and functional correlates. Prog. Neuropsychopharmacol. Biol. Psychiatry 2013, 43, 168–184. [Google Scholar] [CrossRef] [PubMed]
  41. Shenoy, S.; Ibrahim, S. Perinatal Depression and the Role of Synaptic Plasticity in Its Pathogenesis and Treatment. Behav. Sci. 2023, 13, 942. [Google Scholar] [CrossRef]
  42. Brown, J.C.; Higgins, E.S.; George, M.S. Synaptic Plasticity 101: The Story of the AMPA Receptor for the Brain Stimulation Practitioner. Neuromodulation 2022, 25, 1289–1298. [Google Scholar] [CrossRef] [PubMed]
  43. He, J.G.; Zhou, H.Y.; Wang, F.; Chen, J.G. Dysfunction of Glutamatergic Synaptic Transmission in Depression: Focus on AMPA Receptor Trafficking. Biol. Psychiatry Glob. Open Sci. 2023, 3, 187–196. [Google Scholar] [CrossRef] [PubMed]
  44. Brager, D.H.; Johnston, D. Plasticity of intrinsic excitability during long-term depression is mediated through mGluR-dependent changes in I(h) in hippocampal CA1 pyramidal neurons. J. Neurosci. 2007, 27, 13926–13937. [Google Scholar] [CrossRef]
  45. Cavalleri, L.; Merlo Pich, E.; Millan, M.J.; Chiamulera, C.; Kunath, T.; Spano, P.F.; Collo, G. Ketamine enhances structural plasticity in mouse mesencephalic and human iPSC-derived dopaminergic neurons via AMPAR-driven BDNF and mTOR signaling. Mol. Psychiatry 2018, 23, 812–823. [Google Scholar] [CrossRef] [PubMed]
  46. Appelbaum, L.G.; Shenasa, M.A.; Stolz, L.; Daskalakis, Z. Synaptic plasticity and mental health: Methods, challenges and opportunities. Neuropsychopharmacology 2023, 48, 113–120. [Google Scholar] [CrossRef]
  47. Cantone, M.; Bramanti, A.; Lanza, G.; Pennisi, M.; Bramanti, P.; Pennisi, G.; Bella, R. Cortical Plasticity in Depression. ASN Neuro 2017, 9, 1759091417711512. [Google Scholar] [CrossRef]
  48. Hu, Y.T.; Tan, Z.L.; Hirjak, D.; Northoff, G. Brain-wide changes in excitation-inhibition balance of major depressive disorder: A systematic review of topographic patterns of GABA- and glutamatergic alterations. Mol. Psychiatry 2023, 28, 3257–3266. [Google Scholar] [CrossRef]
  49. Fee, C.; Banasr, M.; Sibille, E. Somatostatin-Positive Gamma-Aminobutyric Acid Interneuron Deficits in Depression: Cortical Microcircuit and Therapeutic Perspectives. Biol. Psychiatry 2017, 82, 549–559. [Google Scholar] [CrossRef]
  50. Kuki, T.; Fujihara, K.; Miwa, H.; Tamamaki, N.; Yanagawa, Y.; Mushiake, H. Contribution of parvalbumin and somatostatin-expressing GABAergic neurons to slow oscillations and the balance in beta-gamma oscillations across cortical layers. Front. Neural Circuits 2015, 9, 6. [Google Scholar] [CrossRef]
  51. Chen, G.; Zhang, Y.; Li, X.; Zhao, X.; Ye, Q.; Lin, Y.; Tao, H.W.; Rasch, M.J.; Zhang, X. Distinct Inhibitory Circuits Orchestrate Cortical beta and gamma Band Oscillations. Neuron 2017, 96, 1403–1418.e1406. [Google Scholar] [CrossRef]
  52. Tsuboi, D.; Nagai, T.; Yoshimoto, J.; Kaibuchi, K. Neuromodulator regulation and emotions: Insights from the crosstalk of cell signaling. Front. Mol. Neurosci. 2024, 17, 1376762. [Google Scholar] [CrossRef]
  53. Wagatsuma, N.; Nobukawa, S.; Fukai, T. A microcircuit model involving parvalbumin, somatostatin, and vasoactive intestinal polypeptide inhibitory interneurons for the modulation of neuronal oscillation during visual processing. Cereb. Cortex 2023, 33, 4459–4477. [Google Scholar] [CrossRef]
  54. Tahvili, F.; Vinck, M.; di Volo, M. PV and SOM cells play distinct causal roles in controlling network oscillations and stability. Cell Rep. 2025, 44, 116131. [Google Scholar] [CrossRef] [PubMed]
  55. Mazza, F.; Guet-McCreight, A.; Valiante, T.A.; Griffiths, J.D.; Hay, E. In-silico EEG biomarkers of reduced inhibition in human cortical microcircuits in depression. PLoS Comput. Biol. 2023, 19, e1010986. [Google Scholar] [CrossRef]
  56. Rademacher, J.; Grent-’t-Jong, T.; Rivolta, D.; Sauer, A.; Scheller, B.; Gonzalez-Burgos, G.; Metzner, C.; Uhlhaas, P.J. Computational modeling of ketamine-induced changes in gamma-band oscillations: The contribution of parvalbumin and somatostatin interneurons. PLoS Comput. Biol. 2025, 21, e1013118. [Google Scholar] [CrossRef]
  57. Deng, Z.D.; Luber, B.; Balderston, N.L.; Velez Afanador, M.; Noh, M.M.; Thomas, J.; Altekruse, W.C.; Exley, S.L.; Awasthi, S.; Lisanby, S.H. Device-Based Modulation of Neurocircuits as a Therapeutic for Psychiatric Disorders. Annu. Rev. Pharmacol. Toxicol. 2020, 60, 591–614. [Google Scholar] [CrossRef] [PubMed]
  58. Vinogradov, S.; Chafee, M.V.; Lee, E.; Morishita, H. Psychosis spectrum illnesses as disorders of prefrontal critical period plasticity. Neuropsychopharmacology 2023, 48, 168–185. [Google Scholar] [CrossRef] [PubMed]
  59. Idlett-Ali, S.L.; Salazar, C.A.; Bell, M.S.; Short, E.B.; Rowland, N.C. Neuromodulation for treatment-resistant depression: Functional network targets contributing to antidepressive outcomes. Front. Hum. Neurosci. 2023, 17, 1125074. [Google Scholar] [CrossRef]
  60. Allawala, A.; Bijanki, K.R.; Oswalt, D.; Mathura, R.K.; Adkinson, J.; Pirtle, V.; Shofty, B.; Robinson, M.; Harrison, M.T.; Mathew, S.J.; et al. Prefrontal network engagement by deep brain stimulation in limbic hubs. Front. Hum. Neurosci. 2023, 17, 1291315. [Google Scholar] [CrossRef]
  61. Tanaka, M.; He, Z.; Han, S.; Battaglia, S. Editorial: Noninvasive brain stimulation: A promising approach to study and improve emotion regulation. Front. Behav. Neurosci. 2025, 19, 1633936. [Google Scholar] [CrossRef]
  62. Wang, A.; Sun, J. Personalized EEG-guided brain stimulation targeting in major depression via network controllability and multi-objective optimization. BMC Psychiatry 2025, 25, 723. [Google Scholar] [CrossRef]
  63. Bruijnzeel, A.W. kappa-Opioid receptor signaling and brain reward function. Brain Res. Rev. 2009, 62, 127–146. [Google Scholar] [CrossRef]
  64. Wallace, C.W.; Holleran, K.M.; Slinkard, C.Y.; Centanni, S.W.; Lapish, C.C.; Jones, S.R. Kappa opioid receptors diminish spontaneous dopamine signals in awake mice through multiple mechanisms. Neuropharmacology 2025, 273, 110458. [Google Scholar] [CrossRef]
