Abstract
Spontaneous intracerebral hemorrhage (ICH) is a devastating form of stroke, disproportionately affecting older adults and is associated with high rates of mortality, functional dependence, and long-term cognitive decline. Aging profoundly alters the structure and function of the cerebral vasculature, predisposing the brain to both covert hemorrhage and the development of cerebral microbleeds (CMBs), small, often subclinical lesions that share common pathophysiological mechanisms with ICH. These mechanisms include endothelial dysfunction, impaired cerebral autoregulation, blood–brain barrier breakdown, vascular senescence, and chronic inflammation. Systemic factors such as age-related insulin-like growth factor 1 (IGF-1) deficiency further exacerbate microvascular vulnerability. CMBs and ICH represent distinct yet interconnected manifestations along a continuum of hemorrhagic small vessel disease, with growing recognition of their contribution to vascular cognitive impairment and dementia (VCID). Despite their increasing burden, older adults remain underrepresented in clinical trials, and few therapeutic approaches specifically target aging-related mechanisms. This review synthesizes current knowledge on the cellular, molecular, and systemic drivers of ICH and CMBs in aging, highlights diagnostic and therapeutic challenges, and outlines opportunities for age-sensitive prevention and individualized care strategies.
1. Introduction
Spontaneous intracerebral hemorrhage (ICH) is a devastating form of stroke, resulting from bleeding into the brain parenchyma [,]. Although it accounts for only 10–15% of all strokes, ICH carries the highest mortality and morbidity among all stroke subtypes [,]. Survivors often face profound and persistent functional dependence, long-term cognitive impairment, and diminished quality of life. As global populations continue to age, the burden of ICH is projected to rise sharply. Individuals over the age of 75 now represent a substantial and growing proportion of ICH cases, making it imperative to understand how aging itself [], beyond traditional vascular risk factors such as hypertension or anticoagulant use, predisposes the brain to hemorrhagic injury [].
ICH is increasingly recognized not as a random catastrophic event, but as the endpoint of chronic vascular pathology, particularly cerebral small vessel disease (cSVD) [,,,], which is highly prevalent in older adults [,]. Mechanisms such as vascular endothelial dysfunction, impaired autoregulation, blood–brain barrier (BBB) disruption, and cellular senescence converge to compromise the structural integrity of the cerebral microvasculature. These changes lead to a state of progressive microvascular fragility, which not only predisposes to large parenchymal bleeds but also gives rise to cerebral microbleeds (CMBs), tiny foci of blood leakage that accumulate silently over time [,,].
CMBs, detectable by susceptibility-weighted imaging (SWI) on magnetic resonance imaging (MRI) [], are increasingly viewed as an integral part of the hemorrhagic spectrum of cSVD [,,,,]. Although often labeled “silent,” CMBs are far from clinically irrelevant. Their presence correlates with increased risk of future symptomatic ICH and is strongly associated with vascular cognitive impairment and dementia (VCID) [,,,,,]. In many cases, CMBs coexist with or precede overt hemorrhagic stroke, and they share common risk factors and underlying mechanisms, including chronic hypertension, cerebral amyloid angiopathy (CAA) [,], and age-related endothelial dysfunction. As such, CMBs and ICH should not be regarded as distinct entities, but rather as different manifestations along a continuum of hemorrhagic vascular brain injury [].
This convergence has critical implications for public health. While neurodegenerative diseases like Alzheimer’s disease (AD) remain difficult to prevent or modify at the population level, vascular contributors to cognitive decline, including CMBs and ICH, are, at least in theory, modifiable and even preventable []. Recognizing and targeting the early microvascular changes that preceded over hemorrhage offers a compelling opportunity for intervention, particularly in aging populations at highest risk [].
This review synthesizes current knowledge on the mechanisms by which aging promotes cerebral hemorrhage, with a special focus on cellular and molecular drivers of microvascular weakening. We explore how cSVD underlies both CMBs and larger ICH, review age-specific clinical presentations and outcomes, and examine evolving diagnostic and therapeutic strategies. By bridging basic vascular biology with clinical gerontology and stroke medicine, we aim to highlight the critical need for age-sensitive, mechanism-informed approaches to reduce the burden of hemorrhagic stroke and related cognitive decline in older adults.
2. Pathophysiological Mechanisms Linking Aging to CMBs and ICH
The vulnerability of the aging brain to both CMBs and spontaneous primary ICH [] arise from a multifactorial process involving both local and systemic drivers of cerebrovascular aging [,,,,,,,,,,,,]. These processes, many of which reflect hallmarks of aging, compromise the structural and functional integrity of cerebral vessels and contribute to the continuum of hemorrhagic cSVD []. The key pathophysiological mechanisms underlying these processes are illustrated in Figure 1.
Figure 1.
Pathophysiological mechanisms of intracerebral hemorrhage in aging brain (BBB: blood–brain barrier; ECM: extracellular matrix; IGF-1: insulin-like growth factor 1).
2.1. Endothelial Dysfunction and Blood–Brain Barrier (BBB) Breakdown
With advancing age, endothelial cells in the cerebral microvasculature exhibit reduced nitric oxide (NO) bioavailability [,,,,,,], impaired angiogenic signaling [,], and increased oxidative stress [,,,,]. These changes impair endothelium-dependent vasodilation and compromise the integrity of the BBB [,,,,,,,,,], promoting leakage of plasma proteins and erythrocytes into parenchyma and inducing an inflammatory microenvironment [,]. BBB disruption is a key antecedent of both CMBs and ICH, particularly in the setting of fluctuating blood pressure [].
2.2. Vascular Senescence and Extracellular Matrix (ECM) Remodeling
Cellular senescence [,], an important hallmark of aging [,,,,,,,,], affects various components of the neurovascular unit [,,,,,,], such as endothelial cells [,], pericytes, microglia [] and astrocytes [,], and leads to the release of senescence-associated secretory phenotype (SASP) factors [,], including pro-inflammatory cytokines and matrix metalloproteinases (MMPs) []. These mediators degrade the extracellular matrix (ECM), weaken the basement membrane, and promote loss of vascular elasticity and cohesion [,]. The net effect is increased susceptibility to microvascular rupture. Increased presence of senescent cells has also been linked to endothelial dysfunction [], BBB disruption [,,,,,] and genesis of CMBs [].
2.3. CSVD: A Common Pathological Substrate
CMBs and ICH frequently arise from two major forms of cSVD [,,]. Hypertensive arteriopathy, characterized by arteriolosclerosis, lipohyalinosis, fibrinoid necrosis, and microaneurysm formation, predominantly and typically affects deep perforating arteries and is a major driver of deep ICH and deep CMBs, such as those located in the thalamus, basal ganglia, and brainstem (particularly the pons); however, lobar hemorrhages (including ICH and CMBs) can also develop secondary to hypertensive arteriopathy [,,,,,,]. On the other hand, cerebral amyloid angiopathy (CAA) [,,,] involves the deposition of β-amyloid predominantly in cortical and leptomeningeal vessels, leading to vessel wall weakening and consequent hemorrhages in the parenchyma (i.e., lobar CMBs and lobar ICH) as well as the leptomeninges (i.e., convexity subarachnoid hemorrhage (cSAH) and its chronic manifestation, cortical superficial siderosis (cSS) []. The presence of the above hemorrhagic alterations in a strictly lobar/superficial pattern (i.e., without deep hemorrhages) allows the clinical-radiological diagnosis of probable CAA as per the consecutive versions of the Boston criteria (most recently the v2.0), with the mixed (i.e., lobar and deep) hemorrhagic pattern being considered to be more likely due to hypertensive arteriopathy only, and the cerebellar hemorrhagic alterations being not counted []. CAA is strongly age-dependent and is closely linked to cognitive decline. In both pathologies (i.e., hypertensive arteriopathy and sporadic CAA, microvascular rupture is the culmination of chronic degenerative processes, compounded by systemic factors such as hypertension and impaired cerebral autoregulation. Indeed, the etiology of hemorrhage formation in CAA is likely multifactorial, complex, and expands far beyond pure fragility due to β-amyloid deposition. This is supported by the observations that only a portion of patients with histopathologically defined moderate-to-severe CAA develop hemorrhagic alterations [], and that an increased risk for ICH development in adult patients with Down syndrome (with the trisomy of chromosome 21 resulting in an excess copy of the amyloid precursor protein (APP) and subsequent severe CAA) is not established [].
Reports on hyperacute ICH captured serendipitously during imaging support the hypothesis that hematoma expansion both in hypertensive and CAA related ICH results from multiple sources of bleeding due to a cascade of secondary vessel ruptures with eccentric expansion rather than a single source and continuous bleeding with concentric expansion reflecting the global fragility of the cerebral vasculature [,,].
2.4. Impaired Cerebral Autoregulation and Hemodynamic Stress
Cerebral autoregulation is the intrinsic ability of the brain to maintain stable cerebral blood flow (CBF) despite fluctuations in systemic blood pressure [,,,]. This protective mechanism operates across a wide range of mean arterial pressures (typically 60–150 mmHg) and is essential for shielding the fragile microcirculation from ischemic or hemorrhagic injury. Autoregulation is accomplished through tightly orchestrated myogenic, metabolic, and neurogenic mechanisms that dynamically adjust cerebrovascular tone. In aging, cerebral autoregulation becomes progressively compromised. This decline not only reduces the brain’s ability to maintain perfusion during hypotension but also increases its vulnerability to hypertensive injury, contributing directly to the pathogenesis of both CMBs and ICH [,]. Both older individuals and aged laboratory animals exhibit blunted myogenic responses of cerebral resistance vessels, limiting the ability of the vasculature to constrict or dilate in response to fluctuations in perfusion pressure [,]. This diminished responsiveness compromises the brain’s capacity to buffer hemodynamic stress, increasing vulnerability to both hypoperfusion and hyperperfusion, while age-related changes in hypertensive conditions further disrupt the physiological adaptive remodeling processes of cerebral blood vessels. Research indicates that older mammals exhibit compromised remodeling capacity, characterized by impaired prostanoid signaling pathways, dysfunctional transient receptor potential (TRP) channel activity, and defective calcium signaling mechanisms, all of which collectively impair vascular plasticity and adaptive responses.
2.4.1. Myogenic Dysfunction: Loss of Pressure-Induced Vasoconstriction
The myogenic response, mediated by vascular smooth muscle cells (VSMCs), is a central pillar of cerebral autoregulation []. In response to increased intraluminal pressure, VSMCs contract to maintain constant flow and prevent overperfusion []. Aging impairs this response through multiple mechanisms. First, it was shown earlier that aging impairs hypertension-induced increased pressure-dependent Ca2+ signaling in cerebrovascular smooth muscle cells in a 20-HETE/TRPC channel-dependent manner, leading to decreased pressure-induced constriction of the vessels [,]. Age-related decline in insulin-like growth factor 1 (IGF-1) signaling may play a pivotal role in this dysfunctional response to increased intraluminal pressure []. Second, it is believed that VSMCs senescence and phenotypic switching reduce contractile capacity. Third, ECM stiffening and collagen accumulation diminish vessel compliance. As a result, cerebral arterioles in the aged brain become less capable of constricting in response to systemic pressure increases []. This myogenic failure allows excessive pressure to be transmitted downstream to capillaries and venules, elevating the risk of mechanical rupture and hemorrhage [,].
2.4.2. Impaired Adaptation to Chronic Hypertension
In younger individuals, chronic hypertension induces adaptive remodeling of cerebral arterioles, including wall thickening, increased smooth muscle content, and upward resetting of the autoregulatory curve []. This is completed by the increased 20-HETE-dependent pressure-induced constriction of cerebral vessels mentioned above [,]. These changes expand the upper limit of autoregulatory protection, allowing the brain to tolerate higher systemic pressures without hyperperfusion injury [,,]. However, in aging, this adaptive capacity is blunted [,]. Vascular remodeling becomes maladaptive, characterized by stiffening, fibrosis, and inward hypertrophic remodeling without preserved function [,]. The failure to appropriately reset the autoregulatory range in response to sustained hypertension leaves aged microvessels dangerously exposed during hypertensive surges [,]. This impaired adaptation explains why older adults with moderate or even well-controlled hypertension can still suffer from hemorrhagic lesions. In line with the notion that deep perforating arterioles are the most affected by chronic hypertensive arteriopathy and consequent impaired adaptation, hypertensive surge at presentation (defined as ≥180 mmHg systolic blood pressure (SBP) was found to be an independent predictor of deep ICH location []. Superficial cortical/leptomeningeal vessels are more prone to be burdened by CAA (especially in older people, thus preferentially leading to lobar CMBs, cSAH/cSS, and lobar ICH. However, the prevalence of chronic hypertension was invariably high in patients with both CAA-related and non-CAA-related ICH, suggesting it to be a common underlying risk factor for ICH development in both CAA and hypertensive arteriopathy [].
2.4.3. Functional Consequences: CMBs and ICH as Spectrum Manifestations
When cerebral autoregulatory mechanisms fail, cerebral perfusion becomes passively dependent on systemic blood pressure. In this vulnerable state, hypertensive episodes, especially nocturnal or paroxysmal surges, can exert abrupt and excessive mechanical stress on the cerebral microvasculature [,,]. Fragile vessels, already compromised by cSVD, are unable to buffer these fluctuations, leading to microvascular injury []. This failure of autoregulatory protection is particularly consequential in deep brain regions affected by hypertensive arteriopathy, where sudden pressure increases can trigger focal arteriolar rupture and result in ICH. At the same time, repeated subclinical stress, especially in cortical regions burdened by CAA, promotes chronic leakage, hemosiderin deposition, and the progressive formation of CMBs [,]. Even moderate, transient fluctuations in blood pressure, in the absence of sustained hypertension, can produce cumulative vascular damage over time in older adults with impaired autoregulation.
The underlying pathophysiology involves a combination of myogenic dysfunction, which blunts pressure-induced vasoconstriction, and impaired vascular adaptation to chronic hypertension, which fails to shift the autoregulatory range upward []. Together, these mechanisms create a permissive environment for both chronic microvascular leakage and acute rupture. CMBs and ICH should thus be viewed as distinct yet interconnected manifestations along a shared continuum of hemorrhagic small vessel pathology, where the severity and distribution of damage are shaped by vessel size, the presence of arteriolosclerosis or CAA, and the intensity of hemodynamic stress [].
2.4.4. Interplay with Small Vessel Disease and Neurovascular Uncoupling
The structural changes of cSVD, such as arteriolar wall thickening, perivascular space dilation, and capillary rarefaction [,], further impair the autoregulatory reserve [,]. Concurrent neurovascular uncoupling [] reduces the precision of flow regulation in response to metabolic demand, exacerbating regional perfusion instability [].
Moreover, orthostatic hypotension, nocturnal hypertension, and increased blood pressure variability, all common in older adults, can provoke repeated autoregulatory failure, progressively worsening vascular damage [].
Importantly, autoregulatory failure is both a cause and a consequence of cSVD. The chronic ischemia, BBB leakage, and microvascular rarefaction characteristic of cSVD further erode vascular reactivity, creating a vicious cycle wherein structural damage and functional dysregulation exacerbate one another [,,]. This interplay increases the likelihood of both progressive CMBs accumulation and catastrophic vessel rupture.
2.5. Chronic Sterile Inflammation—Inflammaging
Aging is associated with a state of chronic, low-grade systemic inflammation, termed “inflammaging” [,,,,,], which impacts the cerebral circulation as well. In aging endothelial cells acquire a pro-inflammatory phenotype [,,]. The increased presence of senescent cells in the aging brain through the release of SASP contributes to a heightened state of inflammation [,,,]. Activated microglia and astrocytes release pro-inflammatory cytokines, further disrupting the BBB and promoting vascular injury. Elevated levels of circulating inflammatory mediators such as IL-6, TNF-α, and CRP have also been linked to increased burden of CMBs and higher risk of ICH in older adults.
The role of inflammation in hemorrhage formation is particularly relevant in CAA and has recently gathered increasing interest []. Indeed, CAA has been linked to chronic cellular and molecular inflammatory alterations in the brain both in experimental animals and in humans [,]. In addition, CAA can manifest in a typically subacute (but occasionally chronic) progressive encephalopathy termed CAA-related inflammation (CAA-RI), a condition histopathologically characterized by the accumulation of perivascular and/or transmural (i.e., vasculitic) inflammatory infiltrates in association with CAA-affected vessels, comprising lymphocytes, microglia/macrophages, and frequently multinucleated giant cells. The typical radiological presentation (in addition to CAA-compatible lobar hemorrhagic features) includes the appearance of asymmetric confluent white matter hyperintensities due to vasogenic edema. However, the most recent diagnostic criteria recognize the role of additional radiological features reflecting inflammation, including sulcal non-nulling on the fluid-attenuated inversion recovery (FLAIR) MRI and leptomeningeal contrast enhancement, representing a change in cerebrospinal fluid consistency and an increased leptomeningeal blood vessel permeability, respectively [,]. Importantly, remission of inflammation with clinical improvement can be achieved by immunosuppressive therapy (with corticosteroids in the pipeline) in ~80% percent of the cases; however, spontaneous improvement is also frequently reported (in some ~30–60% of the non-treated) []. Though early studies comparing CAA-RI and non-inflammatory CAA cohorts did not suggest an association between inflammation and ICH formation, a recent retrospective study on patients with sufficient follow-up reported strongly suggestive findings []. These include (1) a remarkable 33.7% incidence of lobar ICH at presentation or within 1 year in definite CAA-RI patients compared to the known ~8.5% annual lobar ICH risk in CAA in general [], (2) a frequent direct topographic association of hallmark white matter changes with CMBs, a feature also being associated with increased probability of true vasculitic pathology, (3) and the observation that clinical improvement (suggesting remission of inflammation) was associated with a significantly decreased probability of lobar ICH development within 1 year []. Recent reports on devastating fatal ICH following systemic thrombolysis with post-mortem findings of silent CAA-RI are in line with this concept [,]. Most recently, a series of CAA patients presenting with “amyloid spells”, pathognomonic cSAH/disseminated CSS, and additional leptomeningeal enhancement (i.e., meeting the novel criteria for probable CAA-RI) demonstrated parallel improvement of the spells and the leptomeningeal enhancement, suggesting a possible link between cSAH/cSS formation and chronic inflammation in CAA []. With the border between inflammatory and non-inflammatory CAA becoming gradually diminished, clinical trials aiming at halting inflammation with the purpose of preventing lobar ICH development in CAA are underway [].
