Abstract
Limbic-predominant age-related TDP-43 encephalopathy neuropathologic change (LATE-NC) is detectable at autopsy in more than one-third of people beyond age 85 years and is robustly associated with dementia independent of other pathologies. Although LATE-NC has a large impact on public health, there remain uncertainties about the underlying biologic mechanisms. Here, we review the literature from human studies that may shed light on pathogenetic mechanisms. It is increasingly clear that certain combinations of pathologic changes tend to coexist in aging brains. Although “pure” LATE-NC is not rare, LATE-NC often coexists in the same brains with Alzheimer disease neuropathologic change, brain arteriolosclerosis, hippocampal sclerosis of aging, and/or age-related tau astrogliopathy (ARTAG). The patterns of pathologic comorbidities provide circumstantial evidence of mechanistic interactions (“synergies”) between the pathologies, and also suggest common upstream influences. As to primary mediators of vulnerability to neuropathologic changes, genetics may play key roles. Genes associated with LATE-NC include TMEM106B, GRN, APOE, SORL1, ABCC9, and others. Although the anatomic distribution of TDP-43 pathology defines the condition, important cofactors for LATE-NC may include Tau pathology, endolysosomal pathways, and blood-brain barrier dysfunction. A review of the human phenomenology offers insights into disease-driving mechanisms, and may provide clues for diagnostic and therapeutic targets.
Keywords: Alzheimer disease neuropathologic change (ADNC), CTE, Carboxy-terminal fragment (CTF), Genome-wide association study (GWAS), Hippocampal sclerosis of aging (HS-A), Frontotemporal lobar degeneration (FTLD), Sulfonylurea receptor type 2 (SUR2)
INTRODUCTION
Limbic-predominant age-related TDP-43 encephalopathy neuropathologic change (LATE-NC) is associated with a clinical syndrome that mimics Alzheimer disease (AD)-type dementia (1). Since the term LATE-NC was introduced in 2019, researchers around the world have found evidence of the large public health impact of the condition. Approximately 1 in 3 individuals beyond age 85 have LATE-NC at autopsy (2). When antemortem cognitive status and other brain pathologies are factored in together, LATE-NC is independently associated with substantial cognitive impairment (3–8). Recent reviews address clinical features of LATE/LATE-NC (9, 10).
Despite progress in the field, uncertainties remain about the biologic mechanisms underlying LATE-NC (10). Homo sapiens appear to have unique susceptibility to LATE-NC, and no model system has been developed for specific experimental study of LATE-NC pathogenesis. Therefore, our understanding of LATE-NC is at least preliminarily dependent on piecing together the relevant human phenomenology. Here, we discuss some of the data that have been gathered related to copathologies and genetic risk factors.
Several underlying factors are important to consider although they are not focal points of the present review: (1) the possible influences of chronological aging, for example senescence; (2) the tendency, particularly in persons of advanced age, for brains to harbor multiple pathologies in the same individual; and (3) challenges inherent to understanding how different pathogenetic factors inter-relate, when each individual topic under discussion is an incompletely understood component of a fast-evolving scientific field. Unlike some other neurodegenerative conditions, LATE-NC is most prevalent among the “oldest-old,” and senescence entails many person-specific influences (1, 11–15). Characteristics of aging-related pathologies include both complexity in an individual and heterogeneity across human populations (16–19). The pathologies may affect one another if present in the same brain; as described below, the subcomponents in “mixed” pathologic subtypes may have characteristics that are subtly different from those of “pure” subtypes. The science cultural context is also relevant; research on dementia, as well as its terminology and classification, are in constant flux.
Despite these caveats, the available pathologic and genetic data may provide insights into potential diagnostic biomarkers and therapeutic targets. The number of identified neuropathologic entities associated with dementia is ever increasing although there are overlapping pathologic mechanisms. This prompts hope that (as in other disease paradigms), a future disease-altering remedy that helps for 1 condition may also help for another. This article is written with an overall goal of reviewing the literature of human studies to generate hypotheses about cause-effect relationships in LATE-NC and associated conditions.
LATE-NC AND COMORBIDITIES THAT ARE ENRICHED IN BRAINS WITH LATE-NC
Before discussing associations between LATE-NC and other pathologies and genetics, it is important to address the pathological phenotype itself. TDP-43 proteinopathy (mislocalized, misfolded TDP-43 protein; Fig. 1A, B) was first recognized as a pathological feature in diseases along what is now considered to be a pathological spectrum that includes amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration with TDP-43 inclusions (FTLD-TDP) (20, 21). However, in recent years, it is increasingly recognized that TDP-43 pathology is analogous to tauopathies (22–24). Like Tau pathology, TDP-43 pathology appears to be stimulated by many “upstream” factors, and both pathologies produce many additional “downstream” deleterious effects that include loss of normal functions, and gain of new toxic functions (24–26). Indeed, numerous (more than 20) different TDPopathies have now been identified (24). Recent studies have provided additional insights into the pathogenetic mechanisms of TDP-43 dysregulation, which include exon mis-splicing, aberrant polyadenylation, and various cell type-specific functional impairments (27–32).