  65. Bekhbat, M.; Li, Z.; Dunlop, B.W.; Treadway, M.T.; Mehta, N.D.; Revill, K.P.; Lucido, M.J.; Hong, C.; Ashchi, A.; Wommack, E.C.; et al. Sustained effects of repeated levodopa (L-DOPA) administration on reward circuitry, effort-based motivation, and anhedonia in depressed patients with higher inflammation. Brain Behav. Immun. 2025, 125, 240–248. [Google Scholar] [CrossRef]
  66. Treadway, M.; Etuk, S.; Cooper, J.; Hossein, S.; Hahn, E.; Betters, S.; Liu, S.; Arulpragasam, A.; DeVries, B.; Irfan, N.; et al. A randomized proof-of-mechanism trial of TNF antagonism for motivational anhedonia and related corticostriatal circuitry in depressed patients with high inflammation. Res. Sq. 2024, 30, 1407–1417. [Google Scholar] [CrossRef]
  67. Felger, J.C.; Treadway, M.T. Inflammation Effects on Motivation and Motor Activity: Role of Dopamine. Neuropsychopharmacology 2017, 42, 216–241. [Google Scholar] [CrossRef]
  68. Lucido, M.J.; Bekhbat, M.; Goldsmith, D.R.; Treadway, M.T.; Haroon, E.; Felger, J.C.; Miller, A.H. Aiding and Abetting Anhedonia: Impact of Inflammation on the Brain and Pharmacological Implications. Pharmacol. Rev. 2021, 73, 1084–1117. [Google Scholar] [CrossRef] [PubMed]
  69. Hasbi, A.; Madras, B.K.; George, S.R. Daily Δ9-Tetrahydrocannabinol and Withdrawal Increase Dopamine D1-D2 Receptor Heteromer to Mediate Anhedonia- and Anxiogenic-like Behavior Through a Dynorphin and Kappa Opioid Receptor Mechanism. Biol. Psychiatry Glob. Open Sci. 2023, 3, 550–566. [Google Scholar] [CrossRef]
  70. Thomas, C.S.; Mohammadkhani, A.; Rana, M.; Qiao, M.; Baimel, C.; Borgland, S.L. Optogenetic stimulation of lateral hypothalamic orexin/dynorphin inputs in the ventral tegmental area potentiates mesolimbic dopamine neurotransmission and promotes reward-seeking behaviours. Neuropsychopharmacology 2022, 47, 728–740. [Google Scholar] [CrossRef] [PubMed]
  71. Albrecht, U. Molecular Mechanisms in Mood Regulation Involving the Circadian Clock. Front. Neurol. 2017, 8, 30. [Google Scholar] [CrossRef]
  72. Focke, C.M.B.; Iremonger, K.J. Rhythmicity matters: Circadian and ultradian patterns of HPA axis activity. Mol. Cell Endocrinol. 2020, 501, 110652. [Google Scholar] [CrossRef]
  73. Nader, N.; Chrousos, G.P.; Kino, T. Interactions of the circadian CLOCK system and the HPA axis. Trends Endocrinol. Metab. 2010, 21, 277–286. [Google Scholar] [CrossRef]
  74. Lightman, S.L.; Birnie, M.T.; Conway-Campbell, B.L. Dynamics of ACTH and Cortisol Secretion and Implications for Disease. Endocr. Rev. 2020, 41, bnaa002. [Google Scholar] [CrossRef] [PubMed]
  75. Carpenter, J.S.; Crouse, J.J.; Shin, M.; Tonini, E.; Hindmarsh, G.; de Haan, Z.; Iorfino, F.; Robillard, R.; Naismith, S.; Scott, E.M.; et al. Evidence for Internal Misalignment of Circadian Rhythms in Youth with Emerging Mood Disorders. J. Biol. Rhythm. 2025, 40, 424–440. [Google Scholar] [CrossRef]
  76. Skubic, C.; Zevnik, U.; Nahtigal, K.; Dolenc Grošelj, L.; Rozman, D. Circadian Biomarkers in Humans: Methodological Insights into the Detection of Melatonin and Cortisol. Biomolecules 2025, 15, 1006. [Google Scholar] [CrossRef] [PubMed]
  77. Schrire, Z.M.; Naismith, S.L.; Pye, J.; Duffy, S.L.; Gordon, C.J.; Lewis, S.J.; Hoyos, C.M. Circadian rhythms and misalignment in older adults and those ‘at risk’for dementia: A study examining correlates of dim-light melatonin onset: Biomarkers (non-neuroimaging)/Prognostic utility. Alzheimer’s Dement. 2020, 16, e045525. [Google Scholar] [CrossRef]
  78. Scott, M.R.; McClung, C.A. Circadian Rhythms in Mood Disorders. Adv. Exp. Med. Biol. 2021, 1344, 153–168. [Google Scholar] [CrossRef]
  79. Johnson, C.; Brzezynski, J.; Bair, A.; Smieszek, S.; Bai, H.; Polymeropoulos, C.; Birznieks, G.; Polymeropoulos, M. 0879 Dim Light Melatonin Onset in a Delayed Sleep-Wake Phase Disorder Cohort: Ongoing Clinical Trial Update. Sleep 2025, 48, A382. [Google Scholar] [CrossRef]
  80. Tanaka, M.; Tóth, F.; Polyák, H.; Szabó, Á.; Mándi, Y.; Vécsei, L. Immune Influencers in Action: Metabolites and Enzymes of the Tryptophan-Kynurenine Metabolic Pathway. Biomedicines 2021, 9, 734. [Google Scholar] [CrossRef]
  81. Badawy, A.A. Kynurenine Pathway of Tryptophan Metabolism: Regulatory and Functional Aspects. Int. J. Tryptophan Res. 2017, 10, 1178646917691938. [Google Scholar] [CrossRef]
  82. Naffaa, M.M. Mechanisms of astrocytic and microglial purinergic signaling in homeostatic regulation and implications for neurological disease. Explor. Neurosci. 2025, 4, 100676. [Google Scholar] [CrossRef]
  83. Bedetta, M.; Pizzo, P.; Lia, A. The Multifaceted Role of P2X7R in Microglia and Astrocytes. Neurochem. Res. 2025, 50, 239. [Google Scholar] [CrossRef]
  84. Batista, A.F.; Khan, K.A.; Papavergi, M.T.; Lemere, C.A. The Importance of Complement-Mediated Immune Signaling in Alzheimer’s Disease Pathogenesis. Int. J. Mol. Sci. 2024, 25, 817. [Google Scholar] [CrossRef]
  85. Liu, Y.D.; Chang, Y.H.; Xie, X.T.; Wang, X.Y.; Ma, H.Y.; Liu, M.C.; Zhang, H.M. PET Imaging Unveils Neuroinflammatory Mechanisms in Psychiatric Disorders: From Microglial Activation to Therapeutic Innovation. Mol. Neurobiol. 2025, 62, 15318–15335. [Google Scholar] [CrossRef]
  86. Werry, E.L.; Bright, F.M.; Piguet, O.; Ittner, L.M.; Halliday, G.M.; Hodges, J.R.; Kiernan, M.C.; Loy, C.T.; Kril, J.J.; Kassiou, M. Recent Developments in TSPO PET Imaging as A Biomarker of Neuroinflammation in Neurodegenerative Disorders. Int. J. Mol. Sci. 2019, 20, 3161. [Google Scholar] [CrossRef] [PubMed]
  87. Enache, D.; Pariante, C.M.; Mondelli, V. Markers of central inflammation in major depressive disorder: A systematic review and meta-analysis of studies examining cerebrospinal fluid, positron emission tomography and post-mortem brain tissue. Brain Behav. Immun. 2019, 81, 24–40. [Google Scholar] [CrossRef] [PubMed]
  88. Dupont, A.C.; Largeau, B.; Santiago Ribeiro, M.J.; Guilloteau, D.; Tronel, C.; Arlicot, N. Translocator Protein-18 kDa (TSPO) Positron Emission Tomography (PET) Imaging and Its Clinical Impact in Neurodegenerative Diseases. Int. J. Mol. Sci. 2017, 18, 785. [Google Scholar] [CrossRef] [PubMed]
  89. Meyer, J.H.; Cervenka, S.; Kim, M.J.; Kreisl, W.C.; Henter, I.D.; Innis, R.B. Neuroinflammation in psychiatric disorders: PET imaging and promising new targets. Lancet Psychiatry 2020, 7, 1064–1074. [Google Scholar] [CrossRef]