With the advent of monoclonal anti-β-amyloid antibodies as proposed and recently approved therapeutic options in AD, a group of radiological alterations has emerged in the past decade as adverse events in clinical trials, collectively termed amyloid-related imaging abnormalities (ARIAs) []. These comprise (1) ARIA-H (ARIA-hemorrhage), including lobar/subarachnoid hemorrhagic alterations typical of CAA, and (2) ARIA-E (ARIA-edema and ARIA-effusion), including white matter vasogenic edema and sulcal hyperintensity (i.e., sulcal non-nulling) on the FLAIR, respectively. Due to the striking resemblance of these alterations and their clinical symptoms to that seen in CAA-RI (especially as per the most recent criteria proposals [], the identical (peri)vasculitic change of CAA vessels reported as histopathological correlate in the few autopsy reports available [,], the propensity to respond to steroid therapy, and the excess risk posed by carrying an ApoE ε4 allele, this adverse reaction has been referred to as iatrogenic CAA-RI or iatrogenic ARIA, as opposed to the spontaneous CAA-RI or spontaneous ARIA []. Iatrogenic ARIA has occasionally been associated with macroscopic lobar ICH in clinical trials, representing one of the most feared complications, sometimes in the context of systemic thrombolysis applied for presumed ischemic stroke [,]. Though the rigorously defined eligibility criteria (e.g., absence of prior spontaneous ARIA-H or ARIA-E and for lecanemab the absence of two ApoE ε4 alleles) will likely reduce the risk of such complications, heightened surveillance for ARIA and related clinical signs will be warranted in the upcoming monoclonal anti-β-amyloid antibody era to address this newly emerged etiology of lobar ICH. Figure 2 shows the hallmark magnetic resonance imaging features of CAA and CAA-RI.
Figure 2.
Hallmark magnetic resonance imaging features of CAA and CAA-RI. (A) Strictly lobar CMBs presenting as small globular hypointensities on SWI (a left parietal CSS can also be appreciated). (B) Lobar ICH (late subacute) in the right parietal lobe on SWI (several surrounding CSS, a small left parietal cSS, and two left frontal CMBs can also be observed). (C) Disseminated CSS in form of gyral hypointensities on the cortical surface on SWI in a patient presenting with TFNEs (a single CMB can also be appreciated right postcentrally). (D) Multiple foci of cSAH on the right prefrontal convexity presenting as a constellation of SWI hypointensity filling the affected sulci with corresponding SNN (i.e., sulcal hyperintensity) on the FLAIR (multiple chronic cSS can also be appreciated with no corresponding SNN). (E) White matter hyperintensities in a multispot pattern on FLAIR in the bilateral subcortical white matter, a feature introduced in the Boston criteria v2.0 []. (F) Severe MRI-visible (i.e., dilated) perivascular spaces in the centrum semiovale on T2, a feature introduced in the Boston criteria v2.0 []. (G) Asymmetric confluent white matter hyperintensity corresponding to vasogenic edema (also known as. ARIA-edema) in the left temporal, parietal, and occipital lobes on FLAIR in a patient with strictly lobar hemorrhagic alterations and subacute cognitive decline, meeting the classical criteria for probable CAA-RI (foci of thin SNN can also be appreciated; the edema completely resolved after corticosteroid therapy). (H) Right parietal SNN without corresponding SWI hypointensity (corresponding to ARIA-effusion) in a patient presenting with TFNEs and disseminated cSS, meeting the proposed extended criteria for probable CAA-RI []. (ARIA, amyloid-related imaging abnormality; CAA, cerebral amyloid angiopathy; CAA-RI, CAA-related inflammation; CMB, cerebral microbleed; cSAH, convexity subarachnoid hemorrhage; cSS, cortical superficial siderosis; FLAIR, fluid-attenuated inversion recovery; ICH, intracerebral hemorrhage; MRI, magnetic resonance imaging; SNN, sulcal non-nulling; SWI, susceptibility-weighted imaging; TFNE, transient focal neurological episode).
2.6. Systemic Endocrine Dysregulation: Role of IGF-1 Deficiency
The vulnerability of the aging brain to hemorrhagic injury is not solely the result of local vascular changes. Increasing evidence suggests that aging is orchestrated, at least in part, by systemic regulatory mechanisms [,,,,,,], circulating factors that influence the function and resilience of distant organs, including the brain []. This concept has been elegantly demonstrated in parabiosis studies, in which the circulatory systems of young and old animals are surgically joined []. Exposure of aged animals to young blood reverses multiple age-related impairments, including declines in cognitive function, neurogenesis, and vascular function, while young animals exposed to old blood exhibit premature vascular aging and reduced endothelial integrity [,,,,,].
These experiments have shifted the paradigm of aging research by revealing that circulating pro-youth or pro-aging factors can directly regulate cerebral vascular health []. Among these, IGF-1 has emerged as a central player in the systemic control of vascular aging [,,,,,,,].
IGF-1 is a pleiotropic growth factor predominantly produced in the liver under the control of growth hormone (GH), and its circulating levels decline markedly with age in both humans and experimental animals [,,,,,,,]. IGF-1 plays a crucial role in maintaining vascular homeostasis through multiple, interrelated mechanisms [,,,]. It supports endothelial function by enhancing the activity of endothelial nitric oxide synthase (eNOS), thereby promoting the production of NO, a key mediator of vasodilation and vascular tone [,,,,]. In parallel, IGF-1 exerts antioxidant effects by modulating the generation of reactive oxygen species (ROS), improving mitochondrial function, and limiting oxidative stress within the vascular wall [,,,,,]. It also plays a central role in preserving BBB integrity, helping to stabilize endothelial tight junctions and suppress inflammatory signaling pathways that compromise barrier permeability [,]. Finally, IGF-1 facilitates vascular repair by promoting endothelial cell survival, stimulating proliferation, and enhancing angiogenesis [,,,,,,]. Through these combined actions, IGF-1 maintains the structural and functional integrity of the cerebral microvasculature and protects against aging-related microvascular damage [].
With aging, the decline in systemic IGF-1 [] signaling leads to endothelial dysfunction, loss of BBB integrity [], and increased microvascular fragility []—key contributors to both CMBs and ICH [,]. Experimental studies in IGF-1-deficient or IGF-1-depleted animal models consistently show an increased burden of CMBs [,,], impaired autoregulatory function [,] and neurovascular uncoupling [], pathological structural adaptation to hypertension [,] and heightened vulnerability to hemorrhagic injury []. Conversely, restoration of IGF-1 levels has been shown to ameliorate vascular dysfunction in aged mice. Human studies further support these findings: lower serum IGF-1 levels in older adults have been associated with increased microvascular pathologies, neurovascular uncoupling [], poorer cognitive outcomes, and elevated risk of spontaneous ICH []. These associations persist even after adjusting for classical vascular risk factors, highlighting IGF-1 as a potential circulating biomarker and therapeutic target in hemorrhagic small vessel disease []. The connection between IGF-1 deficiency and CMBs/ICH formation underscores the importance of endocrine-vascular crosstalk in the pathogenesis of cerebrovascular aging [,,]. Unlike structural vascular changes, systemic hormonal imbalances may be more amenable to therapeutic modulation, offering a promising avenue for preventive interventions in at-risk older populations.
3. Clinical Features and Challenges in Older Adults
ICH in older adults presents distinct clinical challenges stemming from atypical symptomatology, age-related anatomical patterns, diagnostic challenges, and poorer prognoses []. Diagnostic pitfalls arise from atypical clinical presentation and overlapping baseline conditions, often delaying critical imaging and treatment []. Key predictors of outcome in this population include hematoma characteristics, initial neurological status, and age-specific factors such as frailty and pre-existing conditions.
3.1. Atypical Presentations in Older Adults
ICH in older adults frequently presents with more severe symptoms and poorer prognosis compared to younger patients []. Older adults with ICH manifest non-specific symptoms such as acute confusional states, delirium, or unexplained falls, which can also be attributed to baseline cognitive impairment, multiple chronic illnesses, systemic infections, metabolic disturbances, or adverse drug effects []. Additionally, age-associated physiological alterations, such as modified neuroinflammatory mechanisms and compromised cerebral autoregulation, can mask typical clinical signs of ICH and complicate timely recognition and diagnosis, often resulting in worse outcomes [].
3.2. Hemorrhage Location and Aging Patterns
The anatomical distribution of ICH in older adults reflects the influence of different underlying vascular pathologies []. As previously discussed, deep hemorrhages, involving the basal ganglia, thalamus, and brainstem, are strongly associated with hypertensive arteriopathy, as prolonged exposure to elevated blood pressure leads to degenerative changes in the small perforating arteries supplying these regions []. Deep hemorrhages often result in profound motor or sensory deficits due to the involvement of critical subcortical structures, and their clinical course may be further complicated by conditions such as hydrocephalus, particularly in the case of intraventricular hemorrhage [].
On the other hand, lobar hemorrhages affecting the cerebral cortex and immediately subcortical white matter are closely associated with CAA [], a degenerative vasculopathy characterized by the deposition of β-amyloid protein predominantly within the walls of cortical and leptomeningeal blood vessels, leading to vessel fragility and increased susceptibility to hemorrhage [,]. Importantly, CAA-related lobar hemorrhages tend to be recurrent and are often accompanied by cSS []. The pathogenesis of cSS include chronic hemosiderin (iron) deposition due to (likely repeated) episodes of cSAHs, arising from CAA-affected fragile superficial cortical/leptomeningeal small vessels []. The clinical relevance of cSAH/cSS is provided by their propensity to be the anatomical substrate of transient focal neurological episodes (TFNEs, also known as ‘amyloid spells’) and their association with a further increased risk of lobar ICH [,,]. Moreover, lobar ICH is associated with a higher risk of acute symptomatic seizures []. There is also a well-established link between CAA and cognitive decline, highlighting the broader clinical implications of this vascular pathology in the aging brain [].
3.3. Diagnostic Pitfalls
Diagnosis in older adults with ICH is frequently challenged by overlapping comorbidities, pre-existing neurocognitive dysfunction and a higher degree of frailty []. One significant diagnostic pitfall lies in the difficulty of distinguishing new neurological deficits from existing ones, especially for those already diagnosed with conditions like ischemic stroke, dementia, Parkinson’s disease, or chronic subdural hematoma []. Furthermore, cerebral atrophy in advanced age can reduce mass effect and mitigate intracranial pressure elevations, thereby attenuating clinical signs despite significant hemorrhage []. As a result, clinicians may not consider or underestimate the severity of ICH and delay crucial neuroimaging. Likewise, a relatively normal neurological assessment score, such as the National Institutes of Health Stroke Scale (NIHSS) or Glasgow Coma Scale (GCS), may not accurately reflect the underlying severity of injury in this age group []. A low threshold for the early use of neuroimaging, particularly non-contrast CT scans, is critical for distinguishing ICH from other possible causes in this age group.
3.4. Prognosis in Aging Populations
Advanced age is consistently associated with worse outcomes following ICH []. In addition to increased acute-phase mortality, older adults exhibit decreased functional recovery and are more likely to experience persistent disability and post-ICH cognitive decline []. The risk of fatal complications, such as recurrent ICH, expansion of the hematoma, aspiration pneumonia, or multisystem failure, is magnified by age-related physiological vulnerabilities and multiple comorbidities []. The cumulative burden of cSVD, including silent cerebral infarctions, white matter lesions, and CMBs, further compromises plasticity and post-stroke recovery []. Moreover, older adults have a higher likelihood of requiring long-term institutional care or assistance with basic activities of daily living [].
3.5. Predictors of Outcome
Prognosis in older ICH patients is determined by a combination of clinical [], genetic [], radiological, and age-specific factors [,]. Established predictors of a worse outcome include larger hematoma volume [], deep hematoma location [], intraventricular extension, initial low level of consciousness [], advanced age, and an anticoagulated state []. Regarding older adults, additional factors such as baseline frailty, cognitive impairment, and the degree of functional independence prior to ICH play a significant role []. Frailty, as assessed by validated tools like the Clinical Frailty Scale, independently predicts poor neurological recovery, longer hospitalizations, and a greater likelihood of discharge to long-term care []. Similarly, the presence of multiple CMBs and cSS may signal underlying CAA and portend a higher risk of recurrence and long-term cognitive impairment []. Comprehensive assessment is especially important in older ICH patients to guide acute management and rehabilitation [].
4. Management Considerations
The management of ICH in older adults requires a comprehensive approach that integrates acute phase management, personalized treatment planning, and secondary prevention.
4.1. Acute Phase Care
Structured ICH care bundles have been introduced to support the timely and standardized delivery of acute-phase care []. These bundles include rapid triage and neuroimaging, intensive blood pressure control [], immediate reversal of anticoagulation, neurosurgical consultation, and transfer to a dedicated stroke unit []. Care bundles such as the ABC-ICH protocol (Rapid Anticoagulant reversal, Intensive Blood pressure lowering, and a Care pathway for prompt neurosurgical referral) have demonstrated improved outcomes by reducing functional disability and in-hospital mortality, when embedded into daily practice []. The initial emergency response begins with immediate attention to the patient’s airway, breathing, circulation, and rapid neuroimaging []. Once the diagnosis of ICH is confirmed, care bundle protocols are implemented (“code ICH” as an analogy to “code stroke”) [].
Antihypertensive treatment should be initiated in patients presenting within six hours of symptom onset with SBP ≥150 mmHg [,]. Although the benefit is less certain, treatment may still be considered in cases where symptom onset is ≥6 h or unknown if SBP is ≥150 mmHg [,]. The treatment target is an SBP ≤140 mmHg, which should be maintained consistently for seven days []. The first dose of antihypertensive medication should be administered within 30 min of hospital arrival, and the target SBP should be achieved within 60 min [,]. However, abrupt large SBP reductions of 60 mmHg within one hour should be avoided []. Notably, older adults, who may have impaired cerebral autoregulation, are at greater risk for hypoperfusion with aggressive SBP lowering and require more individualized approaches [].
Immediate discontinuation and prompt reversal of anticoagulation are critical steps in the acute management of ICH, as continued anticoagulant activity may contribute to hematoma expansion and worse clinical outcomes []. Reversal protocols must be tailored to the specific type of anticoagulant involved, with a door-to-needle time of ≤30 min [,]. For patients on vitamin K antagonists with an international normalized ratio (INR) ≥1.3, reversal should include intravenous vitamin K and rapid administration of prothrombin complex concentrate (PCC) []. In contrast, patients receiving direct oral anticoagulants (DOACs) require agent-specific reversal strategies, such as idarucizumab for dabigatran [], and andexanet alfa for factor Xa inhibitors like apixaban, rivaroxaban, or edoxaban [,]. PCC may be used as an alternative if a specific DOAC reversal agent is either unavailable or unlicensed []. Heparins can be reversed by protamine-sulphate []. Recombinant activated factor VII (rFVIIa) [] and tranexamic acid may be considered as an adjunctive hemostatic agent in cases where specific reversal therapies are unavailable or contraindicated [,].
Surgical management of ICH in older adults, such as craniotomy or minimally invasive evacuation, should be made on a case-by-case basis by a multidisciplinary team within six hours []. The potential benefits of surgical intervention in this population must be carefully balanced against procedural risks, the likelihood of meaningful neurological recovery, the burden of postoperative rehabilitation, and the patient’s values and goals of care []. The decision to pursue surgical treatment must go beyond radiological findings and consider key age-related factors such as baseline functional status, cognitive reserve, comorbid conditions, and overall frailty []. Suggested criteria include patients with a pre-morbid modified Rankin Scale (mRS) score of ≤2 and a reasonable prognosis, along with one or more of the following: a GCS score ≤13, a supratentorial ICH volume ≥20 mL, posterior fossa ICH, or obstruction of the third or fourth ventricles [,]. The most likely candidates to benefit from minimally invasive surgery are both sexes, age of 30 to 80 years with superficial hematoma, GCS score of ≥9, hematoma volume between 25 and 40 mL, and undergoing surgery within 72 h of the symptom onset []. When the decision of hematoma evacuation has been made, the potential benefit of obtaining histological specimen of the leptomeninges and cortex to provide definite etiological diagnosis of a lobar ICH (with particular focus on CAA) should be kept in mind by neurosurgeons.
ICH care bundle also includes maintenance of normothermia and normoglycemia together with normotonia up to 7 days [,,]. Finally, consistent evidence supports the management of ICH patients in dedicated stroke units, which significantly improves outcomes by reducing mortality and long-term functional dependence compared to general medical wards []. The structured acute care bundles for intracerebral hemorrhage are summarized in Figure 3.
Figure 3.
Acute care bundles for patients with intracerebral hemorrhage (BP, blood pressure; SBP, systolic BP).
4.2. Tailored Strategies for Older Adults
Managing ICH in older adults requires a patient-centered approach that takes into account the complex interplay of age-related factors, biological frailty, pre-existing neurological and cognitive status, comorbidities, and, most critically, the patient’s goals of care []. The potential benefits and risks must be carefully considered when identifying patients who are most likely to derive meaningful benefit from more aggressive treatments []. Multidisciplinary evaluations involving geriatrics, neurology, neurosurgery and ethics teams are essential to determine appropriate treatment plans []. Beyond acute medical management, focused approaches for early mobilization, optimizing nutrition, preventing and managing delirium, and avoiding hospital-associated complications play a critical role in improving quality of life [,,].
4.3. Secondary Prevention
The cornerstone of secondary prevention after ICH is adequate etiological diagnosis. Since the vast majority of deep ICH are likely to be secondary to hypertensive arteriopathy, the comprehensive diagnostic work-up is especially relevant for ICH with lobar location. After ruling out secondary (i.e., structural) etiologies in the acute setting (including vascular malformations and cerebral venous sinus thrombosis), a contrast enhanced MRI is to be performed to evaluate a possible underlying neoplasm. The optimal timing of MRI is uncertain, but ideally after a few (up to 6–8) weeks to allow for adequate visualization by regression and aging of the hematoma, with the most recent guidelines suggesting its repeat after three to six months if negative []. Once secondary etiologies are excluded, thus a primary, i.e., cSVD etiology is suspected, these MRI scans enable the etiological differential diagnosis of CAA and hypertensive arteriopathy, following the probable CAA diagnosis as per the v2.0 Boston criteria []. Figure 4 illustrates the proposed diagnostic work-up for lobar ICH, including acute-phase and follow-up imaging. Though most cases with mixed (i.e., lobar and deep) hemorrhagic alteration patterns are likely to be due to hypertensive arteriopathy only, addressing concomitant CAA in these patients with cerebrospinal fluid and plasma biomarkers as well as with positron emission tomography (PET) is of extensive research interest.
Figure 4.
Proposed diagnostic work-up for lobar intracerebral hemorrhage. (CAA, cerebral amyloid angiopathy; CHA, cerebral hypertensive arteriopathy; CT, computed tomography; CTA, CT angiography; CTV, CT venography; CVST, cerebral venous sinus thrombosis; SVD, small vessel disease; SWI, susceptibility-weighted imaging).