Figure 1.
(A, B) Limbic-predominant age-related TDP-43 encephalopathy neuropathologic changes (LATE-NC) and (D) hippocampal sclerosis of aging (HS-A). (A, B) Sections stained for phosphorylated TDP-43 (pTDP-43) immunohistochemically. The immunostaining signal is brown, nuclei are counterstained blue with hematoxylin. Notable LATE-NC-related features include pTDP-43-immunoreactive neuronal cytoplasmic inclusion (NCI; blue arrow), neurites (yellow arrow), and neuronal intranuclear inclusion (red arrow). (B) Early NCIs (green arrow) with punctate staining as well as a more mature and homogeneously stained one (blue arrow). A capillary is indicated with pink arrowheads, with an associated pTDP-43-immunoreactive Lin body (orange arrow). (C, D) Hippocampal formations at low magnification, stained with H&E preparation. (C) From an individual with minimal hippocampal pathology, for comparison to (D), with severe HS-A; note that the scale bars are the same for (C and D), indicating the large amount of atrophy of the hippocampal formation in (D). Scale bars: A, B = 20 µm; C, D = 4 mm. The pTDP-43 antibody used was the 1D3 clone (250) as reported previously (251).
From neuropathologic and clinical standpoints, there are relatively clear delineations between LATE-NC and other TDP-43 pathologic entities, including FTLD-TDP (10, 33–36). There are also relatively clear delineations between LATE-NC and other neurodegenerative disease categories, including Tau and α-synuclein pathologies. A minority of cases may fall outside of clear-cut diagnostic groupings. The focus of this review is on common neuropathologies; FTLD-TDP (1:1000 lifetime risk [37, 38]) will not be discussed in depth. Nonetheless, there are known to be unusual TDP-43 pathology cases that fall into diagnostic gray areas and some aspects of classification have been debated (33, 35, 39–42).
The staging system for LATE-NC severity (1), was based on prior work (first from Dr Keith Josephs and colleagues), that identified the stereotypical pattern of TDP-43 pathology in aging brains across a spectrum of disease severity (7, 43). The LATE-NC staging system was intended to be useful for routine neuropathological assessments (1). There are 3 recognized stages, based on the anatomic location and severity of TDP-43 pathology: LATE-NC Stage 1 is characterized by any TDP-43 pathology in the amygdala or hippocampal regions, but not a TDP-43-immunoreactive neuronal cytoplasmic inclusion (NCI) in both; LATE-NC Stage 2 indicates that there is at least 1 NCI in both the amygdala and the hippocampus regions; and, brains with LATE-NC Stage 3 have TDP-43 pathology in the amygdala region, hippocampal region, and middle frontal gyrus (35).
Other TDP-43 pathology staging systems have been developed that distinguish 5 or 6 stages of TDP-43 pathologic distribution in aging (7, 43); the additional stages provided a more complete delineation of TDP-43 pathologic expansion outside the medial temporal lobe, that is between LATE-NC stages 2 and 3. There are several reasons that the 3-stage LATE-NC system was chosen by a consensus panel of experts for routine neuropathologic examination (1): (1) The 3-stage system provides markers for the earliest, most usual, and most severe stages; (2) a higher number of stages would introduce more variability with correspondingly lower inter-rater reliability; (3) there are not compelling reasons to add new stages based on clinical-pathological correlation—that is, more pathological stages would not provide added information for clinicians or families to explain symptoms during life; and (4) additional sampling, sectioning, and staining was discouraged due to economic reasons for neuropathology labs. Despite these benefits of a 3-tier staging scheme for widely used diagnostic purposes, sampling more brain regions involved by TDP-43 pathology, using center-specific protocols, has provided insights in the research context (7, 43, 44).
Assessments of “early LATE-NC” (autopsy series that included minimal pathology cases) indicated that there are differing patterns of presumed incipient TDP-43 pathology in LATE-NC (45, 46). Some brains show early TDP-43 pathology primarily associated with neurons that are vulnerable to Tau pathology (47); these microlesions were termed “tangle-associated TDP-43” (48), partly because of their neurofibrillary tangle (NFT)-like histomorphology and partly because the intracellular inclusions often co-stain using antibodies against both Tau and TDP-43 proteins. A different group of LATE-NC cases show TDP-43 pathology in the medial temporal lobe that partially resembles FTLD-TDP staining patterns (45, 49). Still other cases show both Alzheimer disease neuropathologic change (ADNC)-like and FTLD-like patterns in the same brain (46). (There is poor inter-rater reliability in these diagnostic determinations [36]). It is important to note that despite the differing patterns of TDP-43 pathology, as a rule, in early LATE-NC, the pathological spread through the brain starts in or near the amygdala and then mostly maintains medial temporal lobe predominance (46). Even in the ∼15% of LATE-NC cases that progress to LATE-NC Stage 3, the density of TDP-43 pathology in the neocortex tends to remain considerably lower than that seen in FTLD-TDP (2, 33, 34).