  90. Chen, W.; Kullmann, S.; Rhea, E.M. Expanding the understanding of insulin resistance in brain and periphery. Trends Endocrinol. Metab. 2025, online ahead of print. [Google Scholar] [CrossRef]
  91. Lanzillotta, S.; Rolfi, L.R.; Zulli, B.; Barone, E. Metabolic breakdown: Linking insulin resistance and mitochondrial dysfunction to neurodegeneration in Alzheimer’s disease. Neural Regen. Res. 2025, online ahead of print. [Google Scholar] [CrossRef]
  92. Detka, J.; Głombik, K. Insights into a possible role of glucagon-like peptide-1 receptor agonists in the treatment of depression. Pharmacol. Rep. 2021, 73, 1020–1032. [Google Scholar] [CrossRef] [PubMed]
  93. Hölscher, C. Brain insulin resistance: Role in neurodegenerative disease and potential for targeting. Expert. Opin. Investig. Drugs 2020, 29, 333–348. [Google Scholar] [CrossRef]
  94. Gault, V.A.; Hölscher, C. GLP-1 receptor agonists show neuroprotective effects in animal models of diabetes. Peptides 2018, 100, 101–107. [Google Scholar] [CrossRef]
  95. Cooper, D.H.; Ramachandra, R.; Ceban, F.; Di Vincenzo, J.D.; Rhee, T.G.; Mansur, R.B.; Teopiz, K.M.; Gill, H.; Ho, R.; Cao, B.; et al. Glucagon-like peptide 1 (GLP-1) receptor agonists as a protective factor for incident depression in patients with diabetes mellitus: A systematic review. J. Psychiatr. Res. 2023, 164, 80–89. [Google Scholar] [CrossRef]
  96. Baek, S.Y.; Lee, J.; Kim, T.; Lee, H.; Choi, H.S.; Park, H.; Koh, M.; Kim, E.; Jung, M.E.; Iliopoulos, D.; et al. Development of a novel histone deacetylase inhibitor unveils the role of HDAC11 in alleviating depression by inhibition of microglial activation. Biomed. Pharmacother. 2023, 166, 115312. [Google Scholar] [CrossRef] [PubMed]
  97. Du, J.; Liu, R.; Ma, L.; Liu, Y.; Wei, W.; Liu, N.; Cao, Q.; Yu, J. Novel histone deacetylase-5 inhibitor T2943 exerts an anti-depressive effect in mice by enhancing GRID1 expression. Sci. Rep. 2025, 15, 4522. [Google Scholar] [CrossRef] [PubMed]
  98. Sun, Z.; Zhang, B.; Zhou, J.; Luo, Y.; Zhu, X.; Wang, Y.; He, Y.; Zheng, P.; Zhang, L.; Yang, J.; et al. Integrated Single-Cell RNA-seq and ATAC-seq Reveals Heterogeneous Differentiation of CD4+ Naive T Cell Subsets is Associated with Response to Antidepressant Treatment in Major Depressive Disorder. Adv. Sci. 2024, 11, e2308393. [Google Scholar] [CrossRef]
  99. Taavitsainen, S.; Engedal, N.; Cao, S.; Handle, F.; Erickson, A.; Prekovic, S.; Wetterskog, D.; Tolonen, T.; Vuorinen, E.M.; Kiviaho, A.; et al. Single-cell ATAC and RNA sequencing reveal pre-existing and persistent cells associated with prostate cancer relapse. Nat. Commun. 2021, 12, 5307. [Google Scholar] [CrossRef]
  100. Grandi, F.C.; Modi, H.; Kampman, L.; Corces, M.R. Chromatin accessibility profiling by ATAC-seq. Nat. Protoc. 2022, 17, 1518–1552. [Google Scholar] [CrossRef]
  101. Kajumba, M.M.; Kakooza-Mwesige, A.; Nakasujja, N.; Koltai, D.; Canli, T. Treatment-resistant depression: Molecular mechanisms and management. Mol. Biomed. 2024, 5, 43. [Google Scholar] [CrossRef]
  102. Daeli, J.I.D.; Soemara, A. How Do Combined Pharmacological and Psychotherapeutic Interventions Impact Treatment Outcomes for Patients with Treatment-Resistant Major Depressive Disorder?: A Systematic Review. Int. J. Med. Sci. Health Res. 2025, 12, 49–99. [Google Scholar]
  103. Ruberto, V.L.; Jha, M.K.; Murrough, J.W. Pharmacological Treatments for Patients with Treatment-Resistant Depression. Pharmaceuticals 2020, 13, 116. [Google Scholar] [CrossRef] [PubMed]
  104. McIntyre, R.S.; Alsuwaidan, M.; Baune, B.T.; Berk, M.; Demyttenaere, K.; Goldberg, J.F.; Gorwood, P.; Ho, R.; Kasper, S.; Kennedy, S.H.; et al. Treatment-resistant depression: Definition, prevalence, detection, management, and investigational interventions. World Psychiatry 2023, 22, 394–412. [Google Scholar] [CrossRef]
  105. Amasi-Hartoonian, N.; Pariante, C.M.; Cattaneo, A.; Sforzini, L. Understanding treatment-resistant depression using “omics” techniques: A systematic review. J. Affect. Disord. 2022, 318, 423–455. [Google Scholar] [CrossRef]
  106. Garcia, G.; Cotner, C.; Spano, R. Precision medicine in psychiatry: Case series on pharmacogenomic solutions for treatment-resistant depression. J. Am. Assoc. Nurse Pract. 2025, 37, 471–476. [Google Scholar] [CrossRef]
  107. Baum, M.L.; Widge, A.S.; Carpenter, L.L.; McDonald, W.M.; Cohen, B.M.; Nemeroff, C.B. Pharmacogenomic Clinical Support Tools for the Treatment of Depression. Am. J. Psychiatry 2024, 181, 591–607. [Google Scholar] [CrossRef] [PubMed]
  108. Young, J.W. Development of cross-species translational paradigms for psychiatric research in the Research Domain Criteria era. Neurosci. Biobehav. Rev. 2023, 148, 105119. [Google Scholar] [CrossRef]
  109. Iturra-Mena, A.M.; Kangas, B.D.; Luc, O.T.; Potter, D.; Pizzagalli, D.A. Electrophysiological signatures of reward learning in the rodent touchscreen-based Probabilistic Reward Task. Neuropsychopharmacology 2023, 48, 700–709. [Google Scholar] [CrossRef] [PubMed]
  110. Cavanagh, J.F.; Olguin, S.L.; Talledo, J.A.; Kotz, J.E.; Roberts, B.Z.; Nungaray, J.A.; Sprock, J.; Gregg, D.; Bhakta, S.G.; Light, G.A.; et al. Amphetamine alters an EEG marker of reward processing in humans and mice. Psychopharmacology 2022, 239, 923–933. [Google Scholar] [CrossRef]
  111. Pizzagalli, D.A.; Sherwood, R.J.; Henriques, J.B.; Davidson, R.J. Frontal brain asymmetry and reward responsiveness: A source-localization study. Psychol. Sci. 2005, 16, 805–813. [Google Scholar] [CrossRef]
  112. Kheirkhah, M.; Duncan, W.C., Jr.; Yuan, Q.; Wang, P.R.; Jamalabadi, H.; Leistritz, L.; Walter, M.; Goldman, D.; Zarate, C.A., Jr.; Hejazi, N.S. REM density predicts rapid antidepressant response to ketamine in individuals with treatment-resistant depression. Neuropsychopharmacology 2025, 50, 941–946. [Google Scholar] [CrossRef]
  113. Pesonen, A.K.; Gradisar, M.; Kuula, L.; Short, M.; Merikanto, I.; Tark, R.; Räikkönen, K.; Lahti, J. REM sleep fragmentation associated with depressive symptoms and genetic risk for depression in a community-based sample of adolescents. J. Affect. Disord. 2019, 245, 757–763. [Google Scholar] [CrossRef]