In detail, cerebrospinal fluid levels of β-amyloid1–42, β-amyloid1–40, and β-amyloid1–38 have been consistently found decreased compared to controls, reflecting cerebral β-amyloid deposition, with prominent vascular involvement. Since β-amyloid1–40 and β-amyloid1–38 are abundant in CAA vessels but less so in senile plaques, these decreases in β-amyloid1–40 and β-amyloid1–38 appear to be specific to CAA, and not characteristic to AD in general. On the other hand, the concentrations of Tau (as a general marker of neuronal loss) and phosphorylated Tau181 were generally higher compared to controls but lower compared to AD at the cohort levels, reflecting a subpopulation of CAA with concomitant advanced AD neuropathologic change [,]. Study on the plasma levels of β-amyloids and other core AD biomarkers have so far provided incoherent results [,]. The assessment of cerebral β-amyloid and aggregated Tau pathologies by PET can be reasonable alternative to cerebrospinal fluid analyses; however, evidence regarding aggregated Tau PET (by using the tracer [18F]T807) has only recently emerged [,]. Though the classical and clinically approved tracers of β-amyloid (such as [11C]PIB, [18F]florbetapir, [18F]florbetaben, and [18F]flutemetamol) cannot make distinction between vascular and parenchymal plaque pathology, a recently reported ligand [18F]K10-008 might provide a novel vascular-specific alternative in the future [].
Hypertension remains the most significant modifiable risk factor for ICH recurrence, and its management is particularly critical in the older adults []. Regular monitoring and dose adjustments are essential to avoid complications such as orthostatic hypotension and cerebral hypoperfusion, which can lead to falls, syncope, or cognitive decline [].
The resumption of antithrombotic therapy presents a complex clinical challenge and the risk of recurrent ICH versus thromboembolic events need to be weighed []. Factors that increase hemorrhagic risk are older age, already recurrent ICH, lobar ICH, higher number and lobar location of CMBs, CAA (especially when with cSS), uncontrolled hypertension and ApoE ε2 allele [,]. Restarting antiplatelet therapy for an ischemic vascular comorbidity seems safe in ICH survivors and not only decreases the risk of thromboembolic events but also that of recurrent hemorrhage [,]. Patients with an extremely high thromboembolic risk such as mechanical heart valves and left ventricular assistance devices require anticoagulant resumption as soon as possible, no later than two weeks []. Restaring anticoagulation in patients with atrial fibrillation requires more consideration []. For patients at a low to moderate risk of recurrent hemorrhage it is reasonable to resume oral anticoagulation after two months, however for those at a very high hemorrhagic risk left atrial appendage closure may be explored as an alternative []. Healthy diet, smoking cessation, alcohol moderation, and increased physical activity can also optimize vascular health []. In older adults with CAA or extensive lobar hemorrhagic alterations identified on SWI MRI, the risk of recurrent ICH may outweigh the benefits of anticoagulation []. These imaging biomarkers have become critical tools for stratifying hemorrhagic risk, yet there remains no consensus on precise thresholds for safe resumption of anticoagulants, highlighting the need for individualized decision-making [].
4.4. Emerging Therapeutic Directions
Recent advances in understanding the vascular vulnerabilities associated with aging have opened promising paths for preventing and treating ICH and related CMBs. These emerging therapies target the biological mechanisms underlying vascular fragility and secondary brain injury, aiming to modify the disease course beyond traditional acute interventions [].
Chronic neuroinflammation plays a crucial role in secondary injury following ICH []. Prolonged activation of microglia and astrocytes leads to the release of pro-inflammatory cytokines and oxidative stress, exacerbating edema, neuronal injury, and blood–brain barrier disruption [,]. Novel agents targeting this cascade, such as inhibitors of the NLRP3 inflammasome are under active investigation []. These therapies may reduce perihematomal damage and improve neurological recovery by dampening maladaptive inflammatory responses, particularly in older patients with heightened inflammatory vulnerability [].
Senolytic therapies represent another innovative strategy, selectively targeting senescent glial and endothelial cells that accumulate with age and contribute to endothelial dysfunction and vascular instability []. Preclinical studies demonstrate that senolytics can restore vascular integrity, reduce blood–brain barrier disruption, and diminish hemorrhagic risk, providing a novel mechanism for neurovascular protection in aging brains [,].
Additionally, declining systemic levels of IGF-1 with aging have been implicated in weakening neurovascular structures []. Strategies to restore or mimic IGF-1 signaling may strengthen fragile cerebral vessels, enhancing resilience against hemorrhage []. This reflects a broader concept of targeting the biological aging process of the vasculature to prevent ICH [].
In CAA, decreasing neuroinflammation by minocycline in part through decreasing MMP-2 and MMP-9 activity is currently of extensive research interest [,]. In addition, a multicenter phase 2 clinical trial with Mivelsiran, an intrathecally administered RNA interference therapeutic targeting the APP with the aim to decrease the load of pathological β-amyloid and thus the development of future lobar hemorrhagic alterations is underway (NCT06393712). While these approaches remain largely in experimental or early clinical stages, they signify an important shift toward preventative therapies focused on vascular health and biological aging regulation, rather than solely on acute hemorrhage management.
4.5. Ethical and Palliative Care Aspects
The management of ICH in frail older adults, especially those with limited life expectancy or low likelihood of meaningful neurological recovery, demands ethically grounded decision-making []. Clinicians must navigate the delicate balance between the potential benefits of aggressive interventions and the associated physical, emotional, and functional burdens []. The integration of palliative care becomes crucial when life-prolonging therapies are unlikely to improve outcomes or are against the patient’s goals of care [,]. Early palliative involvement focuses on symptom relief, psychosocial support, and ensuring patient comfort and dignity throughout the disease course []. Engaging in early discussions about advanced directives and goals of care allows patients and families to make informed decisions that align with their values and preferences [].
5. Conclusions
ICH in the aging population arises from a complex interplay of vascular aging processes, including endothelial dysfunction, BBB breakdown, impaired autoregulation, chronic inflammation, and systemic endocrine dysregulation. These mechanisms collectively weaken the structural and functional integrity of the cerebral microvasculature, promoting both covert hemorrhage and subclinical lesions such as CMBs. Increasing evidence suggests that CMBs and ICH are not isolated phenomena but rather represent different manifestations along a shared continuum of hemorrhagic small vessel disease.
Despite the rising incidence of ICH, CMBs, and cSAH/cSS in older adults, this population remains underrepresented in clinical trials, limiting the applicability of current evidence to those most at risk. There is an urgent need to develop and validate biomarkers of vascular aging and rupture risk, which could support early detection, risk stratification, and individualized prevention strategies.
Future research should prioritize the mechanistic targeting of aging pathways, including interventions that restore endothelial resilience, stabilize cerebral autoregulation, correct systemic hormonal imbalances such as IGF-1 deficiency, and act against inflammatory alterations accompanied by cSVDs. Furthermore, growing recognition of the link between CMBs, ICH, and VCID highlights the importance of addressing long-term cognitive outcomes in ICH survivors.
Ultimately, advancing our understanding of how aging predisposes the brain to hemorrhagic injury will be essential to guide the development of age-aware, personalized therapeutic approaches. Integrating geroscience principles into stroke research may pave the way for more effective prevention, better post-ICH care, and improved outcomes for older adults at risk of cerebrovascular decline.
Author Contributions
Conceptualization, A.L., B.G. and E.Z.; literature review, A.L., D.L.-E., E.Z., L.S. and P.T.; writing—original draft preparation, A.L., E.Z. and L.S.; writing—review and editing, B.G., D.L.-E., L.S. and P.T.; supervision, B.G.; project administration, B.G.; funding acquisition, L.S. All authors have read and agreed to the published version of the manuscript.
Funding
This work was also supported by the EKÖP-2024-9 New National Excellence Program of the Ministry for Culture and Innovation from the source of the National Research, Development and Innovation Fund. LS was supported by grant 5S 725 (A202) by the University of Szeged. Further financial support was received from the National Research, Development and Innovation Office (OTKA K-134555, OTKA FK-146334 to PT), the National Clinical Neuroscience Laboratory (RRF-2.3.1-21-2022-00011), and the Thematic Excellence Program 2021 Health sub-program of the Ministry for Innovation and Technology in Hungary, within the framework of the EGA-16 project of the University of Pecs (to PT). The funding sources had no role in the study design, in the collection, analysis and interpretation of data, in the writing of the report, or in the decision to submit the article for publication. The content is solely the responsibility of the authors and does not necessarily represent the official views of the funding agencies.
Institutional Review Board Statement
Not applicable.
Informed Consent Statement
Not applicable.
Data Availability Statement
Not applicable.
Acknowledgments
The 4o version of ChatGPT, developed by OpenAI, and Claude 3.5 Sonnet, developed by Anthropic, were used as a language tool to refine our writing and enhance the clarity of our work. The authors have reviewed and edited the output and take full responsibility for the content of this publication.
Conflicts of Interest
The authors declare no conflicts of interest.
References
- Watson, N.; Bonsack, F.; Sukumari-Ramesh, S. Intracerebral Hemorrhage: The Effects of Aging on Brain Injury. Front. Aging Neurosci. 2022, 14, 859067. [Google Scholar] [CrossRef]
- Hostettler, I.C.; Seiffge, D.J.; Werring, D.J. Intracerebral hemorrhage: An update on diagnosis and treatment. Expert. Rev. Neurother. 2019, 19, 679–694. [Google Scholar] [CrossRef]
- Malhotra, K.; Zompola, C.; Theodorou, A.; Katsanos, A.H.; Shoamanesh, A.; Gupta, H.; Beshara, S.; Goyal, N.; Chang, J.; Tayal, A.H.; et al. Prevalence, Characteristics, and Outcomes of Undetermined Intracerebral Hemorrhage: A Systematic Review and Meta-Analysis. Stroke 2021, 52, 3602–3612. [Google Scholar] [CrossRef]
- Rajashekar, D.; Liang, J.W. Intracerebral Hemorrhage. In StatPearls; StatPearls Publishing LLC.: Treasure Island, FL, USA, 2025. [Google Scholar]
- Stein, M.; Misselwitz, B.; Hamann, G.F.; Scharbrodt, W.; Schummer, D.I.; Oertel, M.F. Intracerebral hemorrhage in the very old: Future demographic trends of an aging population. Stroke 2012, 43, 1126–1128. [Google Scholar] [CrossRef]
- Wang, Z.W.; Wan, M.P.; Tai, J.H.; Wang, Y.; Yin, M.Y. Global regional and national burden of intracerebral hemorrhage between 1990 and 2021. Sci. Rep. 2025, 15, 3624. [Google Scholar] [CrossRef]
- Clancy, U.; Kancheva, A.K.; Valdes Hernandez, M.D.C.; Jochems, A.C.C.; Munoz Maniega, S.; Quinn, T.J.; Wardlaw, J.M. Imaging Biomarkers of VCI: A Focused Update. Stroke 2024, 55, 791–800. [Google Scholar] [CrossRef] [PubMed]
- Arndt, P.; Chahem, C.; Luchtmann, M.; Kuschel, J.N.; Behme, D.; Pfister, M.; Neumann, J.; Gortler, M.; Dorner, M.; Pawlitzki, M.; et al. Risk factors for intracerebral hemorrhage in small-vessel disease and non-small-vessel disease etiologies-an observational proof-of-concept study. Front. Neurol. 2024, 15, 1322442. [Google Scholar] [CrossRef] [PubMed]
- Zhu, Y.; Liu, L.; Zhong, L.; Cheng, Y.; Zhang, S.; Wu, B.; Wang, D.; Xu, M. The association between hypertensive angiopathy and cerebral amyloid angiopathy in primary intracerebral hemorrhage. Front. Neurol. 2023, 14, 1257896. [Google Scholar] [CrossRef] [PubMed]
- Chen, S.J.; Tsai, H.H.; Lo, Y.L.; Chen, Y.F.; Tang, S.C.; Jeng, J.S.; Tsai, L.K. Interaction between cerebral small vessel disease, blood pressure, and remote ischemic lesions in acute spontaneous intracerebral hemorrhage. Eur. Stroke J. 2023, 8, 828–835. [Google Scholar] [CrossRef]
- Boulouis, G.; Charidimou, A.; Auriel, E.; Haley, K.E.; van Etten, E.S.; Fotiadis, P.; Reijmer, Y.; Ayres, A.; Schwab, K.M.; Martinez-Ramirez, S.; et al. Intracranial atherosclerosis and cerebral small vessel disease in intracerebral hemorrhage patients. J. Neurol. Sci. 2016, 369, 324–329. [Google Scholar] [CrossRef]
- Gao, Y.; Zong, C.; Liu, H.; Zhang, K.; Yang, H.; Wang, Y.; Li, Y.; Song, B.; Xu, Y. Clinical features and associated factors of coexisting intracerebral hemorrhage in patients with cerebral small vessel disease: A cross-sectional study. Sci. Rep. 2024, 14, 5596. [Google Scholar] [CrossRef]
- Ungvari, Z.; Tarantini, S.; Kirkpatrick, A.C.; Csiszar, A.; Prodan, C.I. Cerebral microhemorrhages: Mechanisms, consequences, and prevention. Am. J. Physiol. Heart Circ. Physiol. 2017, 312, H1128–H1143. [Google Scholar] [CrossRef]
- Caunca, M.R.; Del Brutto, V.; Gardener, H.; Shah, N.; Dequatre-Ponchelle, N.; Cheung, Y.K.; Elkind, M.S.; Brown, T.R.; Cordonnier, C.; Sacco, R.L.; et al. Cerebral Microbleeds, Vascular Risk Factors, and Magnetic Resonance Imaging Markers: The Northern Manhattan Study. J. Am. Heart Assoc. 2016, 5, e003477. [Google Scholar] [CrossRef]
- Markus, H.S.; de Leeuw, F.E. Cerebral small vessel disease: Recent advances and future directions. Int. J. Stroke 2023, 18, 4–14. [Google Scholar] [CrossRef] [PubMed]
- Graff-Radford, J.; Simino, J.; Kantarci, K.; Mosley, T.H., Jr.; Griswold, M.E.; Windham, B.G.; Sharrett, A.R.; Albert, M.S.; Gottesman, R.F.; Jack, C.R., Jr.; et al. Neuroimaging Correlates of Cerebral Microbleeds: The ARIC Study (Atherosclerosis Risk in Communities). Stroke 2017, 48, 2964–2972. [Google Scholar] [CrossRef] [PubMed]
- Greenberg, S.M.; Vernooij, M.W.; Cordonnier, C.; Viswanathan, A.; Al-Shahi Salman, R.; Warach, S.; Launer, L.J.; Van Buchem, M.A.; Breteler, M.M. Cerebral microbleeds: A guide to detection and interpretation. Lancet Neurol. 2009, 8, 165–174. [Google Scholar] [CrossRef] [PubMed]
- Lee, S.H.; Bae, H.J.; Kwon, S.J.; Kim, H.; Kim, Y.H.; Yoon, B.W.; Roh, J.K. Cerebral microbleeds are regionally associated with intracerebral hemorrhage. Neurology 2004, 62, 72–76. [Google Scholar] [CrossRef]
- Shams, S.; Martola, J.; Granberg, T.; Li, X.; Shams, M.; Fereshtehnejad, S.M.; Cavallin, L.; Aspelin, P.; Kristoffersen-Wiberg, M.; Wahlund, L.O. Cerebral microbleeds: Different prevalence, topography, and risk factors depending on dementia diagnosis-the Karolinska Imaging Dementia Study. AJNR Am. J. Neuroradiol. 2015, 36, 661–666. [Google Scholar] [CrossRef]
- Benedictus, M.R.; Prins, N.D.; Goos, J.D.; Scheltens, P.; Barkhof, F.; van der Flier, W.M. Microbleeds, Mortality, and Stroke in Alzheimer Disease: The MISTRAL Study. JAMA Neurol. 2015, 72, 539–545. [Google Scholar] [CrossRef]
- Akoudad, S.; Portegies, M.L.; Koudstaal, P.J.; Hofman, A.; van der Lugt, A.; Ikram, M.A.; Vernooij, M.W. Cerebral Microbleeds Are Associated With an Increased Risk of Stroke: The Rotterdam Study. Circulation 2015, 132, 509–516. [Google Scholar] [CrossRef]
- Ghelmez, D.; Sorin Tuta, S.; Popa, C. Cerebral microbleeds (CMBs)—Relevance for mechanisms of cerebral hemorrhage--analysis of 24 MRI evaluated patients. J. Med. Life 2013, 6, 437–439. [Google Scholar]
- Wardlaw, J.M.; Smith, C.; Dichgans, M. Small vessel disease: Mechanisms and clinical implications. Lancet Neurol. 2019, 18, 684–696. [Google Scholar] [CrossRef]
- Incontri, D.; Marchina, S.; Andreev, A.; Wilson, M.; Wang, J.Y.; Lin, D.; Heistand, E.C.; Carvalho, F.; Selim, M.; Lioutas, V.A. Etiology of Primary Cerebellar Intracerebral Hemorrhage Based on Topographic Localization. Stroke 2023, 54, 3074–3080. [Google Scholar] [CrossRef] [PubMed]
- Okine, D.N.; Knopman, D.S.; Mosley, T.H.; Wong, D.F.; Johansen, M.C.; Walker, K.A.; Jack, C.R., Jr.; Kantarci, K.; Pike, J.R.; Graff-Radford, J.; et al. Cerebral Microbleed Patterns and Cortical Amyloid-beta: The ARIC-PET Study. Stroke 2023, 54, 2613–2620. [Google Scholar] [CrossRef] [PubMed]
- Shoamanesh, A.; Kwok, C.S.; Benavente, O. Cerebral microbleeds: Histopathological correlation of neuroimaging. Cerebrovasc. Dis. 2011, 32, 528–534. [Google Scholar] [CrossRef] [PubMed]
- Iadecola, C. The pathobiology of vascular dementia. Neuron 2013, 80, 844–866. [Google Scholar] [CrossRef]
- Wardlaw, J.M.; Smith, E.E.; Biessels, G.J.; Cordonnier, C.; Fazekas, F.; Frayne, R.; Lindley, R.I.; O’Brien, J.T.; Barkhof, F.; Benavente, O.R.; et al. Neuroimaging standards for research into small vessel disease and its contribution to ageing and neurodegeneration. Lancet Neurol. 2013, 12, 822–838. [Google Scholar] [CrossRef]
- Ungvari, A.; Nyul-Toth, A.; Patai, R.; Csik, B.; Gulej, R.; Nagy, D.; Shanmugarama, S.; Benyo, Z.; Kiss, T.; Ungvari, Z.; et al. Cerebromicrovascular senescence in vascular cognitive impairment: Does accelerated microvascular aging accompany atherosclerosis? Geroscience 2025, 47, 5511–5524. [Google Scholar] [CrossRef]
- Patai, R.; Patel, K.; Csik, B.; Gulej, R.; Nagaraja, R.Y.; Nagy, D.; Chandragiri, S.S.; Shanmugarama, S.; Kordestan, K.V.; Nagykaldi, M.; et al. Aging, mitochondrial dysfunction, and cerebral microhemorrhages: A preclinical evaluation of SS-31 (elamipretide) and development of a high-throughput machine learning-driven imaging pipeline for cerebromicrovascular protection therapeutic screening. Geroscience 2025, 47, 4871–4887. [Google Scholar] [CrossRef]
- Kallai, A.; Ungvari, A.; Csaban, D.; Orfi, Z.; Lehoczki, A.; Harasztdombi, J.; Yabluchanskiy, A.; Benyo, Z.; Szappanos, A.; Tarantini, S.; et al. Clonal hematopoiesis of indeterminate potential (CHIP) in cerebromicrovascular aging: Implications for vascular contributions to cognitive impairment and dementia (VCID). Geroscience 2025, 47, 2739–2775. [Google Scholar] [CrossRef]
- Stankovics, L.; Ungvari, A.; Fekete, M.; Nyul-Toth, A.; Mukli, P.; Patai, R.; Csik, B.; Gulej, R.; Conley, S.; Csiszar, A.; et al. The vasoprotective role of IGF-1 signaling in the cerebral microcirculation: Prevention of cerebral microhemorrhages in aging. Geroscience 2024, 47, 445–455. [Google Scholar] [CrossRef] [PubMed]
- Miller, L.R.; Bickel, M.A.; Vance, M.L.; Vaden, H.; Nagykaldi, D.; Nyul-Toth, A.; Bullen, E.C.; Gautam, T.; Tarantini, S.; Yabluchanskiy, A.; et al. Vascular smooth muscle cell-specific Igf1r deficiency exacerbates the development of hypertension-induced cerebral microhemorrhages and gait defects. Geroscience 2024, 46, 3481–3501. [Google Scholar] [CrossRef] [PubMed]
- Csiszar, A.; Ungvari, A.; Patai, R.; Gulej, R.; Yabluchanskiy, A.; Benyo, Z.; Kovacs, I.; Sotonyi, P.; Kirkpartrick, A.C.; Prodan, C.I.; et al. Atherosclerotic burden and cerebral small vessel disease: Exploring the link through microvascular aging and cerebral microhemorrhages. Geroscience 2024, 46, 5103–5132. [Google Scholar] [CrossRef] [PubMed]
- Faakye, J.; Nyul-Toth, A.; Muranyi, M.; Gulej, R.; Csik, B.; Shanmugarama, S.; Tarantini, S.; Negri, S.; Prodan, C.; Mukli, P.; et al. Preventing spontaneous cerebral microhemorrhages in aging mice: A novel approach targeting cellular senescence with ABT263/navitoclax. Geroscience 2023, 46, 21–37. [Google Scholar] [CrossRef]
- Nyul-Toth, A.; Fulop, G.A.; Tarantini, S.; Kiss, T.; Ahire, C.; Faakye, J.A.; Ungvari, A.; Toth, P.; Toth, A.; Csiszar, A.; et al. Cerebral venous congestion exacerbates cerebral microhemorrhages in mice. Geroscience 2022, 44, 805–816. [Google Scholar] [CrossRef]
- Nyul-Toth, A.; Tarantini, S.; Kiss, T.; Toth, P.; Galvan, V.; Tarantini, A.; Yabluchanskiy, A.; Csiszar, A.; Ungvari, Z. Increases in hypertension-induced cerebral microhemorrhages exacerbate gait dysfunction in a mouse model of Alzheimer’s disease. Geroscience 2020, 42, 1685–1698. [Google Scholar] [CrossRef]
- Tarantini, S.; Valcarcel-Ares, N.M.; Yabluchanskiy, A.; Springo, Z.; Fulop, G.A.; Ashpole, N.; Gautam, T.; Giles, C.B.; Wren, J.D.; Sonntag, W.E.; et al. Insulin-like growth factor 1 deficiency exacerbates hypertension-induced cerebral microhemorrhages in mice, mimicking the aging phenotype. Aging Cell 2017, 16, 469–479. [Google Scholar] [CrossRef]
- Toth, P.; Tarantini, S.; Springo, Z.; Tucsek, Z.; Gautam, T.; Giles, C.B.; Wren, J.D.; Koller, A.; Sonntag, W.E.; Csiszar, A.; et al. Aging exacerbates hypertension-induced cerebral microhemorrhages in mice: Role of resveratrol treatment in vasoprotection. Aging Cell 2015, 14, 400–408. [Google Scholar] [CrossRef]
- Wardlaw, J.M.; Smith, C.; Dichgans, M. Mechanisms of sporadic cerebral small vessel disease: Insights from neuroimaging. Lancet Neurol. 2013, 12, 483–497. [Google Scholar] [CrossRef]
- Iadecola, C.; Duering, M.; Hachinski, V.; Joutel, A.; Pendlebury, S.T.; Schneider, J.A.; Dichgans, M. Vascular Cognitive Impairment and Dementia: JACC Scientific Expert Panel. J. Am. Coll. Cardiol. 2019, 73, 3326–3344. [Google Scholar] [CrossRef]
- Tarantini, S.; Balasubramanian, P.; Delfavero, J.; Csipo, T.; Yabluchanskiy, A.; Kiss, T.; Nyul-Toth, A.; Mukli, P.; Toth, P.; Ahire, C.; et al. Treatment with the BCL-2/BCL-xL inhibitor senolytic drug ABT263/Navitoclax improves functional hyperemia in aged mice. Geroscience 2021, 43, 2427–2440. [Google Scholar] [CrossRef]
- Gulej, R.; Nyul-Toth, A.; Csik, B.; Petersen, B.; Faakye, J.; Negri, S.; Chandragiri, S.S.; Mukli, P.; Yabluchanskiy, A.; Conley, S.; et al. Rejuvenation of cerebromicrovascular function in aged mice through heterochronic parabiosis: Insights into neurovascular coupling and the impact of young blood factors. Geroscience 2024, 46, 327–347. [Google Scholar] [CrossRef] [PubMed]
- Gulej, R.; Nyul-Toth, A.; Csik, B.; Patai, R.; Petersen, B.; Negri, S.; Chandragiri, S.S.; Shanmugarama, S.; Mukli, P.; Yabluchanskiy, A.; et al. Young blood-mediated cerebromicrovascular rejuvenation through heterochronic parabiosis: Enhancing blood-brain barrier integrity and capillarization in the aged mouse brain. Geroscience 2024, 46, 4415–4442. [Google Scholar] [CrossRef] [PubMed]
- Kiss, T.; Nyul-Toth, A.; Gulej, R.; Tarantini, S.; Csipo, T.; Mukli, P.; Ungvari, A.; Balasubramanian, P.; Yabluchanskiy, A.; Benyo, Z.; et al. Old blood from heterochronic parabionts accelerates vascular aging in young mice: Transcriptomic signature of pathologic smooth muscle remodeling. Geroscience 2022, 44, 953–981. [Google Scholar] [CrossRef] [PubMed]
- Kiss, T.; Tarantini, S.; Csipo, T.; Balasubramanian, P.; Nyul-Toth, A.; Yabluchanskiy, A.; Wren, J.D.; Garman, L.; Huffman, D.M.; Csiszar, A.; et al. Circulating anti-geronic factors from heterochonic parabionts promote vascular rejuvenation in aged mice: Transcriptional footprint of mitochondrial protection, attenuation of oxidative stress, and rescue of endothelial function by young blood. Geroscience 2020, 42, 727–748. [Google Scholar] [CrossRef]
- Pospiech, E.; Bar, A.; Pisarek-Pacek, A.; Karas, A.; Branicki, W.; Chlopicki, S. Epigenetic clock in the aorta and age-related endothelial dysfunction in mice. Geroscience 2024, 46, 3993–4002. [Google Scholar] [CrossRef]
- Waigi, E.W.; Pernomian, L.; Crockett, A.M.; Costa, T.J.; Townsend, P., Jr.; Webb, R.C.; McQuail, J.A.; McCarthy, C.G.; Hollis, F.; Wenceslau, C.F. Vascular dysfunction occurs prior to the onset of amyloid pathology and Abeta plaque deposits colocalize with endothelial cells in the hippocampus of female APPswe/PSEN1dE9 mice. Geroscience 2024, 46, 5517–5536. [Google Scholar] [CrossRef]
- Murugesan, N.; Demarest, T.G.; Madri, J.A.; Pachter, J.S. Brain regional angiogenic potential at the neurovascular unit during normal aging. Neurobiol. Aging 2012, 33, 1004.e1–1004.e16. [Google Scholar] [CrossRef]
- Benderro, G.F.; Lamanna, J.C. Hypoxia-induced angiogenesis is delayed in aging mouse brain. Brain Res. 2011, 1389, 50–60. [Google Scholar] [CrossRef]
- Ionescu-Tucker, A.; Cotman, C.W. Emerging roles of oxidative stress in brain aging and Alzheimer’s disease. Neurobiol. Aging 2021, 107, 86–95. [Google Scholar] [CrossRef]
- Shao, A.; Lin, D.; Wang, L.; Tu, S.; Lenahan, C.; Zhang, J. Oxidative Stress at the Crossroads of Aging, Stroke and Depression. Aging Dis. 2020, 11, 1537–1566. [Google Scholar] [CrossRef] [PubMed]
- Sivandzade, F.; Prasad, S.; Bhalerao, A.; Cucullo, L. NRF2 and NF-B interplay in cerebrovascular and neurodegenerative disorders: Molecular mechanisms and possible therapeutic approaches. Redox Biol. 2019, 21, 101059. [Google Scholar] [CrossRef] [PubMed]
- Forstermann, U.; Sessa, W.C. Nitric oxide synthases: Regulation and function. Eur. Heart J. 2012, 33, 829–837. [Google Scholar] [CrossRef] [PubMed]
- Donato, A.J.; Morgan, R.G.; Walker, A.E.; Lesniewski, L.A. Cellular and molecular biology of aging endothelial cells. J. Mol. Cell Cardiol. 2015, 89, 122–135. [Google Scholar] [CrossRef]
- Cummins, M.J.; Cresswell, E.T.; Bevege, R.J.; Smith, D.W. Aging disrupts blood-brain and blood-spinal cord barrier homeostasis, but does not increase paracellular permeability. Geroscience 2024, 47, 263–285. [Google Scholar] [CrossRef]
- Ting, K.K.; Coleman, P.; Kim, H.J.; Zhao, Y.; Mulangala, J.; Cheng, N.C.; Li, W.; Gunatilake, D.; Johnstone, D.M.; Loo, L.; et al. Vascular senescence and leak are features of the early breakdown of the blood-brain barrier in Alzheimer’s disease models. Geroscience 2023, 45, 3307–3331. [Google Scholar] [CrossRef]
- Zachariou, V.; Pappas, C.; Bauer, C.E.; Shao, X.; Liu, P.; Lu, H.; Wang, D.J.J.; Gold, B.T. Regional differences in the link between water exchange rate across the blood-brain barrier and cognitive performance in normal aging. Geroscience 2024, 46, 265–282. [Google Scholar] [CrossRef]
- van Dinther, M.; Voorter, P.H.M.; Zhang, E.; van Kuijk, S.M.J.; Jansen, J.F.A.; van Oostenbrugge, R.J.; Backes, W.H.; Staals, J. The neurovascular unit and its correlation with cognitive performance in patients with cerebral small vessel disease: A canonical correlation analysis approach. Geroscience 2024, 46, 5061–5073. [Google Scholar] [CrossRef]
- Nyul-Toth, A.; Patai, R.; Csiszar, A.; Ungvari, A.; Gulej, R.; Mukli, P.; Yabluchanskiy, A.; Benyo, Z.; Sotonyi, P.; Prodan, C.I.; et al. Linking peripheral atherosclerosis to blood-brain barrier disruption: Elucidating its role as a manifestation of cerebral small vessel disease in vascular cognitive impairment. Geroscience 2024, 46, 6511–6536. [Google Scholar] [CrossRef]
- Montagne, A.; Barnes, S.R.; Nation, D.A.; Kisler, K.; Toga, A.W.; Zlokovic, B.V. Imaging subtle leaks in the blood-brain barrier in the aging human brain: Potential pitfalls, challenges, and possible solutions. Geroscience 2022, 44, 1339–1351. [Google Scholar] [CrossRef]
- Kerkhofs, D.; Wong, S.M.; Zhang, E.; Uiterwijk, R.; Hoff, E.I.; Jansen, J.F.A.; Staals, J.; Backes, W.H.; van Oostenbrugge, R.J. Blood-brain barrier leakage at baseline and cognitive decline in cerebral small vessel disease: A 2-year follow-up study. Geroscience 2021, 43, 1643–1652. [Google Scholar] [CrossRef]
- Verheggen, I.C.M.; de Jong, J.J.A.; van Boxtel, M.P.J.; Postma, A.A.; Jansen, J.F.A.; Verhey, F.R.J.; Backes, W.H. Imaging the role of blood-brain barrier disruption in normal cognitive ageing. Geroscience 2020, 42, 1751–1764. [Google Scholar] [CrossRef]
- Verheggen, I.C.M.; de Jong, J.J.A.; van Boxtel, M.P.J.; Gronenschild, E.; Palm, W.M.; Postma, A.A.; Jansen, J.F.A.; Verhey, F.R.J.; Backes, W.H. Increase in blood-brain barrier leakage in healthy, older adults. Geroscience 2020, 42, 1183–1193. [Google Scholar] [CrossRef] [PubMed]
- Sweeney, M.D.; Zhao, Z.; Montagne, A.; Nelson, A.R.; Zlokovic, B.V. Blood-Brain Barrier: From Physiology to Disease and Back. Physiol. Rev. 2019, 99, 21–78. [Google Scholar] [CrossRef] [PubMed]
- Sweeney, M.D.; Kisler, K.; Montagne, A.; Toga, A.W.; Zlokovic, B.V. The role of brain vasculature in neurodegenerative disorders. Nat. Neurosci. 2018, 21, 1318–1331. [Google Scholar] [CrossRef] [PubMed]
- Real, M.G.C.; Falcione, S.R.; Boghozian, R.; Clarke, M.; Todoran, R.; St Pierre, A.; Zhang, Y.; Joy, T.; Jickling, G.C. Endothelial Cell Senescence Effect on the Blood-Brain Barrier in Stroke and Cognitive Impairment. Neurology 2024, 103, e210063. [Google Scholar] [CrossRef]
- Kumar, M.; Yan, P.; Kuchel, G.A.; Xu, M. Cellular Senescence as a Targetable Risk Factor for Cardiovascular Diseases: Therapeutic Implications: JACC Family Series. JACC Basic Transl. Sci. 2024, 9, 522–534. [Google Scholar] [CrossRef]
- Casanova, R.; Walker, K.A.; Justice, J.N.; Anderson, A.; Duggan, M.R.; Cordon, J.; Barnard, R.T.; Lu, L.; Hsu, F.C.; Sedaghat, S.; et al. Associations of plasma proteomics and age-related outcomes with brain age in a diverse cohort. Geroscience 2024, 46, 3861–3873. [Google Scholar] [CrossRef]