  114. Lechinger, J.; Koch, J.; Weinhold, S.L.; Seeck-Hirschner, M.; Stingele, K.; Kropp-Näf, C.; Braun, M.; Drews, H.J.; Aldenhoff, J.; Huchzermeier, C.; et al. REM density is associated with treatment response in major depression: Antidepressant pharmacotherapy vs. psychotherapy. J. Psychiatr. Res. 2021, 133, 67–72. [Google Scholar] [CrossRef]
  115. Luc, O.T.; Kangas, B.D. Validation of a touchscreen probabilistic reward task for mice: A reverse-translated assay with cross-species continuity. Cogn. Affect. Behav. Neurosci. 2024, 24, 281–288. [Google Scholar] [CrossRef]
  116. Palmer, D.; Dumont, J.R.; Dexter, T.D.; Prado, M.A.M.; Finger, E.; Bussey, T.J.; Saksida, L.M. Touchscreen cognitive testing: Cross-species translation and co-clinical trials in neurodegenerative and neuropsychiatric disease. Neurobiol. Learn. Mem. 2021, 182, 107443. [Google Scholar] [CrossRef]
  117. Molendijk, M.L.; de Kloet, E.R. Coping with the forced swim stressor: Current state-of-the-art. Behav. Brain Res. 2019, 364, 1–10. [Google Scholar] [CrossRef] [PubMed]
  118. Commons, K.G.; Cholanians, A.B.; Babb, J.A.; Ehlinger, D.G. The Rodent Forced Swim Test Measures Stress-Coping Strategy, Not Depression-like Behavior. ACS Chem. Neurosci. 2017, 8, 955–960. [Google Scholar] [CrossRef] [PubMed]
  119. Molendijk, M.L.; de Kloet, E.R. Immobility in the forced swim test is adaptive and does not reflect depression. Psychoneuroendocrinology 2015, 62, 389–391. [Google Scholar] [CrossRef] [PubMed]
  120. Parekh, P.K.; Johnson, S.B.; Liston, C. Synaptic Mechanisms Regulating Mood State Transitions in Depression. Annu. Rev. Neurosci. 2022, 45, 581–601. [Google Scholar] [CrossRef]
  121. Aleksandrova, L.R.; Wang, Y.T.; Phillips, A.G. Evaluation of the Wistar-Kyoto rat model of depression and the role of synaptic plasticity in depression and antidepressant response. Neurosci. Biobehav. Rev. 2019, 105, 1–23, Erratum in Neurosci Biobehav Rev. 2020, 116, 162–163. [Google Scholar] [CrossRef] [PubMed]
  122. Petković, A.; Chaudhury, D. Encore: Behavioural animal models of stress, depression and mood disorders. Front. Behav. Neurosci. 2022, 16, 931964. [Google Scholar] [CrossRef] [PubMed]
  123. Della Valle, A.; De Carlo, S.; Sonsini, G.; Pilati, S.; Perali, A.; Ubaldi, M.; Ciccocioppo, R. Machine learning-based model for behavioural analysis in rodents applied to the forced swim test. Sci. Rep. 2025, 15, 22314. [Google Scholar] [CrossRef]
  124. Cuthbert, B.N. Research Domain Criteria (RDoC): Progress and Potential. Curr. Dir. Psychol. Sci. 2022, 31, 107–114. [Google Scholar] [CrossRef]
  125. Anderzhanova, E.; Kirmeier, T.; Wotjak, C.T. Animal models in psychiatric research: The RDoC system as a new framework for endophenotype-oriented translational neuroscience. Neurobiol. Stress. 2017, 7, 47–56. [Google Scholar] [CrossRef] [PubMed]
  126. Tanaka, M. From Serendipity to Precision: Integrating AI, Multi-Omics, and Human-Specific Models for Personalized Neuropsychiatric Care. Biomedicines 2025, 13, 167. [Google Scholar] [CrossRef]
  127. Nusslock, R.; Alloy, L.B. Reward processing and mood-related symptoms: An RDoC and translational neuroscience perspective. J. Affect. Disord. 2017, 216, 3–16. [Google Scholar] [CrossRef]
  128. Cavanagh, J.F.; Gregg, D.; Light, G.A.; Olguin, S.L.; Sharp, R.F.; Bismark, A.W.; Bhakta, S.G.; Swerdlow, N.R.; Brigman, J.L.; Young, J.W. Electrophysiological biomarkers of behavioral dimensions from cross-species paradigms. Transl. Psychiatry 2021, 11, 482, Erratum in Transl Psychiatry. 2022, 12, 290. [Google Scholar] [CrossRef]
  129. Fernandes, B.S.; Karmakar, C.; Tamouza, R.; Tran, T.; Yearwood, J.; Hamdani, N.; Laouamri, H.; Richard, J.R.; Yolken, R.; Berk, M.; et al. Precision psychiatry with immunological and cognitive biomarkers: A multi-domain prediction for the diagnosis of bipolar disorder or schizophrenia using machine learning. Transl. Psychiatry 2020, 10, 162. [Google Scholar] [CrossRef] [PubMed]
  130. Sanislow, C.A.; Ferrante, M.; Pacheco, J.; Rudorfer, M.V.; Morris, S.E. Advancing Translational Research Using NIMH Research Domain Criteria and Computational Methods. Neuron 2019, 101, 779–782. [Google Scholar] [CrossRef]
  131. Morris, S.E.; Sanislow, C.A.; Pacheco, J.; Vaidyanathan, U.; Gordon, J.A.; Cuthbert, B.N. Revisiting the seven pillars of RDoC. BMC Med. 2022, 20, 220. [Google Scholar] [CrossRef] [PubMed]
  132. Arns, M.; van Dijk, H.; Luykx, J.J.; van Wingen, G.; Olbrich, S. Stratified psychiatry: Tomorrow’s precision psychiatry? Eur. Neuropsychopharmacol. 2022, 55, 14–19. [Google Scholar] [CrossRef] [PubMed]
  133. Milaneschi, Y.; Kappelmann, N.; Ye, Z.; Lamers, F.; Moser, S.; Jones, P.B.; Burgess, S.; Penninx, B.; Khandaker, G.M. Association of inflammation with depression and anxiety: Evidence for symptom-specificity and potential causality from UK Biobank and NESDA cohorts. Mol. Psychiatry 2021, 26, 7393–7402, Erratum in Mol Psychiatry. 2022, 27, 1856. [Google Scholar] [CrossRef]
  134. Tanaka, M. Special Issue “Translating Molecular Psychiatry: From Biomarkers to Personalized Therapies”. Int. J. Mol. Sci. 2025, 26, 10238. [Google Scholar] [CrossRef]
  135. Bekhbat, M.; Treadway, M.T.; Goldsmith, D.R.; Woolwine, B.J.; Haroon, E.; Miller, A.H.; Felger, J.C. Gene signatures in peripheral blood immune cells related to insulin resistance and low tyrosine metabolism define a sub-type of depression with high CRP and anhedonia. Brain Behav. Immun. 2020, 88, 161–165. [Google Scholar] [CrossRef]
  136. Baune, B.T.; Fromme, S.E.; Aberg, M.; Adli, M.; Afantitis, A.; Akkouh, I.; Andreassen, O.A.; Angulo, C.; Barlati, S.; Brasso, C.; et al. A stratified treatment algorithm in psychiatry: A program on stratified pharmacogenomics in severe mental illness (Psych-STRATA): Concept, objectives and methodologies of a multidisciplinary project funded by Horizon Europe. Eur. Arch. Psychiatry Clin. Neurosci. 2025, 275, 1453–1464. [Google Scholar] [CrossRef]
  137. Abi-Dargham, A.; Horga, G. The search for imaging biomarkers in psychiatric disorders. Nat. Med. 2016, 22, 1248–1255. [Google Scholar] [CrossRef] [PubMed]