- Ruggiero, A.D.; Vemuri, R.; Blawas, M.; Long, M.; DeStephanis, D.; Williams, A.G.; Chen, H.; Justice, J.N.; Macauley, S.L.; Day, S.M.; et al. Long-term dasatinib plus quercetin effects on aging outcomes and inflammation in nonhuman primates: Implications for senolytic clinical trial design. Geroscience 2023, 45, 2785–2803. [Google Scholar] [CrossRef]
- Fielding, R.A.; Atkinson, E.J.; Aversa, Z.; White, T.A.; Heeren, A.A.; Achenbach, S.J.; Mielke, M.M.; Cummings, S.R.; Pahor, M.; Leeuwenburgh, C.; et al. Associations between biomarkers of cellular senescence and physical function in humans: Observations from the lifestyle interventions for elders (LIFE) study. Geroscience 2022, 44, 2757–2770. [Google Scholar] [CrossRef]
- Fang, Y.; Peck, M.R.; Quinn, K.; Chapman, J.E.; Medina, D.; McFadden, S.A.; Bartke, A.; Hascup, E.R.; Hascup, K.N. Senolytic intervention improves cognition, metabolism, and adiposity in female APP(NL)(-F/NL-F) mice. Geroscience 2024, 47, 1123–1138. [Google Scholar] [CrossRef]
- Cummings, S.R.; Lui, L.Y.; Zaira, A.; Mau, T.; Fielding, R.A.; Atkinson, E.J.; Patel, S.; LeBrasseur, N. Biomarkers of cellular senescence and major health outcomes in older adults. Geroscience 2024, 47, 3407–3415. [Google Scholar] [CrossRef]
- Boone, I.; Tuerlings, M.; Coutinho de Almeida, R.; Lehmann, J.; Ramos, Y.; Nelissen, R.; Slagboom, E.; de Keizer, P.; Meulenbelt, I. Identified senescence endotypes in aged cartilage are reflected in the blood metabolome. Geroscience 2024, 46, 2359–2369. [Google Scholar] [CrossRef] [PubMed]
- Teuliere, J.; Bernard, C.; Corel, E.; Lapointe, F.J.; Martens, J.; Lopez, P.; Bapteste, E. Network analyses unveil ageing-associated pathways evolutionarily conserved from fungi to animals. Geroscience 2023, 45, 1059–1080. [Google Scholar] [CrossRef] [PubMed]
- Marcozzi, S.; Bigossi, G.; Giuliani, M.E.; Giacconi, R.; Piacenza, F.; Cardelli, M.; Brunetti, D.; Segala, A.; Valerio, A.; Nisoli, E.; et al. Cellular senescence and frailty: A comprehensive insight into the causal links. Geroscience 2023, 45, 3267–3305. [Google Scholar] [CrossRef] [PubMed]
- Gulej, R.; Nyul-Toth, A.; Ahire, C.; DelFavero, J.; Balasubramanian, P.; Kiss, T.; Tarantini, S.; Benyo, Z.; Pacher, P.; Csik, B.; et al. Elimination of senescent cells by treatment with Navitoclax/ABT263 reverses whole brain irradiation-induced blood-brain barrier disruption in the mouse brain. Geroscience 2023, 45, 2983–3002. [Google Scholar] [CrossRef]
- Lambert, M.; Miquel, G.; Villeneuve, L.; Thorin-Trescases, N.; Thorin, E. The senolytic ABT-263 improves cognitive functions in middle-aged male, but not female, atherosclerotic LDLr(-/-);hApoB(100)(+/+) mice. Geroscience 2025, 47, 4577–4600. [Google Scholar] [CrossRef]
- Kieronska-Rudek, A.; Kij, A.; Bar, A.; Kurpinska, A.; Mohaissen, T.; Grosicki, M.; Stojak, M.; Sternak, M.; Buczek, E.; Proniewski, B.; et al. Phylloquinone improves endothelial function, inhibits cellular senescence, and vascular inflammation. Geroscience 2024, 46, 4909–4935. [Google Scholar] [CrossRef]
- Yang, C.; Pang, Y.; Huang, Y.; Ye, F.; Chen, X.; Gao, Y.; Zhang, C.; Yao, L.; Gao, J. Single-cell transcriptomics identifies premature aging features of TERC-deficient mouse brain and bone marrow. Geroscience 2022, 44, 2139–2155. [Google Scholar] [CrossRef]
- Kiss, T.; Nyul-Toth, A.; DelFavero, J.; Balasubramanian, P.; Tarantini, S.; Faakye, J.; Gulej, R.; Ahire, C.; Ungvari, A.; Yabluchanskiy, A.; et al. Spatial transcriptomic analysis reveals inflammatory foci defined by senescent cells in the white matter, hippocampi and cortical grey matter in the aged mouse brain. Geroscience 2022, 44, 661–681. [Google Scholar] [CrossRef]
- Kiss, T.; Nyul-Toth, A.; Balasubramanian, P.; Tarantini, S.; Ahire, C.; DelFavero, J.; Yabluchanskiy, A.; Csipo, T.; Farkas, E.; Wiley, G.; et al. Single-cell RNA sequencing identifies senescent cerebromicrovascular endothelial cells in the aged mouse brain. Geroscience 2020, 42, 429–444. [Google Scholar] [CrossRef]
- Kellogg, C.M.; Pham, K.; Machalinski, A.H.; Porter, H.L.; Blankenship, H.E.; Tooley, K.B.; Stout, M.B.; Rice, H.C.; Sharpe, A.L.; Beckstead, M.J.; et al. Microglial MHC-I induction with aging and Alzheimer’s is conserved in mouse models and humans. Geroscience 2023, 45, 3019–3043. [Google Scholar] [CrossRef]
- Csipo, T.; Lipecz, A.; Ashpole, N.M.; Balasubramanian, P.; Tarantini, S. Astrocyte senescence contributes to cognitive decline. Geroscience 2020, 42, 51–55. [Google Scholar] [CrossRef] [PubMed]
- Alshaebi, F.; Sciortino, A.; Kayed, R. The Role of Glial Cell Senescence in Alzheimer’s Disease. J. Neurochem. 2025, 169, e70051. [Google Scholar] [CrossRef] [PubMed]
- Fang, Y.; Medina, D.; Stockwell, R.; McFadden, S.; Quinn, K.; Peck, M.R.; Bartke, A.; Hascup, K.N.; Hascup, E.R. Sexual dimorphic metabolic and cognitive responses of C57BL/6 mice to Fisetin or Dasatinib and quercetin cocktail oral treatment. Geroscience 2023, 45, 2835–2850. [Google Scholar] [CrossRef] [PubMed]
- Baier, M.P.; Ranjit, R.; Owen, D.B.; Wilson, J.L.; Stiles, M.A.; Masingale, A.M.; Thomas, Z.; Bredegaard, A.; Sherry, D.M.; Logan, S. Cellular Senescence Is a Central Driver of Cognitive Disparities in Aging. Aging Cell 2025, 24, e70041. [Google Scholar] [CrossRef]
- Munoz-Espin, D.; Serrano, M. Cellular senescence: From physiology to pathology. Nat. Rev. Mol. Cell Biol. 2014, 15, 482–496. [Google Scholar] [CrossRef]
- Kollikowski, A.M.; Pham, M.; Marz, A.G.; Feick, J.; Vogt, M.L.; Xiong, Y.; Strinitz, M.; Vollmuth, C.; Essig, F.; Neugebauer, H.; et al. MMP-9 release into collateral blood vessels before endovascular thrombectomy to assess the risk of major intracerebral haemorrhages and poor outcome for acute ischaemic stroke: A proof-of-concept study. EBioMedicine 2024, 103, 105095. [Google Scholar] [CrossRef]
- Ho, W.M.; Chen, C.M.; Lee, Y.S.; Chang, K.H.; Chen, H.W.; Chen, S.T.; Chen, Y.C. Association of MMP-9 Haplotypes and TIMP-1 Polymorphism with Spontaneous Deep Intracerebral Hemorrhage in the Taiwan Population. PLoS ONE 2015, 10, e0125397. [Google Scholar] [CrossRef]
- Patai, R.; Csik, B.; Nyul-Toth, A.; Gulej, R.; Vali Kordestan, K.; Chandragiri, S.S.; Shanmugarama, S.; Tarantini, S.; Mukli, P.; Ungvari, A.; et al. Persisting blood-brain barrier disruption following cisplatin treatment in a mouse model of chemotherapy-associated cognitive impairment. Geroscience 2025, 47, 3835–3847. [Google Scholar] [CrossRef]
- Csik, B.; Vali Kordestan, K.; Gulej, R.; Patai, R.; Nyul-Toth, A.; Shanmugarama, S.; Mukli, P.; Ungvari, A.; Balsara, K.E.; McNall, R.Y.; et al. Cisplatin and methotrexate induce brain microvascular endothelial and microglial senescence in mouse models of chemotherapy-associated cognitive impairment. Geroscience 2025, 47, 3447–3459. [Google Scholar] [CrossRef] [PubMed]
- Patai, R.; Kiss, T.; Gulej, R.; Nyul-Toth, A.; Csik, B.; Chandragiri, S.S.; Shanmugarama, S.; Tarantini, S.; Ungvari, A.; Pacher, P.; et al. Transcriptomic profiling of senescence effects on blood-brain barrier-related gene expression in brain capillary endothelial cells in a mouse model of paclitaxel-induced chemobrain. Geroscience 2025, 47, 3677–3691. [Google Scholar] [CrossRef] [PubMed]
- Xu, M.; Zhu, Y.; Song, X.; Zhong, X.; Yu, X.; Wang, D.; Cheng, Y.; Tao, W.; Wu, B.; Liu, M. Pathological Changes of Small Vessel Disease in Intracerebral Hemorrhage: A Systematic Review and Meta-analysis. Transl. Stroke Res. 2024, 15, 533–544. [Google Scholar] [CrossRef] [PubMed]
- Hainsworth, A.H.; Markus, H.S.; Schneider, J.A. Cerebral Small Vessel Disease, Hypertension, and Vascular Contributions to Cognitive Impairment and Dementia. Hypertension 2024, 81, 75–86. [Google Scholar] [CrossRef]
- Gorelick, P.B.; Sorond, F.A. Advancing our knowledge about cerebral small vessel diseases. Lancet Neurol. 2023, 22, 972–973. [Google Scholar] [CrossRef]
- Ren, B.; Tan, L.; Song, Y.; Li, D.; Xue, B.; Lai, X.; Gao, Y. Cerebral Small Vessel Disease: Neuroimaging Features, Biochemical Markers, Influencing Factors, Pathological Mechanism and Treatment. Front. Neurol. 2022, 13, 843953. [Google Scholar] [CrossRef]
- Hase, Y.; Polvikoski, T.M.; Firbank, M.J.; Craggs, L.J.L.; Hawthorne, E.; Platten, C.; Stevenson, W.; Deramecourt, V.; Ballard, C.; Kenny, R.A.; et al. Small vessel disease pathological changes in neurodegenerative and vascular dementias concomitant with autonomic dysfunction. Brain Pathol. 2020, 30, 191–202. [Google Scholar] [CrossRef]
- Low, A.; Mak, E.; Rowe, J.B.; Markus, H.S.; O’Brien, J.T. Inflammation and cerebral small vessel disease: A systematic review. Ageing Res. Rev. 2019, 53, 100916. [Google Scholar] [CrossRef]
- Craggs, L.J.; Yamamoto, Y.; Deramecourt, V.; Kalaria, R.N. Microvascular pathology and morphometrics of sporadic and hereditary small vessel diseases of the brain. Brain Pathol. 2014, 24, 495–509. [Google Scholar] [CrossRef]
- Gouw, A.A.; Seewann, A.; van der Flier, W.M.; Barkhof, F.; Rozemuller, A.M.; Scheltens, P.; Geurts, J.J. Heterogeneity of small vessel disease: A systematic review of MRI and histopathology correlations. J. Neurol. Neurosurg. Psychiatry 2011, 82, 126–135. [Google Scholar] [CrossRef]
- Koemans, E.A.; Chhatwal, J.P.; van Veluw, S.J.; van Etten, E.S.; van Osch, M.J.P.; van Walderveen, M.A.A.; Sohrabi, H.R.; Kozberg, M.G.; Shirzadi, Z.; Terwindt, G.M.; et al. Progression of cerebral amyloid angiopathy: A pathophysiological framework. Lancet Neurol. 2023, 22, 632–642. [Google Scholar] [CrossRef]
- Jakel, L.; De Kort, A.M.; Klijn, C.J.M.; Schreuder, F.; Verbeek, M.M. Prevalence of cerebral amyloid angiopathy: A systematic review and meta-analysis. Alzheimer’s Dement. 2022, 18, 10–28. [Google Scholar] [CrossRef]
- Viswanathan, A.; Greenberg, S.M. Cerebral amyloid angiopathy in the elderly. Ann. Neurol. 2011, 70, 871–880. [Google Scholar] [CrossRef]
- Fakan, B.; Reisz, Z.; Zadori, D.; Vecsei, L.; Klivenyi, P.; Szalardy, L. Predictors of localization, outcome, and etiology of spontaneous intracerebral hemorrhages: Focus on cerebral amyloid angiopathy. J. Neural Transm. 2020, 127, 963–972. [Google Scholar] [CrossRef] [PubMed]
- Charidimou, A.; Boulouis, G.; Frosch, M.P.; Baron, J.C.; Pasi, M.; Albucher, J.F.; Banerjee, G.; Barbato, C.; Bonneville, F.; Brandner, S.; et al. The Boston criteria version 2.0 for cerebral amyloid angiopathy: A multicentre, retrospective, MRI-neuropathology diagnostic accuracy study. Lancet Neurol. 2022, 21, 714–725. [Google Scholar] [CrossRef] [PubMed]
- Szalardy, L.; Lee, S.; Kim, A.; Kovacs, G.G. Distinct cerebral amyloid angiopathy patterns in adult Down syndrome. J. Neurol. Sci. 2025, 476, 123601. [Google Scholar] [CrossRef] [PubMed]
- Gunda, B.; Böjti, P.; Kozák, L.R. Hyperacute Spontaneous Intracerebral Hemorrhage During Computed Tomography Scanning. JAMA Neurol. 2021, 78, 365–366. [Google Scholar] [CrossRef]
- Gunda, B.; Böjti, P.; Takács, T.; Zhubi, E.; Bereczki, D.; Varga, A.; Kozák, L.R. Spontaneous intracerebral hemorrhage during computed tomography scanning-assessment of hyperacute hematoma growth. Geroscience 2025. online ahead of print. [Google Scholar] [CrossRef]
- Jeong, D.; Jhaveri, M.D.; Prabhakaran, S. Magnetic resonance imaging characteristics at onset of spontaneous intracerebral hemorrhage. Arch. Neurol. 2011, 68, 826–827. [Google Scholar] [CrossRef]
- Liu, J.; Zhu, Y.S.; Hill, C.; Armstrong, K.; Tarumi, T.; Hodics, T.; Hynan, L.S.; Zhang, R. Cerebral autoregulation of blood velocity and volumetric flow during steady-state changes in arterial pressure. Hypertension 2013, 62, 973–979. [Google Scholar] [CrossRef]
- Harder, D.R.; Narayanan, J.; Gebremedhin, D. Pressure-induced myogenic tone and role of 20-HETE in mediating autoregulation of cerebral blood flow. Am. J. Physiol. Heart Circ. Physiol. 2011, 300, H1557–H1565. [Google Scholar] [CrossRef]
- Traon, A.P.; Costes-Salon, M.C.; Galinier, M.; Fourcade, J.; Larrue, V. Dynamics of cerebral blood flow autoregulation in hypertensive patients. J. Neurol. Sci. 2002, 195, 139–144. [Google Scholar] [CrossRef]
- Willie, C.K.; Tzeng, Y.C.; Fisher, J.A.; Ainslie, P.N. Integrative regulation of human brain blood flow. J. Physiol. 2014, 592, 841–859. [Google Scholar] [CrossRef] [PubMed]
- Young, A.P.; Zhu, J.; Bagher, A.M.; Denovan-Wright, E.M.; Howlett, S.E.; Kelly, M.E.M. Endothelin B receptor dysfunction mediates elevated myogenic tone in cerebral arteries from aged male Fischer 344 rats. Geroscience 2021, 43, 1447–1463. [Google Scholar] [CrossRef] [PubMed]
- Adamek, S.; Vyskocil, F. Potassium-selective microelectrode revealed difference in threshold potassium concentration for cortical spreading depression in female and male rat brain. Brain Res. 2011, 1370, 215–219. [Google Scholar] [CrossRef] [PubMed]
- Silverman, A.; Petersen, N.H. Physiology, Cerebral Autoregulation. In StatPearls; StatPearls Publishing LLC.: Treasure Island, FL, USA, 2025. [Google Scholar]
- Tan, C.O.; Hamner, J.W.; Taylor, J.A. The role of myogenic mechanisms in human cerebrovascular regulation. J. Physiol. 2013, 591, 5095–5105. [Google Scholar] [CrossRef]
- Toth, P.; Csiszar, A.; Tucsek, Z.; Sosnowska, D.; Gautam, T.; Koller, A.; Laniado Schwartzman, M.; Sonntag, W.E.; Ungvari, Z.I. Role of 20-HETE, TRP channels & BKCa in dysregulation of pressure-induced Ca2+ signaling and myogenic constriction of cerebral arteries in aged hypertensive mice. Am. J. Physiol. Heart Circ. Physiol. 2013, 305, H1698–H1708. [Google Scholar] [CrossRef]
- Toth, P.; Tucsek, Z.; Sosnowska, D.; Gautam, T.; Mitschelen, M.; Tarantini, S.; Deak, F.; Koller, A.; Sonntag, W.E.; Csiszar, A.; et al. Age-related autoregulatory dysfunction and cerebromicrovascular injury in mice with angiotensin II-induced hypertension. J. Cereb. Blood Flow. Metab. 2013, 33, 1732–1742. [Google Scholar] [CrossRef]
- Toth, P.; Tucsek, Z.; Tarantini, S.; Sosnowska, D.; Gautam, T.; Mitschelen, M.; Koller, A.; Sonntag, W.E.; Csiszar, A.; Ungvari, Z. IGF-1 deficiency impairs cerebral myogenic autoregulation in hypertensive mice. J. Cereb. Blood Flow. Metab. 2014, 34, 1887–1897. [Google Scholar] [CrossRef]
- Springo, Z.; Toth, P.; Tarantini, S.; Ashpole, N.M.; Tucsek, Z.; Sonntag, W.E.; Csiszar, A.; Koller, A.; Ungvari, Z.I. Aging impairs myogenic adaptation to pulsatile pressure in mouse cerebral arteries. J. Cereb. Blood Flow. Metab. 2015, 35, 527–530. [Google Scholar] [CrossRef]
- Roman, R.J.; Van Dokkum, R.P. Commentary on the special issue on the impact of myogenic tone in health and disease. Curr. Vasc. Pharmacol. 2014, 12, 779. [Google Scholar] [CrossRef]
- Pires, P.W.; Dams Ramos, C.M.; Matin, N.; Dorrance, A.M. The effects of hypertension on the cerebral circulation. Am. J. Physiol. Heart Circ. Physiol. 2013, 304, H1598–H1614. [Google Scholar] [CrossRef]