  138. Guo, Q.; Guo, L.; Wang, Y.; Shang, S. Efficacy and safety of eight enhanced therapies for treatment-resistant depression: A systematic review and network meta-analysis of RCTs. Psychiatry Res. 2024, 339, 116018. [Google Scholar] [CrossRef]
  139. Łysik, A.; Logoń, K.; Szczygieł, A.; Wołoszczak, J.; Wrześniewska, M.; Leszek, J. Innovative approaches in the treatment-resistant depression: Exploring different therapeutic pathways. GeroScience 2025, 47, 5543–5558. [Google Scholar] [CrossRef]
  140. Salahudeen, M.S.; Wright, C.M.; Peterson, G.M. Esketamine: New hope for the treatment of treatment-resistant depression? A narrative review. Ther. Adv. Drug Saf. 2020, 11, 2042098620937899. [Google Scholar] [CrossRef]
  141. Brietzke, E.; Hawken, E.R.; Idzikowski, M.; Pong, J.; Kennedy, S.H.; Soares, C.N. Integrating digital phenotyping in clinical characterization of individuals with mood disorders. Neurosci. Biobehav. Rev. 2019, 104, 223–230. [Google Scholar] [CrossRef] [PubMed]
  142. Baldwin, H.; Loebel-Davidsohn, L.; Oliver, D.; Salazar de Pablo, G.; Stahl, D.; Riper, H.; Fusar-Poli, P. Real-World Implementation of Precision Psychiatry: A Systematic Review of Barriers and Facilitators. Brain Sci. 2022, 12, 934. [Google Scholar] [CrossRef] [PubMed]
  143. Dhieb, D.; Bastaki, K. Pharmaco-Multiomics: A New Frontier in Precision Psychiatry. Int. J. Mol. Sci. 2025, 26, 1082. [Google Scholar] [CrossRef]
  144. Zanardi, R.; Prestifilippo, D.; Fabbri, C.; Colombo, C.; Maron, E.; Serretti, A. Precision psychiatry in clinical practice. Int. J. Psychiatry Clin. Pract. 2021, 25, 19–27. [Google Scholar] [CrossRef] [PubMed]
  145. Fusar-Poli, P.; Manchia, M.; Koutsouleris, N.; Leslie, D.; Woopen, C.; Calkins, M.E.; Dunn, M.; Tourneau, C.L.; Mannikko, M.; Mollema, T.; et al. Ethical considerations for precision psychiatry: A roadmap for research and clinical practice. Eur. Neuropsychopharmacol. 2022, 63, 17–34. [Google Scholar] [CrossRef]
  146. Brown, J.E.H.; Young, J.L.; Martinez-Martin, N. Psychiatric genomics, mental health equity, and intersectionality: A framework for research and practice. Front. Psychiatry 2022, 13, 1061705. [Google Scholar] [CrossRef]
  147. Giusti-Rodríguez, P.; Okewole, N.; Jain, S.; Montalvo-Ortiz, J.L.; Peterson, R.E. Diversifying Psychiatric Genomics: Globally Inclusive Strategies Toward Health Equity. Psychiatr. Clin. N. Am. 2025, 48, 241–256. [Google Scholar] [CrossRef]
  148. Blackburn, T.P. Depressive disorders: Treatment failures and poor prognosis over the last 50 years. Pharmacol. Res. Perspect. 2019, 7, e00472. [Google Scholar] [CrossRef]
  149. Oasi, O.; Critchfield, K.L.; Werbart, A. Editorial: Rethinking unsuccessful psychotherapies: When and how do treatments fail? Front. Psychol. 2024, 15, 1514654. [Google Scholar] [CrossRef]
  150. Petter, J.; Schumacher, L.; Echterhoff, J.; Klein, J.P.; Schramm, E.; Härter, M.; Hautzinger, M.; Kriston, L. Heterogeneity of Treatment Outcomes Across Therapists and Sites in a Randomized Multicentre Psychotherapy Trial. Clin. Psychol. Psychother. 2025, 32, e70087. [Google Scholar] [CrossRef] [PubMed]
  151. Huneke, N.T.M.; Fusetto Veronesi, G.; Garner, M.; Baldwin, D.S.; Cortese, S. Expectancy Effects, Failure of Blinding Integrity, and Placebo Response in Trials of Treatments for Psychiatric Disorders: A Narrative Review. JAMA Psychiatry 2025, 82, 531–538. [Google Scholar] [CrossRef]
  152. Bjurner, P.; Isacsson, N.H.; Abdesslem, F.B.; Boman, M.; Forsell, E.; Kaldo, V. Study protocol for a triple-blind randomised controlled trial evaluating a machine learning-based predictive clinical decision support tool for internet-delivered cognitive behaviour therapy (ICBT) for depression and anxiety. Internet Interv. 2025, 40, 100816. [Google Scholar] [CrossRef]
  153. Lutz, W.; Deisenhofer, A.K.; Rubel, J.; Bennemann, B.; Giesemann, J.; Poster, K.; Schwartz, B. Prospective evaluation of a clinical decision support system in psychological therapy. J. Consult. Clin. Psychol. 2022, 90, 90–106. [Google Scholar] [CrossRef]
  154. Kwan, J.L.; Lo, L.; Ferguson, J.; Goldberg, H.; Diaz-Martinez, J.P.; Tomlinson, G.; Grimshaw, J.M.; Shojania, K.G. Computerised clinical decision support systems and absolute improvements in care: Meta-analysis of controlled clinical trials. BMJ 2020, 370, m3216. [Google Scholar] [CrossRef]
  155. Rybak, Y.E.; Lai, K.S.P.; Ramasubbu, R.; Vila-Rodriguez, F.; Blumberger, D.M.; Chan, P.; Delva, N.; Giacobbe, P.; Gosselin, C.; Kennedy, S.H.; et al. Treatment-resistant major depressive disorder: Canadian expert consensus on definition and assessment. Depress. Anxiety 2021, 38, 456–467. [Google Scholar] [CrossRef] [PubMed]
  156. Mora, F.; Ramos-Quiroga, J.A.; Baca-García, E.; Crespo, J.M.; Gutiérrez-Rojas, L.; Madrazo, A.; Pérez Costillas, L.; Saiz, P.A.; Tordera, V.; Vieta, E. Treatment-resistant depression and intranasal esketamine: Spanish consensus on theoretical aspects. Front. Psychiatry 2025, 16, 1623659. [Google Scholar] [CrossRef] [PubMed]
  157. Ivarsson, M.; Danielsson, H.; Imms, C. Measurement issues in longitudinal studies of mental health problems in children with neurodevelopmental disorders. BMC Psychol. 2025, 13, 267. [Google Scholar] [CrossRef] [PubMed]
  158. Lee, W.; Bindman, J.; Ford, T.; Glozier, N.; Moran, P.; Stewart, R.; Hotopf, M. Bias in psychiatric case-control studies: Literature survey. Br. J. Psychiatry 2007, 190, 204–209. [Google Scholar] [CrossRef]
  159. Abbaz Yazdian, F.; Khodabakhshi-Koolaee, A. Exploring the Counselors and Psychotherapists Perceptions of Therapeutic Errors in the Treatment Room. SAGE Open 2024, 14, 21582440241257320. [Google Scholar] [CrossRef]
  160. Hayes, S.C.; Hofmann, S.G. “Third-wave” cognitive and behavioral therapies and the emergence of a process-based approach to intervention in psychiatry. World Psychiatry 2021, 20, 363–375. [Google Scholar] [CrossRef]
  161. Tanaka, M. Parkinson’s Disease: Bridging Gaps, Building Biomarkers, and Reimagining Clinical Translation. Cells 2025, 14, 1161. [Google Scholar] [CrossRef]
  162. Meehan, A.J.; Lewis, S.J.; Fazel, S.; Fusar-Poli, P.; Steyerberg, E.W.; Stahl, D.; Danese, A. Clinical prediction models in psychiatry: A systematic review of two decades of progress and challenges. Mol. Psychiatry 2022, 27, 2700–2708. [Google Scholar] [CrossRef]