- Toth, P.; Tarantini, S.; Csiszar, A.; Ungvari, Z. Functional vascular contributions to cognitive impairment and dementia: Mechanisms and consequences of cerebral autoregulatory dysfunction, endothelial impairment, and neurovascular uncoupling in aging. Am. J. Physiol. Heart Circ. Physiol. 2017, 312, H1–H20. [Google Scholar] [CrossRef] [PubMed]
- Sadoshima, S.; Yoshida, F.; Ibayashi, S.; Shiokawa, O.; Fujishima, M. Upper limit of cerebral autoregulation during development of hypertension in spontaneously hypertensive rats--effect of sympathetic denervation. Stroke 1985, 16, 477–481. [Google Scholar] [CrossRef] [PubMed]
- Barry, D.I.; Lassen, N.A. Cerebral blood flow autoregulation in hypertension and effects of antihypertensive drugs. J. Hypertens. Suppl. 1984, 2, S519–S526. [Google Scholar] [PubMed]
- Barry, D.I.; Strandgaard, S.; Graham, D.I.; Braendstrup, O.; Svendsen, U.G.; Vorstrup, S.; Hemmingsen, R.; Bolwig, T.G. Cerebral blood flow in rats with renal and spontaneous hypertension: Resetting of the lower limit of autoregulation. J. Cereb. Blood Flow. Metab. 1982, 2, 347–353. [Google Scholar] [CrossRef] [PubMed]
- Humphrey, J.D. Mechanisms of Vascular Remodeling in Hypertension. Am. J. Hypertens. 2021, 34, 432–441. [Google Scholar] [CrossRef]
- Zeng, X.; Yang, Y. Molecular Mechanisms Underlying Vascular Remodeling in Hypertension. Rev. Cardiovasc. Med. 2024, 25, 72. [Google Scholar] [CrossRef]
- Ungvari, Z.; Muranyi, M.; Gulej, R.; Negri, S.; Nyul-Toth, A.; Csik, B.; Patai, R.; Conley, S.; Milan, M.; Bagwell, J.; et al. Longitudinal detection of gait alterations associated with hypertension-induced cerebral microhemorrhages in mice: Predictive role of stride length and stride time asymmetry and increased gait entropy. Geroscience 2024, 46, 4743–4760. [Google Scholar] [CrossRef]
- Toth, P.; Szarka, N.; Farkas, E.; Ezer, E.; Czeiter, E.; Amrein, K.; Ungvari, Z.; Hartings, J.A.; Buki, A.; Koller, A. Traumatic brain injury-induced autoregulatory dysfunction and spreading depression-related neurovascular uncoupling: Pathomechanisms, perspectives, and therapeutic implications. Am. J. Physiol. Heart Circ. Physiol. 2016, 311, H1118–H1131. [Google Scholar] [CrossRef]
- Gatti, L.; Tinelli, F.; Scelzo, E.; Arioli, F.; Di Fede, G.; Obici, L.; Pantoni, L.; Giaccone, G.; Caroppo, P.; Parati, E.A.; et al. Understanding the Pathophysiology of Cerebral Amyloid Angiopathy. Int. J. Mol. Sci. 2020, 21, 3435. [Google Scholar] [CrossRef]
- Martin-Jimenez, P.; Sanchez-Tornero, M.; Llamas-Velasco, S.; Guerrero-Molina, M.P.; Gonzalez-Sanchez, M.; Herrero-San Martin, A.; Blanco-Palmero, V.; Calleja-Castano, P.; Francisco-Gonzalo, J.; Hilario, A.; et al. Cerebral amyloid angiopathy-related inflammation: Clinical features and treatment response in a case series. Neurologia 2023, 38, 550–559. [Google Scholar] [CrossRef]
- Norling, A.M.; Gerstenecker, A.T.; Buford, T.W.; Khan, B.; Oparil, S.; Lazar, R.M. The role of exercise in the reversal of IGF-1 deficiencies in microvascular rarefaction and hypertension. Geroscience 2020, 42, 141–158. [Google Scholar] [CrossRef] [PubMed]
- Sleight, E.; Stringer, M.S.; Clancy, U.; Arteaga, C.; Jaime Garcia, D.; Hewins, W.; Jochems, A.C.C.; Hamilton, O.K.L.; Manning, C.; Morgan, A.G.; et al. Cerebrovascular Reactivity in Patients With Small Vessel Disease: A Cross-Sectional Study. Stroke 2023, 54, 2776–2784. [Google Scholar] [CrossRef] [PubMed]
- Liu, Z.; Ma, H.; Guo, Z.N.; Wang, L.; Qu, Y.; Fan, L.; Liu, X.; Liu, J.; Zhu, Y.; Yang, Y. Impaired dynamic cerebral autoregulation is associated with the severity of neuroimaging features of cerebral small vessel disease. CNS Neurosci. Ther. 2022, 28, 298–306. [Google Scholar] [CrossRef] [PubMed]
- Toth, L.; Czigler, A.; Hegedus, E.; Komaromy, H.; Amrein, K.; Czeiter, E.; Yabluchanskiy, A.; Koller, A.; Orsi, G.; Perlaki, G.; et al. Age-related decline in circulating IGF-1 associates with impaired neurovascular coupling responses in older adults. Geroscience 2022, 44, 2771–2783. [Google Scholar] [CrossRef]
- Venkat, P.; Chopp, M.; Chen, J. New insights into coupling and uncoupling of cerebral blood flow and metabolism in the brain. Croat. Med. J. 2016, 57, 223–228. [Google Scholar] [CrossRef]
- Dani, M.; Dirksen, A.; Taraborrelli, P.; Panagopolous, D.; Torocastro, M.; Sutton, R.; Lim, P.B. Orthostatic hypotension in older people: Considerations, diagnosis and management. Clin. Med. 2021, 21, e275–e282. [Google Scholar] [CrossRef]
- Dupre, N.; Drieu, A.; Joutel, A. Pathophysiology of cerebral small vessel disease: A journey through recent discoveries. J. Clin. Investig. 2024, 134, e172841. [Google Scholar] [CrossRef]
- Andonian, B.J.; Hippensteel, J.A.; Abuabara, K.; Boyle, E.M.; Colbert, J.F.; Devinney, M.J.; Faye, A.S.; Kochar, B.; Lee, J.; Litke, R.; et al. Inflammation and aging-related disease: A transdisciplinary inflammaging framework. Geroscience 2024, 47, 515–542. [Google Scholar] [CrossRef]
- Meier, H.C.S.; Mitchell, C.; Karadimas, T.; Faul, J.D. Systemic inflammation and biological aging in the Health and Retirement Study. Geroscience 2023, 45, 3257–3265. [Google Scholar] [CrossRef]
- Lu, W.H.; Guyonnet, S.; Martinez, L.O.; Lucas, A.; Parini, A.; Vellas, B.; de Souto Barreto, P. Association between aging-related biomarkers and longitudinal trajectories of intrinsic capacity in older adults. Geroscience 2023, 45, 3409–3418. [Google Scholar] [CrossRef] [PubMed]
- Bencivenga, L.; Strumia, M.; Rolland, Y.; Martinez, L.; Cestac, P.; Guyonnet, S.; Andrieu, S.; Parini, A.; Lucas, A.; Vellas, B.; et al. Biomarkers of mitochondrial dysfunction and inflammaging in older adults and blood pressure variability. Geroscience 2023, 45, 797–809. [Google Scholar] [CrossRef] [PubMed]
- Franceschi, C.; Campisi, J. Chronic inflammation (inflammaging) and its potential contribution to age-associated diseases. J. Gerontol. A Biol. Sci. Med. Sci. 2014, 69 (Suppl. 1), S4–S9. [Google Scholar] [CrossRef] [PubMed]
- Santoro, A.; Bientinesi, E.; Monti, D. Immunosenescence and inflammaging in the aging process: Age-related diseases or longevity? Ageing Res. Rev. 2021, 71, 101422. [Google Scholar] [CrossRef]
- Csiszar, A.; Gautam, T.; Sosnowska, D.; Tarantini, S.; Banki, E.; Tucsek, Z.; Toth, P.; Losonczy, G.; Koller, A.; Reglodi, D.; et al. Caloric restriction confers persistent anti-oxidative, pro-angiogenic, and anti-inflammatory effects and promotes anti-aging miRNA expression profile in cerebromicrovascular endothelial cells of aged rats. Am. J. Physiol. Heart Circ. Physiol. 2014, 307, H292–H306. [Google Scholar] [CrossRef]
- Csiszar, A.; Wang, M.; Lakatta, E.G.; Ungvari, Z.I. Inflammation and endothelial dysfunction during aging: Role of NF-κB. J. Appl. Physiol. 2008, 105, 1333–1341. [Google Scholar] [CrossRef]
- Ungvari, Z.I.; Orosz, Z.; Labinskyy, N.; Rivera, A.; Xiangmin, Z.; Smith, K.E.; Csiszar, A. Increased mitochondrial H2O2 production promotes endothelial NF-kB activation in aged rat arteries. Am. J. Physiol. Heart Circ. Physiol. 2007, 293, H37–H47. [Google Scholar] [CrossRef]
- Csik, B.; Nyul-Toth, A.; Gulej, R.; Patai, R.; Kiss, T.; Delfavero, J.; Nagaraja, R.Y.; Balasubramanian, P.; Shanmugarama, S.; Ungvari, A.; et al. Senescent Endothelial Cells in Cerebral Microcirculation Are Key Drivers of Age-Related Blood-Brain Barrier Disruption, Microvascular Rarefaction, and Neurovascular Coupling Impairment in Mice. Aging Cell 2025, 24, e70048. [Google Scholar] [CrossRef]
- Voigt, S.; Koemans, E.A.; Rasing, I.; van Etten, E.S.; Terwindt, G.M.; Baas, F.; Kaushik, K.; van Es, A.; van Buchem, M.A.; van Osch, M.J.P.; et al. Minocycline for sporadic and hereditary cerebral amyloid angiopathy (BATMAN): Study protocol for a placebo-controlled randomized double-blind trial. Trials 2023, 24, 378. [Google Scholar] [CrossRef]
- Koemans, E.A.; van Etten, E.S. Cerebral amyloid angiopathy: One single entity? Curr. Opin. Neurol. 2025, 38, 29–34. [Google Scholar] [CrossRef]
- Szalardy, L.; Fakan, B.; Maszlag-Torok, R.; Ferencz, E.; Reisz, Z.; Radics, B.L.; Csizmadia, S.; Szpisjak, L.; Annus, A.; Zadori, D.; et al. Identifying diagnostic and prognostic factors in cerebral amyloid angiopathy-related inflammation: A systematic analysis of published and seven new cases. Neuropathol. Appl. Neurobiol. 2024, 50, e12946. [Google Scholar] [CrossRef] [PubMed]
- Charidimou, A. Cerebral Amyloid Angiopathy-Related Inflammation Spectrum Disorders: Introduction of a Novel Concept and Diagnostic Criteria. Ann. Neurol. 2025, 97, 470–474. [Google Scholar] [CrossRef] [PubMed]
- Fandler-Hofler, S.; Obergottsberger, L.; Ambler, G.; Eppinger, S.; Wunsch, G.; Kneihsl, M.; Seiffge, D.; Banerjee, G.; Wilson, D.; Nash, P.; et al. Association of the Presence and Pattern of MRI Markers of Cerebral Small Vessel Disease With Recurrent Intracerebral Hemorrhage. Neurology 2023, 101, e794–e804. [Google Scholar] [CrossRef] [PubMed]
- Babarczy, K.; Radics, B.L.; Horvath, O.; Klivenyi, P.; Szalardy, L. Fatal Outcome of Intravenous Thrombolysis With an Unexpected Finding of Amyloid-beta-Related Angiitis-A Case Report Highlighting a Relevant Scenario With Acute Focal Neurological Deficits and Minimal Radiological Presentation. Neuropathology 2025, 45, e70013. [Google Scholar] [CrossRef]
- Reisz, Z.; Troakes, C.; Sztriha, L.K.; Bodi, I. Fatal thrombolysis-related intracerebral haemorrhage associated with amyloid-beta-related angiitis in a middle-aged patient—Case report and literature review. BMC Neurol. 2022, 22, 500. [Google Scholar] [CrossRef]
- Sellimi, A.; Panteleienko, L.; Mallon, D.; Fandler-Hofler, S.; Oliver, R.; Harvey, V.; Zandi, M.S.; Banerjee, G.; Werring, D.J. Inflammation in Cerebral Amyloid Angiopathy-Related Transient Focal Neurological Episodes. Ann. Neurol. 2025, 97, 475–482. [Google Scholar] [CrossRef]
- Sperling, R.A.; Jack, C.R., Jr.; Black, S.E.; Frosch, M.P.; Greenberg, S.M.; Hyman, B.T.; Scheltens, P.; Carrillo, M.C.; Thies, W.; Bednar, M.M.; et al. Amyloid-related imaging abnormalities in amyloid-modifying therapeutic trials: Recommendations from the Alzheimer’s Association Research Roundtable Workgroup. Alzheimers Dement. 2011, 7, 367–385. [Google Scholar] [CrossRef]
- Solopova, E.; Romero-Fernandez, W.; Harmsen, H.; Ventura-Antunes, L.; Wang, E.; Shostak, A.; Maldonado, J.; Donahue, M.J.; Schultz, D.; Coyne, T.M.; et al. Fatal iatrogenic cerebral β-amyloid-related arteritis in a woman treated with lecanemab for Alzheimer’s disease. Nat. Commun. 2023, 14, 8220. [Google Scholar] [CrossRef]
- Reish, N.J.; Jamshidi, P.; Stamm, B.; Flanagan, M.E.; Sugg, E.; Tang, M.; Donohue, K.L.; McCord, M.; Krumpelman, C.; Mesulam, M.M.; et al. Multiple Cerebral Hemorrhages in a Patient Receiving Lecanemab and Treated with t-PA for Stroke. N. Engl. J. Med. 2023, 388, 478–479. [Google Scholar] [CrossRef]
- Piazza, F.; Lucia, D.D.; Guzzi, F.; Pascarella, R.; DeBernardi, E.; Antolini, L.; Basso, G.; Zedde, M. Spontaneous and iatrogenic ARIA: Mechanistic insights from CAA-related inflammation. Alzheimer’s Dement. 2024, 20, e085575. [Google Scholar] [CrossRef]
- Zimmer, J.A.; Ardayfio, P.; Wang, H.; Khanna, R.; Evans, C.D.; Lu, M.; Sparks, J.; Andersen, S.; Lauzon, S.; Nery, E.S.M.; et al. Amyloid-Related Imaging Abnormalities With Donanemab in Early Symptomatic Alzheimer Disease: Secondary Analysis of the TRAILBLAZER-ALZ and ALZ 2 Randomized Clinical Trials. JAMA Neurol. 2025, 82, 461–469. [Google Scholar] [CrossRef] [PubMed]
- Landsberger, T.; Amit, I.; Alon, U. Geroprotective interventions converge on gene expression programs of reduced inflammation and restored fatty acid metabolism. Geroscience 2024, 46, 1627–1639. [Google Scholar] [CrossRef] [PubMed]
- Yousefzadeh, M.J.; Wilkinson, J.E.; Hughes, B.; Gadela, N.; Ladiges, W.C.; Vo, N.; Niedernhofer, L.J.; Huffman, D.M.; Robbins, P.D. Heterochronic parabiosis regulates the extent of cellular senescence in multiple tissues. Geroscience 2020, 42, 951–961. [Google Scholar] [CrossRef] [PubMed]
- Gulej, R.; Patai, R.; Ungvari, A.; Kallai, A.; Tarantini, S.; Yabluchanskiy, A.; Huffman, D.M.; Conboy, M.J.; Conboy, I.M.; Kivimaki, M.; et al. Impacts of systemic milieu on cerebrovascular and brain aging: Insights from heterochronic parabiosis, blood exchange, and plasma transfer experiments. Geroscience 2025. online ahead of print. [Google Scholar] [CrossRef]
- Ximerakis, M.; Holton, K.M.; Giadone, R.M.; Ozek, C.; Saxena, M.; Santiago, S.; Adiconis, X.; Dionne, D.; Nguyen, L.; Shah, K.M.; et al. Heterochronic parabiosis reprograms the mouse brain transcriptome by shifting aging signatures in multiple cell types. Nat. Aging 2023, 3, 327–345. [Google Scholar] [CrossRef]
- Poudel, S.B.; Ruff, R.R.; He, Z.; Dixit, M.; Yildirim, G.; Jayarathne, H.; Manchanayake, D.H.; Basta-Pljakic, J.; Duran-Ortiz, S.; Schaffler, M.B.; et al. The impact of inactivation of the GH/IGF axis during aging on healthspan. Geroscience 2024, 47, 3027–3042. [Google Scholar] [CrossRef]
- Tarantini, S.; Nyul-Toth, A.; Yabluchanskiy, A.; Csipo, T.; Mukli, P.; Balasubramanian, P.; Ungvari, A.; Toth, P.; Benyo, Z.; Sonntag, W.E.; et al. Endothelial deficiency of insulin-like growth factor-1 receptor (IGF1R) impairs neurovascular coupling responses in mice, mimicking aspects of the brain aging phenotype. Geroscience 2021, 43, 2387–2394. [Google Scholar] [CrossRef]
- Tarantini, S.; Balasubramanian, P.; Yabluchanskiy, A.; Ashpole, N.M.; Logan, S.; Kiss, T.; Ungvari, A.; Nyul-Toth, A.; Schwartzman, M.L.; Benyo, Z.; et al. IGF1R signaling regulates astrocyte-mediated neurovascular coupling in mice: Implications for brain aging. Geroscience 2021, 43, 901–911. [Google Scholar] [CrossRef]
- Ungvari, Z.; Csiszar, A. The emerging role of IGF-1 deficiency in cardiovascular aging: Recent advances. J. Gerontol. A Biol. Sci. Med. Sci. 2012, 67, 599–610. [Google Scholar] [CrossRef]
- Gong, Z.; Kennedy, O.; Sun, H.; Wu, Y.; Williams, G.A.; Klein, L.; Cardoso, L.; Matheny, R.W., Jr.; Hubbard, G.B.; Ikeno, Y.; et al. Reductions in serum IGF-1 during aging impair health span. Aging Cell 2014, 13, 408–418. [Google Scholar] [CrossRef]
- Bailey-Downs, L.C.; Mitschelen, M.; Sosnowska, D.; Toth, P.; Pinto, J.T.; Ballabh, P.; Valcarcel-Ares, M.N.; Farley, J.; Koller, A.; Henthorn, J.C.; et al. Liver-specific knockdown of IGF-1 decreases vascular oxidative stress resistance by impairing the Nrf2-dependent antioxidant response: A novel model of vascular aging. J. Gerontol. A Biol. Sci. Med. Sci. 2012, 67, 313–329. [Google Scholar] [CrossRef]
- Ungvari, Z.; Sosnowska, D.; Podlutsky, A.; Koncz, P.; Sonntag, W.E.; Csiszar, A. Free radical production, antioxidant capacity, and oxidative stress response signatures in fibroblasts from Lewis dwarf rats: Effects of life span-extending peripubertal GH treatment. J. Gerontol. A Biol. Sci. Med. Sci. 2011, 66, 501–510. [Google Scholar] [CrossRef]