  163. Figueiredo Godoy, A.C.; Frota, F.F.; Araújo, L.P.; Valenti, V.E.; Pereira, E.; Detregiachi, C.R.P.; Galhardi, C.M.; Caracio, F.C.; Haber, R.S.A.; Fornari Laurindo, L.; et al. Neuroinflammation and Natural Antidepressants: Balancing Fire with Flora. Biomedicines 2025, 13, 1129. [Google Scholar] [CrossRef]
  164. Haslbeck, J.; Ryan, O.; Robinaugh, D.J.; Waldorp, L.J.; Borsboom, D. Modeling psychopathology: From data models to formal theories. Psychol. Methods 2022, 27, 930. [Google Scholar] [CrossRef]
  165. Halassa, M.M.; Frank, M.J.; Garety, P.; Ongur, D.; Airan, R.D.; Sanacora, G.; Dzirasa, K.; Suresh, S.; Fitzpatrick, S.M.; Rothman, D.L. Developing algorithmic psychiatry via multi-level spanning computational models. Cell Rep. Med. 2025, 6, 102094. [Google Scholar] [CrossRef] [PubMed]
  166. Tan, T.; Wang, W.; Liu, T.; Zhong, P.; Conrow-Graham, M.; Tian, X.; Yan, Z. Neural circuits and activity dynamics underlying sex-specific effects of chronic social isolation stress. Cell Rep. 2021, 34, 108874. [Google Scholar] [CrossRef] [PubMed]
  167. Bowman, R.; Frankfurt, M.; Luine, V. Sex differences in anxiety and depression: Insights from adult rodent models of chronic stress and neural plasticity. Front. Behav. Neurosci. 2025, 19, 1591973. [Google Scholar] [CrossRef]
  168. Mingardi, J.; Giovenzana, M.; Nicosia, N.; Misztak, P.; Ieraci, A.; Musazzi, L. Sex and Circadian Rhythm Dependent Behavioral Effects of Chronic Stress in Mice and Modulation of Clock Genes in the Prefrontal Cortex. Int. J. Mol. Sci. 2025, 26, 6410. [Google Scholar] [CrossRef] [PubMed]
  169. Bekhbat, M.; Ulukaya, G.B.; Bhasin, M.K.; Felger, J.C.; Miller, A.H. Cellular and immunometabolic mechanisms of inflammation in depression: Preliminary findings from single cell RNA sequencing and a tribute to Bruce McEwen. Neurobiol. Stress. 2022, 19, 100462. [Google Scholar] [CrossRef]
  170. Koppe, G.; Meyer-Lindenberg, A.; Durstewitz, D. Deep learning for small and big data in psychiatry. Neuropsychopharmacology 2021, 46, 176–190. [Google Scholar] [CrossRef]
  171. Driessen, E.; Efthimiou, O.; Wienicke, F.J.; Breunese, J.; Cuijpers, P.; Debray, T.P.A.; Fisher, D.J.; Fokkema, M.; Furukawa, T.A.; Hollon, S.D.; et al. Developing a multivariable prediction model to support personalized selection among five major empirically-supported treatments for adult depression. Study protocol of a systematic review and individual participant data network meta-analysis. PLoS ONE 2025, 20, e0322124. [Google Scholar] [CrossRef]
  172. Luedtke, A.; Sadikova, E.; Kessler, R.C. Sample size requirements for multivariate models to predict between-patient differences in best treatments of major depressive disorder. Clin. Psychol. Sci. 2019, 7, 445–461. [Google Scholar] [CrossRef]
  173. Watkins, E.R.; Newbold, A. Factorial Designs Help to Understand How Psychological Therapy Works. Front. Psychiatry 2020, 11, 429. [Google Scholar] [CrossRef]
  174. Ciolino, J.D.; Scholtens, D.M.; Bonner, L.B. Factorial Clinical Trial Designs. JAMA 2025, 333, 532–533. [Google Scholar] [CrossRef]
  175. Watkins, E.; Newbold, A.; Tester-Jones, M.; Javaid, M.; Cadman, J.; Collins, L.M.; Graham, J.; Mostazir, M. Implementing multifactorial psychotherapy research in online virtual environments (IMPROVE-2): Study protocol for a phase III trial of the MOST randomized component selection method for internet cognitive-behavioural therapy for depression. BMC Psychiatry 2016, 16, 345. [Google Scholar] [CrossRef]
  176. Kotecha, G.; Ventz, S.; Fortini, S.; Trippa, L. Uncertainty directed factorial clinical trials. Biostatistics 2024, 25, 833–851. [Google Scholar] [CrossRef] [PubMed]
  177. Victor, T.A.; Khalsa, S.S.; Simmons, W.K.; Feinstein, J.S.; Savitz, J.; Aupperle, R.L.; Yeh, H.W.; Bodurka, J.; Paulus, M.P. Tulsa 1000: A naturalistic study protocol for multilevel assessment and outcome prediction in a large psychiatric sample. BMJ Open 2018, 8, e016620. [Google Scholar] [CrossRef] [PubMed]
  178. Nelson, B.; McGorry, P.D.; Wichers, M.; Wigman, J.T.W.; Hartmann, J.A. Moving from Static to Dynamic Models of the Onset of Mental Disorder: A Review. JAMA Psychiatry 2017, 74, 528–534. [Google Scholar] [CrossRef]
  179. Harkness, A.R.; Reynolds, S.M.; Lilienfeld, S.O. A review of systems for psychology and psychiatry: Adaptive systems, personality psychopathology five (PSY-5), and the DSM-5. J. Pers. Assess. 2014, 96, 121–139. [Google Scholar] [CrossRef]
Figure 1. Translational timeline linking preclinical models, diagnostic frameworks, and first-line treatments in depression. The schematic illustrates the historical progression from early stress- and monoamine-based preclinical models, through key diagnostic milestones, to successive generations of pharmacological, psychotherapeutic, neuromodulatory, and digital interventions. The timeline highlights periods in which advances in experimental models, clinical nosology, and frontline care evolved in parallel, as well as phases in which these domains diverged, helping to contextualize current efforts to reconnect mechanism, diagnosis, and treatment within a systems psychiatry framework. Created in BioRender. Tanaka, M. (2025) https://BioRender.com/3omdcgb (accessed on 1 December 2025). BA, behavioral activation; CBT, cognitive behavioral therapy; CMS, chronic mild stress; CSDS, chronic social defeat stress; DREADD, designer receptors exclusively activated by designer drugs; DSM, Diagnostic and Statistical Manual of Mental Disorders; GCaMP, green fluorescent protein–calmodulin–M13 peptide; HAM-D, Hamilton Depression Rating Scale; ICD, International Classification of Diseases; IPT, interpersonal psychotherapy; LPS, lipopolysaccharide; MADRS, Montgomery–Åsberg Depression Rating Scale; MAOI, monoamine oxidase inhibitor; MBCT, mindfulness-based cognitive therapy; NMDA, N-methyl-D-aspartate; RDoC, Research Domain Criteria; rTMS, repetitive transcranial magnetic stimulation; SNRI, serotonin–norepinephrine reuptake inhibitor; SSRI, selective serotonin reuptake inhibitor; TCA, tricyclic antidepressant.