- Hao, C.N.; Geng, Y.J.; Li, F.; Yang, T.; Su, D.F.; Duan, J.L.; Li, Y. Insulin-like growth factor-1 receptor activation prevents hydrogen peroxide-induced oxidative stress, mitochondrial dysfunction and apoptosis. Apoptosis 2011, 16, 1118–1127. [Google Scholar] [CrossRef] [PubMed]
- Higashi, Y.; Sukhanov, S.; Anwar, A.; Shai, S.Y.; Delafontaine, P. IGF-1, oxidative stress and atheroprotection. Trends Endocrinol. Metab. 2010, 21, 245–254. [Google Scholar] [CrossRef] [PubMed]
- Sukhanov, S.; Higashi, Y.; Shai, S.Y.; Vaughn, C.; Mohler, J.; Li, Y.; Song, Y.H.; Titterington, J.; Delafontaine, P. IGF-1 reduces inflammatory responses, suppresses oxidative stress, and decreases atherosclerosis progression in ApoE-deficient mice. Arterioscler. Thromb. Vasc. Biol. 2007, 27, 2684–2690. [Google Scholar] [CrossRef] [PubMed]
- Obradovic, M.; Zafirovic, S.; Soskic, S.; Stanimirovic, J.; Trpkovic, A.; Jevremovic, D.; Isenovic, E.R. Effects of IGF-1 on the Cardiovascular System. Curr. Pharm. Des. 2019, 25, 3715–3725. [Google Scholar] [CrossRef]
- Higashi, Y.; Gautam, S.; Delafontaine, P.; Sukhanov, S. IGF-1 and cardiovascular disease. Growth Horm. IGF Res. 2019, 45, 6–16. [Google Scholar] [CrossRef]
- Higashi, Y.; Sukhanov, S.; Anwar, A.; Shai, S.Y.; Delafontaine, P. Aging, atherosclerosis, and IGF-1. J. Gerontol. A Biol. Sci. Med. Sci. 2012, 67, 626–639. [Google Scholar] [CrossRef]
- Macvanin, M.; Gluvic, Z.; Radovanovic, J.; Essack, M.; Gao, X.; Isenovic, E.R. New insights on the cardiovascular effects of IGF-1. Front. Endocrinol. 2023, 14, 1142644. [Google Scholar] [CrossRef]
- Gulej, R.; Csik, B.; Faakye, J.; Tarantini, S.; Shanmugarama, S.; Chandragiri, S.S.; Mukli, P.; Conley, S.; Csiszar, A.; Ungvari, Z.; et al. Endothelial deficiency of insulin-like growth factor-1 receptor leads to blood-brain barrier disruption and accelerated endothelial senescence in mice, mimicking aspects of the brain aging phenotype. Microcirculation 2024, 31, e12840. [Google Scholar] [CrossRef]
- Labandeira-Garcia, J.L.; Costa-Besada, M.A.; Labandeira, C.M.; Villar-Cheda, B.; Rodriguez-Perez, A.I. Insulin-Like Growth Factor-1 and Neuroinflammation. Front. Aging Neurosci. 2017, 9, 365. [Google Scholar] [CrossRef] [PubMed]
- Lin, S.; Zhang, Q.; Shao, X.; Zhang, T.; Xue, C.; Shi, S.; Zhao, D.; Lin, Y. IGF-1 promotes angiogenesis in endothelial cells/adipose-derived stem cells co-culture system with activation of PI3K/Akt signal pathway. Cell Prolif. 2017, 50, e12390. [Google Scholar] [CrossRef] [PubMed]
- Tarantini, S.; Tucsek, Z.; Valcarcel-Ares, M.N.; Toth, P.; Gautam, T.; Giles, C.B.; Ballabh, P.; Wei, J.Y.; Wren, J.D.; Ashpole, N.M.; et al. Circulating IGF-1 deficiency exacerbates hypertension-induced microvascular rarefaction in the mouse hippocampus and retrosplenial cortex: Implications for cerebromicrovascular and brain aging. Age 2016, 38, 273–289. [Google Scholar] [CrossRef]
- Humpert, P.M.; Djuric, Z.; Zeuge, U.; Oikonomou, D.; Seregin, Y.; Laine, K.; Eckstein, V.; Nawroth, P.P.; Bierhaus, A. Insulin stimulates the clonogenic potential of angiogenic endothelial progenitor cells by IGF-1 receptor-dependent signaling. Mol. Med. 2008, 14, 301–308. [Google Scholar] [CrossRef] [PubMed]
- Delafontaine, P.; Song, Y.H.; Li, Y. Expression, regulation, and function of IGF-1, IGF-1R, and IGF-1 binding proteins in blood vessels. Arterioscler. Thromb. Vasc. Biol. 2004, 24, 435–444. [Google Scholar] [CrossRef]
- Shigematsu, S.; Yamauchi, K.; Nakajima, K.; Iijima, S.; Aizawa, T.; Hashizume, K. IGF-1 regulates migration and angiogenesis of human endothelial cells. Endocr. J. 1999, 46, S59–S62. [Google Scholar] [CrossRef]
- Higashi, Y.; Quevedo, H.C.; Tiwari, S.; Sukhanov, S.; Shai, S.Y.; Anwar, A.; Delafontaine, P. Interaction between insulin-like growth factor-1 and atherosclerosis and vascular aging. Front. Horm. Res. 2014, 43, 107–124. [Google Scholar] [CrossRef]
- Bickel, M.A.; Csik, B.; Gulej, R.; Ungvari, A.; Nyul-Toth, A.; Conley, S.M. Cell non-autonomous regulation of cerebrovascular aging processes by the somatotropic axis. Front. Endocrinol. 2023, 14, 1087053. [Google Scholar] [CrossRef]
- Miller, L.R.; Bickel, M.A.; Tarantini, S.; Runion, M.E.; Matacchiera, Z.; Vance, M.L.; Hibbs, C.; Vaden, H.; Nagykaldi, D.; Martin, T.; et al. IGF1R deficiency in vascular smooth muscle cells impairs myogenic autoregulation and cognition in mice. Front. Aging Neurosci. 2024, 16, 1320808. [Google Scholar] [CrossRef]
- Toth, P.; Tarantini, S.; Ashpole, N.M.; Tucsek, Z.; Milne, G.L.; Valcarcel-Ares, N.M.; Menyhart, A.; Farkas, E.; Sonntag, W.E.; Csiszar, A.; et al. IGF-1 deficiency impairs neurovascular coupling in mice: Implications for cerebromicrovascular aging. Aging Cell 2015, 14, 1034–1044. [Google Scholar] [CrossRef] [PubMed]
- Fulop, G.A.; Ramirez-Perez, F.I.; Kiss, T.; Tarantini, S.; Valcarcel Ares, M.N.; Toth, P.; Yabluchanskiy, A.; Conley, S.M.; Ballabh, P.; Martinez-Lemus, L.A.; et al. IGF-1 deficiency Promotes Pathological Remodeling of Cerebral Arteries: A Potential Mechanism Contributing to the Pathogenesis of Intracerebral Hemorrhages in Aging. J. Gerontol. A Biol. Sci. Med. Sci. 2018, 74, 446–454. [Google Scholar] [CrossRef] [PubMed]
- Sonntag, W.E.; Deak, F.; Ashpole, N.; Toth, P.; Csiszar, A.; Freeman, W.; Ungvari, Z. Insulin-like growth factor-1 in CNS and cerebrovascular aging. Front. Aging Neurosci. 2013, 5, 27. [Google Scholar] [CrossRef] [PubMed]
- Batista, A.; Osorio, R.; Varela, A.; Guilherme, P.; Marreiros, A.; Pais, S.; Nzwalo, H. Prediction of short-term prognosis in elderly patients with spontaneous intracerebral hemorrhage. Eur. Geriatr. Med. 2021, 12, 1267–1273. [Google Scholar] [CrossRef]
- Wen, D.; Chen, R.; Zhang, T.; Li, H.; Zheng, J.; Fu, W.; You, C.; Ma, L. “Atypical” Mild Clinical Presentation in Elderly Patients With Ruptured Intracranial Aneurysm: Causes and Clinical Characteristics. Front. Surg. 2022, 9, 927351. [Google Scholar] [CrossRef]
- Huang, B.; Chen, A.; Sun, Y.; He, Q. The Role of Aging in Intracerebral Hemorrhage. Brain Sci. 2024, 14, 613. [Google Scholar] [CrossRef]
- Potter, T.; Lioutas, V.A.; Tano, M.; Pan, A.; Meeks, J.; Woo, D.; Seshadri, S.; Selim, M.; Vahidy, F. Cognitive Impairment After Intracerebral Hemorrhage: A Systematic Review of Current Evidence and Knowledge Gaps. Front. Neurol. 2021, 12, 716632. [Google Scholar] [CrossRef]
- Kuohn, L.R.; Witsch, J.; Steiner, T.; Sheth, K.N.; Kamel, H.; Navi, B.B.; Merkler, A.E.; Murthy, S.B.; Mayer, S.A. Early Deterioration, Hematoma Expansion, and Outcomes in Deep Versus Lobar Intracerebral Hemorrhage: The FAST Trial. Stroke 2022, 53, 2441–2448. [Google Scholar] [CrossRef]
- Jolink, W.M.T.; Wiegertjes, K.; Rinkel, G.J.E.; Algra, A.; de Leeuw, F.E.; Klijn, C.J.M. Location-specific risk factors for intracerebral hemorrhage: Systematic review and meta-analysis. Neurology 2020, 95, e1807–e1818. [Google Scholar] [CrossRef]
- Morotti, A.; Li, Q.; Nawabi, J.; Mazzacane, F.; Schlunk, F.; Shoamanesh, A.; Busto, G.; Cavallini, A.; Palmerini, F.; Paciaroni, M.; et al. Volume Tolerance and Prognostic Impact of Hematoma Expansion in Deep and Lobar Intracerebral Hemorrhage. Stroke 2025, 56, 1224–1231. [Google Scholar] [CrossRef]
- Cordonnier, C.; Klijn, C.; Smith, E.E.; Al-Shahi Salman, R.; Chwalisz, B.K.; van Etten, E.; Muir, R.T.; Piazza, F.; Schreiber, S.; Schreuder, F.H.; et al. Diagnosis and management of cerebral amyloid angiopathy: A scientific statement from the International CAA Association and the World Stroke Organization. Int. J. Stroke 2025, 20, 949–967. [Google Scholar] [CrossRef]
- McCarter, S.J.; Lesnick, T.G.; Lowe, V.; Mielke, M.M.; Constantopoulos, E.; Rabinstein, A.A.; Przybelski, S.A.; Botha, H.; Jones, D.T.; Ramanan, V.K.; et al. Cerebral Amyloid Angiopathy Pathology and Its Association With Amyloid-beta PET Signal. Neurology 2021, 97, e1799–e1808. [Google Scholar] [CrossRef] [PubMed]
- Charidimou, A.; Linn, J.; Vernooij, M.W.; Opherk, C.; Akoudad, S.; Baron, J.C.; Greenberg, S.M.; Jäger, H.R.; Werring, D.J. Cortical superficial siderosis: Detection and clinical significance in cerebral amyloid angiopathy and related conditions. Brain 2015, 138, 2126–2139. [Google Scholar] [CrossRef] [PubMed]
- Charidimou, A.; Peeters, A.; Fox, Z.; Gregoire, S.M.; Vandermeeren, Y.; Laloux, P.; Jäger, H.R.; Baron, J.C.; Werring, D.J. Spectrum of transient focal neurological episodes in cerebral amyloid angiopathy: Multicentre magnetic resonance imaging cohort study and meta-analysis. Stroke 2012, 43, 2324–2330. [Google Scholar] [CrossRef] [PubMed]
- Charidimou, A.; Boulouis, G.; Roongpiboonsopit, D.; Xiong, L.; Pasi, M.; Schwab, K.M.; Rosand, J.; Gurol, M.E.; Greenberg, S.M.; Viswanathan, A. Cortical superficial siderosis and recurrent intracerebral hemorrhage risk in cerebral amyloid angiopathy: Large prospective cohort and preliminary meta-analysis. Int. J. Stroke 2019, 14, 723–733. [Google Scholar] [CrossRef]
- Guo, X.; Zhong, R.; Han, Y.; Zhang, H.; Zhang, X.; Lin, W. Incidence and relevant factors for seizures after spontaneous intracerebral hemorrhage: A systematic review and meta-analysis. Seizure 2022, 101, 30–38. [Google Scholar] [CrossRef]
- Rabin, J.S.; Nichols, E.; La Joie, R.; Casaletto, K.B.; Palta, P.; Dams-O’Connor, K.; Kumar, R.G.; George, K.M.; Satizabal, C.L.; Schneider, J.A.; et al. Cerebral amyloid angiopathy interacts with neuritic amyloid plaques to promote tau and cognitive decline. Brain 2022, 145, 2823–2833. [Google Scholar] [CrossRef]
- Kanai, M.; Noguchi, M.; Kubo, H.; Nozoe, M.; Kitano, T.; Izawa, K.P.; Mase, K.; Shimada, S. Pre-Stroke Frailty and Stroke Severity in Elderly Patients with Acute Stroke. J. Stroke Cerebrovasc. Dis. 2020, 29, 105346. [Google Scholar] [CrossRef]
- Blaauw, J.; den Hertog, H.M.; van Zundert, J.M.; van der Gaag, N.A.; Jellema, K.; Dammers, R.; Kho, K.H.; Groen, R.J.M.; Lingsma, H.F.; van der Naalt, J.; et al. Transient neurological deficit in patients with chronic subdural hematoma: A retrospective cohort analysis. J. Neurol. 2022, 269, 3180–3188. [Google Scholar] [CrossRef]
- Chen, C.; Xie, Y.; Pu, M.; Deng, L.; Li, Z.; Yang, T.; Yin, H.; Zhang, Z.; Lv, X.; Liu, X.; et al. Age-related differences in risk factors, clinical characteristics, and outcomes for intracerebral hemorrhage. Front. Aging Neurosci. 2023, 15, 1264124. [Google Scholar] [CrossRef]
- Inoue, Y.; Miyashita, F.; Minematsu, K.; Toyoda, K. Clinical Characteristics and Outcomes of Intracerebral Hemorrhage in Very Elderly. J. Stroke Cerebrovasc. Dis. 2018, 27, 97–102. [Google Scholar] [CrossRef] [PubMed]
- Kim, M.G.; Gandhi, C.; Azizkhanian, I.; Epstein, B.; Mittal, A.; Lee, N.; Santarelli, J.; Schmidt, M.; Al-Mufti, F.; Bowers, C.A. Frailty and spontaneous intracerebral hemorrhage: Does the modified frailty index predict mortality? Clin. Neurol. Neurosurg. 2020, 194, 105816. [Google Scholar] [CrossRef] [PubMed]
- Morotti, A.; Nawabi, J.; Pilotto, A.; Toffali, M.; Busto, G.; Mazzacane, F.; Cavallini, A.; Laudisi, M.; Gentile, L.; Viola, M.M.; et al. Functional outcome improvement from 3 to 12 months after intracerebral hemorrhage. Eur. Stroke J. 2024, 9, 391–397. [Google Scholar] [CrossRef] [PubMed]
- Ong, S.H.; Tan, A.Y.X.; Tan, B.; Yeo, L.; Tan, L.F.; Teo, K.; Yeo, T.T.; Nga, V.D.W.; Lim, M.J.R. The effect of frailty on mortality and functional outcomes in spontaneous intracerebral haemorrhage. Clin. Neurol. Neurosurg. 2024, 246, 108539. [Google Scholar] [CrossRef]
- Mai, L.M.; Joundi, R.A.; Katsanos, A.H.; Selim, M.; Shoamanesh, A. Pathophysiology of Intracerebral Hemorrhage: Recovery Trajectories. Stroke 2025, 56, 783–793. [Google Scholar] [CrossRef]
- Mayerhofer, E.; Parodi, L.; Prapiadou, S.; Malik, R.; Rosand, J.; Georgakis, M.K.; Anderson, C.D. Genetic Risk Score Improves Risk Stratification for Anticoagulation-Related Intracerebral Hemorrhage. Stroke 2023, 54, 791–799. [Google Scholar] [CrossRef]
- Goeldlin, M.B.; Fandler-Hofler, S.; Pezzini, A.; Manikantan, A.; Rauch, J.; Hald, S.M.; Kristensen, M.L.; Obergottsberger, L.; Sembill, J.A.; Haupenthal, D.; et al. Location and Timing of Recurrent, Nontraumatic Intracerebral Hemorrhage. JAMA Neurol. 2025, 82, 355–363. [Google Scholar] [CrossRef]
- Ohashi, S.N.; DeLong, J.H.; Kozberg, M.G.; Mazur-Hart, D.J.; van Veluw, S.J.; Alkayed, N.J.; Sansing, L.H. Role of Inflammatory Processes in Hemorrhagic Stroke. Stroke 2023, 54, 605–619. [Google Scholar] [CrossRef]
- Morotti, A.; Boulouis, G.; Dowlatshahi, D.; Li, Q.; Shamy, M.; Al-Shahi Salman, R.; Rosand, J.; Cordonnier, C.; Goldstein, J.N.; Charidimou, A. Intracerebral haemorrhage expansion: Definitions, predictors, and prevention. Lancet Neurol. 2023, 22, 159–171. [Google Scholar] [CrossRef]
- Teo, K.C.; Fong, S.M.; Leung, W.C.Y.; Leung, I.Y.H.; Wong, Y.K.; Choi, O.M.Y.; Yam, K.K.; Lo, R.C.N.; Cheung, R.T.F.; Ho, S.L.; et al. Location-Specific Hematoma Volume Cutoff and Clinical Outcomes in Intracerebral Hemorrhage. Stroke 2023, 54, 1548–1557. [Google Scholar] [CrossRef]
- Patel, K.; Shrier, W.E.J.; Sengupta, N.; Hunt, D.C.E.; Hodgson, L.E. Frailty, Assessed by the Rockwood Clinical Frailty Scale and 1-Year Outcomes Following Ischaemic Stroke in a Non-Specialist UK Stroke Centre. J. Stroke Cerebrovasc. Dis. 2022, 31, 106451. [Google Scholar] [CrossRef]
- Parry-Jones, A.R.; Jarhult, S.J.; Kreitzer, N.; Morotti, A.; Toni, D.; Seiffge, D.; Mendelow, A.D.; Patel, H.; Brouwers, H.B.; Klijn, C.J.; et al. Acute care bundles should be used for patients with intracerebral haemorrhage: An expert consensus statement. Eur. Stroke J. 2024, 9, 295–302. [Google Scholar] [CrossRef] [PubMed]
- Li, Q.; Morotti, A.; Warren, A.; Qureshi, A.I.; Dowlatshahi, D.; Falcone, G.; Sheth, K.N.; Shoamanesh, A.; Murthy, S.B.; Viswanathan, A.; et al. Intensive Blood Pressure Reduction is Associated with Reduced Hematoma Growth in Fast Bleeding Intracerebral Hemorrhage. Ann. Neurol. 2025. online ahead of print. [Google Scholar] [CrossRef] [PubMed]
- Morotti, A.; Boulouis, G.; Nawabi, J.; Li, Q.; Charidimou, A.; Pasi, M.; Schlunk, F.; Shoamanesh, A.; Katsanos, A.H.; Mazzacane, F.; et al. Association Between Hematoma Expansion Severity and Outcome and Its Interaction With Baseline Intracerebral Hemorrhage Volume. Neurology 2023, 101, e1606–e1613. [Google Scholar] [CrossRef] [PubMed]
- Parry-Jones, A.R.; Sammut-Powell, C.; Paroutoglou, K.; Birleson, E.; Rowland, J.; Lee, S.; Cecchini, L.; Massyn, M.; Emsley, R.; Bray, B.; et al. An Intracerebral Hemorrhage Care Bundle Is Associated with Lower Case Fatality. Ann. Neurol. 2019, 86, 495–503. [Google Scholar] [CrossRef]
- Li, Q.; Yakhkind, A.; Alexandrov, A.W.; Alexandrov, A.V.; Anderson, C.S.; Dowlatshahi, D.; Frontera, J.A.; Hemphill, J.C.; Ganti, L.; Kellner, C.; et al. Code ICH: A Call to Action. Stroke 2024, 55, 494–505. [Google Scholar] [CrossRef]