Figure 1. Translational timeline linking preclinical models, diagnostic frameworks, and first-line treatments in depression. The schematic illustrates the historical progression from early stress- and monoamine-based preclinical models, through key diagnostic milestones, to successive generations of pharmacological, psychotherapeutic, neuromodulatory, and digital interventions. The timeline highlights periods in which advances in experimental models, clinical nosology, and frontline care evolved in parallel, as well as phases in which these domains diverged, helping to contextualize current efforts to reconnect mechanism, diagnosis, and treatment within a systems psychiatry framework. Created in BioRender. Tanaka, M. (2025) https://BioRender.com/3omdcgb (accessed on 1 December 2025). BA, behavioral activation; CBT, cognitive behavioral therapy; CMS, chronic mild stress; CSDS, chronic social defeat stress; DREADD, designer receptors exclusively activated by designer drugs; DSM, Diagnostic and Statistical Manual of Mental Disorders; GCaMP, green fluorescent protein–calmodulin–M13 peptide; HAM-D, Hamilton Depression Rating Scale; ICD, International Classification of Diseases; IPT, interpersonal psychotherapy; LPS, lipopolysaccharide; MADRS, Montgomery–Åsberg Depression Rating Scale; MAOI, monoamine oxidase inhibitor; MBCT, mindfulness-based cognitive therapy; NMDA, N-methyl-D-aspartate; RDoC, Research Domain Criteria; rTMS, repetitive transcranial magnetic stimulation; SNRI, serotonin–norepinephrine reuptake inhibitor; SSRI, selective serotonin reuptake inhibitor; TCA, tricyclic antidepressant.
Biomedicines 14 00035 g001
Figure 2. Evolution of depression trial design across eligibility, endpoints, timing, stratification and durability. The schematic contrasts conventional antidepressant trials (Then, left) with emerging systems-psychiatry-informed designs (Now, right). Eligibility. Earlier trials typically enrolled narrowly defined adults with “pure” major depressive disorder (MDD), excluding common comorbidities, older age, suicidality, and medical complexity; contemporary designs move toward staged, better-characterized, and more inclusive samples, with comorbidities, treatment resistance, and functional impairment captured as structured baseline variables rather than automatic exclusions. Endpoints. Traditional studies relied on a single clinician-rated symptom scale (for example, HAM-D or MADRS) at one acute time point; modern trials incorporate multi-domain outcome batteries, including anhedonia and cognition, functioning and quality of life, patient-reported outcomes, and mechanistic readouts (for example, inflammatory markers, circuit or plasticity measures, digital behavior). Timing. Classic trials emphasized 6–8-week acute response with relatively sparse visits; new designs layer dense early sampling windows (hours to days for rapid-acting agents) onto longer follow-up to 6–12 months, enabling characterization of onset, offset, and relapse dynamics. Stratification. Earlier studies used minimal or post hoc subgrouping (for example, melancholic versus atypical) and randomization only at the syndrome level; contemporary designs prespecify strata and enrichment rules based on clinical staging, comorbidity clusters, biomarkers (such as inflammation or KYN metrics), and cognitive or circuit biotypes, and sometimes deploy adaptive or umbrella/basket architectures. Durability. Historically, continuation and maintenance phases were optional or handled in separate extension studies; current trials increasingly embed durability as a co-primary objective, tracking time to relapse, sustained response, taper/discontinuation effects, and real-world effectiveness via registries or digital follow-up, thereby aligning trial design with the long-term course of depressive illness. Created in BioRender. Tanaka, M. (2025) https://BioRender.com/evdupmh (accessed on 1 December 2025). HAM-D, Hamilton Depression Rating Scale; KYN, kynurenine; MADRS, Montgomery–Åsberg Depression Rating Scale; MDD, major depressive disorder.
Figure 2. Evolution of depression trial design across eligibility, endpoints, timing, stratification and durability. The schematic contrasts conventional antidepressant trials (Then, left) with emerging systems-psychiatry-informed designs (Now, right). Eligibility. Earlier trials typically enrolled narrowly defined adults with “pure” major depressive disorder (MDD), excluding common comorbidities, older age, suicidality, and medical complexity; contemporary designs move toward staged, better-characterized, and more inclusive samples, with comorbidities, treatment resistance, and functional impairment captured as structured baseline variables rather than automatic exclusions. Endpoints. Traditional studies relied on a single clinician-rated symptom scale (for example, HAM-D or MADRS) at one acute time point; modern trials incorporate multi-domain outcome batteries, including anhedonia and cognition, functioning and quality of life, patient-reported outcomes, and mechanistic readouts (for example, inflammatory markers, circuit or plasticity measures, digital behavior). Timing. Classic trials emphasized 6–8-week acute response with relatively sparse visits; new designs layer dense early sampling windows (hours to days for rapid-acting agents) onto longer follow-up to 6–12 months, enabling characterization of onset, offset, and relapse dynamics. Stratification. Earlier studies used minimal or post hoc subgrouping (for example, melancholic versus atypical) and randomization only at the syndrome level; contemporary designs prespecify strata and enrichment rules based on clinical staging, comorbidity clusters, biomarkers (such as inflammation or KYN metrics), and cognitive or circuit biotypes, and sometimes deploy adaptive or umbrella/basket architectures. Durability. Historically, continuation and maintenance phases were optional or handled in separate extension studies; current trials increasingly embed durability as a co-primary objective, tracking time to relapse, sustained response, taper/discontinuation effects, and real-world effectiveness via registries or digital follow-up, thereby aligning trial design with the long-term course of depressive illness. Created in BioRender. Tanaka, M. (2025) https://BioRender.com/evdupmh (accessed on 1 December 2025). HAM-D, Hamilton Depression Rating Scale; KYN, kynurenine; MADRS, Montgomery–Åsberg Depression Rating Scale; MDD, major depressive disorder.