- Ma, L.; Hu, X.; Song, L.; Chen, X.; Ouyang, M.; Billot, L.; Li, Q.; Malavera, A.; Li, X.; Munoz-Venturelli, P.; et al. The third Intensive Care Bundle with Blood Pressure Reduction in Acute Cerebral Haemorrhage Trial (INTERACT3): An international, stepped wedge cluster randomised controlled trial. Lancet 2023, 402, 27–40. [Google Scholar] [CrossRef]
- Anderson, C.S.; Heeley, E.; Huang, Y.; Wang, J.; Stapf, C.; Delcourt, C.; Lindley, R.; Robinson, T.; Lavados, P.; Neal, B.; et al. Rapid blood-pressure lowering in patients with acute intracerebral hemorrhage. N. Engl. J. Med. 2013, 368, 2355–2365. [Google Scholar] [CrossRef]
- Steiner, T.; Purrucker, J.C.; Aguiar de Sousa, D.; Apostolaki-Hansson, T.; Beck, J.; Christensen, H.; Cordonnier, C.; Downer, M.B.; Eilertsen, H.; Gartly, R.; et al. European Stroke Organisation (ESO) and European Association of Neurosurgical Societies (EANS) guideline on stroke due to spontaneous intracerebral haemorrhage. Eur. Stroke J. 2025. online ahead of print. [Google Scholar] [CrossRef]
- Parry-Jones, A.R.; Moullaali, T.J.; Ziai, W.C. Treatment of intracerebral hemorrhage: From specific interventions to bundles of care. Int. J. Stroke 2020, 15, 945–953. [Google Scholar] [CrossRef]
- Mutimer, C.A.; Yassi, N.; Wu, T.Y. Blood Pressure Management in Intracerebral Haemorrhage: When, how much, and for how long? Curr. Neurol. Neurosci. Rep. 2024, 24, 181–189. [Google Scholar] [CrossRef]
- Sheth, K.N.; Solomon, N.; Alhanti, B.; Messe, S.R.; Xian, Y.; Bhatt, D.L.; Hemphill, J.C.; Frontera, J.A.; Chang, R.C.; Danelich, I.M.; et al. Time to Anticoagulation Reversal and Outcomes After Intracerebral Hemorrhage. JAMA Neurol. 2024, 81, 363–372. [Google Scholar] [CrossRef]
- Greenberg, S.M.; Ziai, W.C.; Cordonnier, C.; Dowlatshahi, D.; Francis, B.; Goldstein, J.N.; Hemphill, J.C., 3rd; Johnson, R.; Keigher, K.M.; Mack, W.J.; et al. 2022 Guideline for the Management of Patients With Spontaneous Intracerebral Hemorrhage: A Guideline From the American Heart Association/American Stroke Association. Stroke 2022, 53, e282–e361. [Google Scholar] [CrossRef]
- Christensen, H.; Cordonnier, C.; Korv, J.; Lal, A.; Ovesen, C.; Purrucker, J.C.; Toni, D.; Steiner, T. European Stroke Organisation Guideline on Reversal of Oral Anticoagulants in Acute Intracerebral Haemorrhage. Eur. Stroke J. 2019, 4, 294–306. [Google Scholar] [CrossRef]
- Pollack, C.V., Jr.; Reilly, P.A.; van Ryn, J.; Eikelboom, J.W.; Glund, S.; Bernstein, R.A.; Dubiel, R.; Huisman, M.V.; Hylek, E.M.; Kam, C.W.; et al. Idarucizumab for Dabigatran Reversal—Full Cohort Analysis. N. Engl. J. Med. 2017, 377, 431–441. [Google Scholar] [CrossRef] [PubMed]
- Costa, O.S.; Connolly, S.J.; Sharma, M.; Beyer-Westendorf, J.; Christoph, M.J.; Lovelace, B.; Coleman, C.I. Andexanet alfa versus four-factor prothrombin complex concentrate for the reversal of apixaban- or rivaroxaban-associated intracranial hemorrhage: A propensity score-overlap weighted analysis. Crit. Care 2022, 26, 180. [Google Scholar] [CrossRef] [PubMed]
- Huttner, H.B.; Gerner, S.T.; Kuramatsu, J.B.; Connolly, S.J.; Beyer-Westendorf, J.; Demchuk, A.M.; Middeldorp, S.; Zotova, E.; Altevers, J.; Andersohn, F.; et al. Hematoma Expansion and Clinical Outcomes in Patients With Factor-Xa Inhibitor-Related Atraumatic Intracerebral Hemorrhage Treated Within the ANNEXA-4 Trial Versus Real-World Usual Care. Stroke 2022, 53, 532–543. [Google Scholar] [CrossRef] [PubMed]
- Christensen, H.; Casolla, B.; Frontera, J.A.; Grundtvig, J.; Nielsen, J.D.; Petersson, J.; Steiner, T. Principles of reversal of anticoagulation in patients with intracerebral hemorrhage related to oral anticoagulants. Eur. Stroke J. 2025, 10, 4–13. [Google Scholar] [CrossRef]
- Naidech, A.M.; Grotta, J.; Elm, J.; Janis, S.; Dowlatshahi, D.; Toyoda, K.; Steiner, T.; Mayer, S.A.; Khanolkar, P.; Denlinger, J.; et al. Recombinant factor VIIa for hemorrhagic stroke treatment at earliest possible time (FASTEST): Protocol for a phase III, double-blind, randomized, placebo-controlled trial. Int. J. Stroke 2022, 17, 806–809. [Google Scholar] [CrossRef]
- Meretoja, A.; Yassi, N.; Wu, T.Y.; Churilov, L.; Sibolt, G.; Jeng, J.S.; Kleinig, T.; Spratt, N.J.; Thijs, V.; Wijeratne, T.; et al. Tranexamic acid in patients with intracerebral haemorrhage (STOP-AUST): A multicentre, randomised, placebo-controlled, phase 2 trial. Lancet Neurol. 2020, 19, 980–987. [Google Scholar] [CrossRef]
- Sprigg, N.; Flaherty, K.; Appleton, J.P.; Al-Shahi Salman, R.; Bereczki, D.; Beridze, M.; Christensen, H.; Ciccone, A.; Collins, R.; Czlonkowska, A.; et al. Tranexamic acid for hyperacute primary IntraCerebral Haemorrhage (TICH-2): An international randomised, placebo-controlled, phase 3 superiority trial. Lancet 2018, 391, 2107–2115. [Google Scholar] [CrossRef] [PubMed]
- Sondag, L.; Schreuder, F.; Boogaarts, H.D.; Rovers, M.M.; Vandertop, W.P.; Dammers, R.; Klijn, C.J.M.; Dutch Ich Surgery Trial Study Group, p.o.t.C.c. Neurosurgical Intervention for Supratentorial Intracerebral Hemorrhage. Ann. Neurol. 2020, 88, 239–250. [Google Scholar] [CrossRef] [PubMed]
- Kellner, C.P.; Song, R.; Ali, M.; Nistal, D.A.; Samarage, M.; Dangayach, N.S.; Liang, J.; McNeill, I.; Zhang, X.; Bederson, J.B.; et al. Time to Evacuation and Functional Outcome After Minimally Invasive Endoscopic Intracerebral Hemorrhage Evacuation. Stroke 2021, 52, e536–e539. [Google Scholar] [CrossRef] [PubMed]
- Zhou, X.; Chen, J.; Li, Q.; Ren, G.; Yao, G.; Liu, M.; Dong, Q.; Guo, J.; Li, L.; Guo, J.; et al. Minimally invasive surgery for spontaneous supratentorial intracerebral hemorrhage: A meta-analysis of randomized controlled trials. Stroke 2012, 43, 2923–2930. [Google Scholar] [CrossRef]
- Middleton, S.; McElduff, P.; Ward, J.; Grimshaw, J.M.; Dale, S.; D’Este, C.; Drury, P.; Griffiths, R.; Cheung, N.W.; Quinn, C.; et al. Implementation of evidence-based treatment protocols to manage fever, hyperglycaemia, and swallowing dysfunction in acute stroke (QASC): A cluster randomised controlled trial. Lancet 2011, 378, 1699–1706. [Google Scholar] [CrossRef]
- Langhorne, P.; Ramachandra, S.; Stroke Unit Trialists, C. Organised inpatient (stroke unit) care for stroke: Network meta-analysis. Cochrane Database Syst. Rev. 2020, 4, CD000197. [Google Scholar] [CrossRef]
- Hickman, L.D.; Phillips, J.L.; Newton, P.J.; Halcomb, E.J.; Al Abed, N.; Davidson, P.M. Multidisciplinary team interventions to optimise health outcomes for older people in acute care settings: A systematic review. Arch. Gerontol. Geriatr. 2015, 61, 322–329. [Google Scholar] [CrossRef]
- Yen, H.C.; Jeng, J.S.; Chen, W.S.; Pan, G.S.; Chuang Pt Bs, W.Y.; Lee, Y.Y.; Teng, T. Early Mobilization of Mild-Moderate Intracerebral Hemorrhage Patients in a Stroke Center: A Randomized Controlled Trial. Neurorehabil Neural Repair. 2020, 34, 72–81. [Google Scholar] [CrossRef]
- De Kort, A.M.; Kuiperij, H.B.; Marques, T.M.; Jäkel, L.; van den Berg, E.; Kersten, I.; van Berckel-Smit, H.E.P.; Duering, M.; Stoops, E.; Abdo, W.F.; et al. Decreased Cerebrospinal Fluid Amyloid β 38, 40, 42, and 43 Levels in Sporadic and Hereditary Cerebral Amyloid Angiopathy. Ann. Neurol. 2023, 93, 1173–1186. [Google Scholar] [CrossRef]
- Charidimou, A.; Boulouis, G. Core CSF Biomarker Profile in Cerebral Amyloid Angiopathy: Updated Meta-Analysis. Neurology 2024, 103, e209795. [Google Scholar] [CrossRef]
- Theodorou, A.; Tsantzali, I.; Stefanou, M.I.; Sacco, S.; Katsanos, A.H.; Shoamanesh, A.; Karapanayiotides, T.; Koutroulou, I.; Stamati, P.; Werring, D.J.; et al. CSF and plasma biomarkers in cerebral amyloid angiopathy: A single-center study and a systematic review/meta-analysis. Eur. Stroke J. 2025, 10, 278–288. [Google Scholar] [CrossRef]
- Muir, R.T.; Stukas, S.; Cooper, J.G.; Beaudin, A.E.; McCreary, C.R.; Gee, M.; Nelles, K.; Nukala, N.; Valencia, J.; Kirmess, K.M.; et al. Plasma biomarkers distinguish Boston Criteria 2.0 cerebral amyloid angiopathy from healthy controls. Alzheimer’s Dement. 2025, 21, e70010. [Google Scholar] [CrossRef]
- Charidimou, A.; Farid, K.; Tsai, H.H.; Tsai, L.K.; Yen, R.F.; Baron, J.C. Amyloid-PET burden and regional distribution in cerebral amyloid angiopathy: A systematic review and meta-analysis of biomarker performance. J. Neurol. Neurosurg. Psychiatry 2018, 89, 410–417. [Google Scholar] [CrossRef] [PubMed]
- Tsai, H.H.; Liu, C.J.; Lee, B.C.; Chen, Y.F.; Yen, R.F.; Jeng, J.S.; Tsai, L.K. Cerebral tau pathology in cerebral amyloid angiopathy. Brain Commun. 2024, 6, fcae086. [Google Scholar] [CrossRef] [PubMed]
- Chen, J.; Xie, C.; Wu, R.; Zhang, D.; Wen, F.; Nie, H.; Xue, L.; Zha, Z.; Wang, J. Optimization and automation of the radiosynthesis of PET ligand targeting Aβ for imaging cerebral amyloid angiopathy. Appl. Radiat. Isot. 2025, 225, 112037. [Google Scholar] [CrossRef] [PubMed]
- Myserlis, E.P.; Mayerhofer, E.; Abramson, J.R.; Teo, K.C.; Montgomery, B.E.; Sugita, L.; Warren, A.D.; Goldstein, J.N.; Gurol, M.E.; Viswanathan, A.; et al. Lobar intracerebral hemorrhage and risk of subsequent uncontrolled blood pressure. Eur. Stroke J. 2022, 7, 280–288. [Google Scholar] [CrossRef]
- Fernando, S.M.; Qureshi, D.; Talarico, R.; Tanuseputro, P.; Dowlatshahi, D.; Sood, M.M.; Smith, E.E.; Hill, M.D.; McCredie, V.A.; Scales, D.C.; et al. Intracerebral Hemorrhage Incidence, Mortality, and Association With Oral Anticoagulation Use: A Population Study. Stroke 2021, 52, 1673–1681. [Google Scholar] [CrossRef]
- O’Donnell, M.J.; Xavier, D.; Liu, L.; Zhang, H.; Chin, S.L.; Rao-Melacini, P.; Rangarajan, S.; Islam, S.; Pais, P.; McQueen, M.J.; et al. Risk factors for ischaemic and intracerebral haemorrhagic stroke in 22 countries (the INTERSTROKE study): A case-control study. Lancet 2010, 376, 112–123. [Google Scholar] [CrossRef]
- Biffi, A.; Anderson, C.D.; Jagiella, J.M.; Schmidt, H.; Kissela, B.; Hansen, B.M.; Jimenez-Conde, J.; Pires, C.R.; Ayres, A.M.; Schwab, K.; et al. APOE genotype and extent of bleeding and outcome in lobar intracerebral haemorrhage: A genetic association study. Lancet Neurol. 2011, 10, 702–709. [Google Scholar] [CrossRef]
- Collaboration, R. Effects of antiplatelet therapy after stroke due to intracerebral haemorrhage (RESTART): A randomised, open-label trial. Lancet 2019, 393, 2613–2623. [Google Scholar] [CrossRef]
- Cordonnier, C.; Demchuk, A.; Ziai, W.; Anderson, C.S. Intracerebral haemorrhage: Current approaches to acute management. Lancet 2018, 392, 1257–1268. [Google Scholar] [CrossRef]
- Frontera, J.A.; Lewin, J.J., 3rd; Rabinstein, A.A.; Aisiku, I.P.; Alexandrov, A.W.; Cook, A.M.; del Zoppo, G.J.; Kumar, M.A.; Peerschke, E.I.; Stiefel, M.F.; et al. Guideline for Reversal of Antithrombotics in Intracranial Hemorrhage: A Statement for Healthcare Professionals from the Neurocritical Care Society and Society of Critical Care Medicine. Neurocrit. Care 2016, 24, 6–46. [Google Scholar] [CrossRef]
- Mavridis, T.; Archontakis-Barakakis, P.; Chlorogiannis, D.D.; Charidimou, A. Left atrial appendage occlusion in patients with atrial fibrillation and previous Intracranial Hemorrhage or Cerebral Amyloid Angiopathy: A systematic review and meta-analysis. Int. J. Stroke 2025. online ahead of print. [Google Scholar] [CrossRef]
- Diener, H.C.; Hankey, G.J. Primary and Secondary Prevention of Ischemic Stroke and Cerebral Hemorrhage: JACC Focus Seminar. J. Am. Coll. Cardiol. 2020, 75, 1804–1818. [Google Scholar] [CrossRef] [PubMed]
- Charidimou, A.; Imaizumi, T.; Moulin, S.; Biffi, A.; Samarasekera, N.; Yakushiji, Y.; Peeters, A.; Vandermeeren, Y.; Laloux, P.; Baron, J.C.; et al. Brain hemorrhage recurrence, small vessel disease type, and cerebral microbleeds: A meta-analysis. Neurology 2017, 89, 820–829. [Google Scholar] [CrossRef] [PubMed]
- Hwang, B.Y.; Appelboom, G.; Ayer, A.; Kellner, C.P.; Kotchetkov, I.S.; Gigante, P.R.; Haque, R.; Kellner, M.; Connolly, E.S. Advances in neuroprotective strategies: Potential therapies for intracerebral hemorrhage. Cerebrovasc. Dis. 2011, 31, 211–222. [Google Scholar] [CrossRef]
- Tschoe, C.; Bushnell, C.D.; Duncan, P.W.; Alexander-Miller, M.A.; Wolfe, S.Q. Neuroinflammation after Intracerebral Hemorrhage and Potential Therapeutic Targets. J. Stroke 2020, 22, 29–46. [Google Scholar] [CrossRef] [PubMed]
- Ma, Q.; Chen, S.; Hu, Q.; Feng, H.; Zhang, J.H.; Tang, J. NLRP3 inflammasome contributes to inflammation after intracerebral hemorrhage. Ann. Neurol. 2014, 75, 209–219. [Google Scholar] [CrossRef]
- Fang, M.; Xia, F.; Wang, J.; Wang, C.; Teng, B.; You, S.; Li, M.; Chen, X.; Hu, X. The NLRP3 inhibitor, OLT1177 attenuates brain injury in experimental intracerebral hemorrhage. Int. Immunopharmacol. 2024, 131, 111869. [Google Scholar] [CrossRef]
- Ren, H.; Han, R.; Chen, X.; Liu, X.; Wan, J.; Wang, L.; Yang, X.; Wang, J. Potential therapeutic targets for intracerebral hemorrhage-associated inflammation: An update. J. Cereb. Blood Flow. Metab. 2020, 40, 1752–1768. [Google Scholar] [CrossRef]
- Yabluchanskiy, A.; Tarantini, S.; Balasubramanian, P.; Kiss, T.; Csipo, T.; Fulop, G.A.; Lipecz, A.; Ahire, C.; DelFavero, J.; Nyul-Toth, A.; et al. Pharmacological or genetic depletion of senescent astrocytes prevents whole brain irradiation-induced impairment of neurovascular coupling responses protecting cognitive function in mice. Geroscience 2020, 42, 409–428. [Google Scholar] [CrossRef]
- Bax, F.; Warren, A.; Fouks, A.A.; van den Brink, H.; van Veluw, S.J.; Kozberg, M.G.; Greenberg, S.M. Minocycline in Severe Cerebral Amyloid Angiopathy: A Single-Center Cohort Study. J. Am. Heart Assoc. 2024, 13, e033464. [Google Scholar] [CrossRef]
- Mc Lernon, S.; Werring, D.; Terry, L. Clinicians’ Perceptions of the Appropriateness of Neurocritical Care for Patients with Spontaneous Intracerebral Hemorrhage (ICH): A Qualitative Study. Neurocrit. Care 2021, 35, 162–171. [Google Scholar] [CrossRef]
- Holloway, R.G.; Arnold, R.M.; Creutzfeldt, C.J.; Lewis, E.F.; Lutz, B.J.; McCann, R.M.; Rabinstein, A.A.; Saposnik, G.; Sheth, K.N.; Zahuranec, D.B.; et al. Palliative and end-of-life care in stroke: A statement for healthcare professionals from the American Heart Association/American Stroke Association. Stroke 2014, 45, 1887–1916. [Google Scholar] [CrossRef]
- Visvanathan, A.; Mead, G.; Dennis, M.; Whiteley, W.; Doubal, F.; Lawton, J. Maintaining hope after a disabling stroke: A longitudinal qualitative study of patients’ experiences, views, information needs and approaches towards making treatment decisions. PLoS ONE 2019, 14, e0222500. [Google Scholar] [CrossRef]
- Madhok, D.Y.; Vitt, J.R.; MacIsaac, D.; Hsia, R.Y.; Kim, A.S.; Hemphill, J.C. Early Do-Not-Resuscitate Orders and Outcome After Intracerebral Hemorrhage. Neurocrit. Care 2021, 34, 492–499. [Google Scholar] [CrossRef]
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. |
© 2025 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).