Biomedicines 14 00035 g002
Table 1. Cross-species bridge map for depression-relevant constructs and translatable assays. Cross-species bridge map linking human constructs to preclinical assays, quantitative readouts, and clinical analogs that can be implemented in contemporary trials. “Status” summarizes translational maturity (routine, emerging, or exploratory), while “Design tip” highlights one concrete way to embed each bridge into mechanistically anchored, heterogeneity-aware study designs.
Table 1. Cross-species bridge map for depression-relevant constructs and translatable assays. Cross-species bridge map linking human constructs to preclinical assays, quantitative readouts, and clinical analogs that can be implemented in contemporary trials. “Status” summarizes translational maturity (routine, emerging, or exploratory), while “Design tip” highlights one concrete way to embed each bridge into mechanistically anchored, heterogeneity-aware study designs.
Human ConstructPreclinical AssayReadoutClinical AnalogStatusDesign Tip
Anhedonia/motivational deficitEffort-based decision tasks (progressive ratio, T-maze barrier, operant sucrose)Breakpoint, lever presses, willingness to work under stress or inflammationProbabilistic reward tasks, EEfRT, ventral striatal BOLD, anhedonia scalesEmerging trial biomarkerSeparate hedonic “liking” from motivational “wanting”; include stress/inflammation challenge blocks.
Negative affect/threat biasFear conditioning and extinction; chronic social defeatFreezing/avoidance, extinction curves, startle, social withdrawalFear-learning and extinction tasks, startle paradigms, threat-bias tasks in anxious/MDD subgroupsRobust basic science; limited clinical useUse as domain-specific endpoint in anxious and trauma-loaded depression; pair behavior with EEG/fMRI.
Cognitive control/executive dysfunctionAttentional set-shifting, 5-CSRTT, reversal learningErrors, omissions, reaction times, perseveration indexesSet-shifting (e.g., CANTAB), n-back, Stroop, Trail Making, DLPFC activationSecondary endpoint in several trialsPre-stratify “cognitively loaded” depression; link change to functioning and return-to-work outcomes.
Sleep and circadian disruptionRodent EEG/EMG with chronic stress or light-cycle shift; REM-deprivation modelsREM latency/density, NREM slow-wave power, activity rhythms, phase shiftsPolysomnography, actigraphy, DLMO, sleep/circadian questionnairesStrong observational; emerging endpointsAlign dosing and assessments with chronotype; treat sleep/circadian metrics as primary modifiable targets.
HPA axis and stress reactivityChronic mild stress, restraint, social defeat; Dex/CRH challengesCorticosterone profiles, GR sensitivity, coping style, stress-induced behavioral shiftCortisol awakening response, DST, lab stress tests, hair cortisolMixed but promising for subtypingSample across diurnal cycle; co-model stress markers with symptom domains (anergy, anxiety, cognitive fog).
Inflammation–KYN steeringLPS/IFN-α or stress-sensitized immune activation; Trp–KYN pathway assaysKYN/Trp ratio, QA/KYNA balance, microglial activation, cytokine panelsCRP, IL-6/TNF panels, plasma KYN/Trp, symptom clusters (anergia, anhedonia, psychomotor slowing)High translational interestPre-specify “inflammation-high” strata; collect longitudinal KYN panels and align with treatment response.
Metabolic–endocrine loadHigh-fat diet, genetic obesity, insulin-resistance modelsGlucose tolerance, insulin signaling, adiposity, spontaneous activityBMI, waist-to-hip ratio, HOMA-IR, HbA1c, metabolic-syndrome indicesGrowing but underused in trialsEmbed metabolic panels into TRD studies; design dedicated obesity/T2D depression trials with functional endpoints.
Synaptic plasticity/rapid-acting responseKetamine/psychedelic paradigms; LTP/LTD, in vivo spine imaging, AMPA-forward assaysSpine density, AMPA/NMDA ratio, LTP/LTD magnitude, early oscillatory changesEarly EEG/MEG plasticity markers, TMS-LTP readouts, 24–72 h symptom and cognition shiftsStrong mechanistic, clinical for ketamineBuild in early (24–72 h) windows and plasticity markers as key secondary endpoints in rapid-acting trials.
Network-level connectivity biotypesChemogenetic/optogenetic PFC–striatal/limbic manipulation; rodent rsfMRI/EEGResting-state connectivity, oscillatory coupling, causal node influence, behavior under circuit controlrsfMRI biotypes, TMS-EEG connectivity, SCC/vmPFC network markers for neuromodulation targetingEmerging targeting toolRequire “target engagement” thresholds for drugs/devices; enrich samples by baseline network topology.
Digital behavior and passive monitoringHome-cage automated monitoring of movement, sleep, and social interactionContinuous activity, sleep–wake structure, social proximity, exploration patternsSmartphone-based mobility, call/text patterns, speech and behavior passively captured by sensorsEarly exploratoryPredefine digital endpoints (e.g., mobility, social withdrawal) and link them to functional and relapse outcomes.
5-CSRTT, five-choice serial reaction time task; BMI, body mass index; AMPA, α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid; BOLD, blood oxygen-level-dependent; CANTAB, Cambridge Neuropsychological Test Automated Battery; Dex/CRH, dexamethasone/corticotropin-releasing hormone; DLPFC, dorsolateral prefrontal cortex; DLMO, dim light melatonin onset; DST, dexamethasone suppression test; EEG, electroencephalograph; EMG, electromyography; fMRI, functional magnetic resonance imaging; GR, HbA1c, glycated hemoglobin; glucocorticoid receptor; HOMA-IR, homeostatic model assessment of insulin resistance; HPA, hypothalamic–pituitary–adrenal; IFN-α interferon-alpha; IL-6, interleukin-6; KYN, kynurenine; LTD, long-term depression; LPS, lipopolysaccharide; LTP, long-term potentiation; MEG, magnetoencephalography; NMDA, N-methyl-D-aspartate; NREM, non-rapid eye movement; PFC, prefrontal cortex; QA, quinolinic acid; REM, rapid eye movement; rsfMRI, resting-state functional magnetic resonance imaging; SCC, subcallosal cingulate; T2D, type 2 diabetes; TMS, transcranial magnetic stimulation; TNF, tumor necrosis factor; TRD, treatment-resistant depression; Trp, tryptophan; vmPFC, ventromedial prefrontal cortex.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Tanaka, M. From Monoamines to Systems Psychiatry: Rewiring Depression Science and Care (1960s–2025). Biomedicines 2026, 14, 35. https://doi.org/10.3390/biomedicines14010035

AMA Style

Tanaka M. From Monoamines to Systems Psychiatry: Rewiring Depression Science and Care (1960s–2025). Biomedicines. 2026; 14(1):35. https://doi.org/10.3390/biomedicines14010035

Chicago/Turabian Style

Tanaka, Masaru. 2026. "From Monoamines to Systems Psychiatry: Rewiring Depression Science and Care (1960s–2025)" Biomedicines 14, no. 1: 35. https://doi.org/10.3390/biomedicines14010035

APA Style

Tanaka, M. (2026). From Monoamines to Systems Psychiatry: Rewiring Depression Science and Care (1960s–2025). Biomedicines, 14(1), 35. https://doi.org/10.3390/biomedicines14010035

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Article metric data becomes available approximately 24 hours after publication online.
Back to TopTop