Alzheimer disease neuropathologic change
The existence of both ADNC and LATE-NC in the same brain at autopsy is common and the combination is impactful according to clinical-pathologic studies (50–52). Having both ADNC and LATE-NC as co-pathologies is associated with a swiftly progressing and severe disease course, with more brain atrophy, in comparison to either LATE-NC or ADNC alone (7, 52–55). Cases with ADNC+LATE-NC also are disproportionately likely to have comorbid Lewy body disease (LBD) (53, 56); this suggests widespread proteinopathy that theoretically may extend beyond Tau, Aβ, TDP-43, and α-synuclein (57). The exact associations between Aβ and LATE-NC are unclear but neuritic amyloid plaques (which tend to contain Tau pathology) are associated with increased TDP-43 pathology in LATE-NC (58). Regardless, approximately 50% of brains with severe ADNC (Braak NFT stages V or VI) have LATE-NC, whereas only ∼25% of cases without any ADNC (i.e. lacking amyloid plaques or advanced Braak NFT stages) have LATE-NC (2). See Figure 2 for a heuristic, visual schematic of ADNC, LATE-NC, and hippocampal sclerosis (HS) according to observations from community-based autopsy cohorts.
Figure 2.
A schematic representation of some of the common pathologies in human populations—how they often overlap and are driven by separate pathogenetic mechanisms. Note that around half of severe Alzheimer disease neuropathologic change (ADNC) cases have comorbid LATE-NC. Somewhat fewer than half of LATE-NC cases have comorbid hippocampal sclerosis of aging (HS-A). Cases with LATE-NC+HS-A lacking ADNC should be considered a subtype of “pure” LATE-NC because it is likely that TDP-43-related mechanisms drive the HS-A pathology. A minority of cases with HS pathology lack LATE-NC; these are associated with other mechanisms and do not map onto the same disease paradigm as HS-A.
“Pathologic synergy” refers to hypothesized interaction(s) between different subtypes of pathology and implies that 1 pathology promotes the other, or that both directly exacerbate each other (59–61). Tauopathy is a subcomponent of ADNC that may synergize most directly with TDP-43 proteinopathy in LATE-NC (47, 60–64). In comparing between Tau and TDP-43 pathologies, both are common but Tau pathology is more common; some degree of Tau pathology is universal by middle age (65). Both Tau NFTs and TDP-43 NCIs are by definition intracellular within neurons, and NFTs and NCIs are often seen in the same neuronal cytoplasm (47, 48, 60, 66). More severe ADNC is associated with higher likelihood of LATE-NC (2, 67), but see (68). Further, the presence of LATE-NC is also associated with more severe (higher Braak NFT stage) primary age-related tauopathy (PART) (2), which lacks amyloid plaques by definition (69). Numerous other dementia-related conditions have been shown to comorbidly express the pathological phenotypes of Tau and TDP-43 proteinopathies (24, 70, 71).
The association between ADNC and LATE-NC appears to remain robust across the aging spectrum but it is most pronounced in advanced old age. To convey data related to odds ratios of LATE-NC for high ADNC versus no/low/intermediate ADNC stratified by age at death (categorized into 4 responses) in the NACC Neuropathology Dataset see Figure 3 (72, 73). These data derive from a sample exactly as described in a recent paper using NACC data (74). As in other autopsy cohorts, LATE-NC is relatively likely to occur in cases with high ADNC. The same data were analyzed to convey the estimated probabilities of different neuropathologic findings across the aging spectrum (Fig. 4). Note that LATE-NC maintains an increase in prevalence year-by-year in the oldest-old whereas ADNC severity peaks earlier in the human aging spectrum, as shown previously (1, 74–76).
Figure 3.
Shown are data from the NACC Neuropathology Data set (n = 2240 cases) using methods as described in detail previously (74) to indicate the associations between LATE-NC with Alzheimer disease neuropathologic change (ADNC; panel A) and with brain arteriolosclerosis (B-ASC; panel B) across the aging spectrum. In this cohort, ∼25% of cases had LATE-NC. Odds ratios of LATE-NC for high ADNC versus no, low, and intermediate ADNC (A) and for moderate and severe B-ASC vs none and mild B-ASC (B) estimated by logistic regression models unadjusted (red and orange) and adjusted for sex and the number of ε4 alleles of APOE (blue and green).
Figure 4.
Shown are the same data as in Figure 3 (n = 2240 NACC cases) as described in detail previously (74) to indicate the estimated probabilities of subtypes of pathology, along the human aging spectrum. Since NACC has many clinic-based cohorts that may be less population-representative (252), we split out the cases from the University of Kentucky ADRC (UK-ADRC), which follows a community-based cohort (253). Estimated probabilities with 95% confidence intervals over age at death by logistic regression models for severe ADNC (red), LATE-NC Stage >1 (blue), and moderate-to-severe brain arteriolosclerosis (B-ASC; green) in the NACC cohort excluding the UK-ADRC cohort (A) and in the UK-ADRC cohort itself (B). Note that the probability of severe ADNC declines whereas LATE-NC and B-ASC pathologies increase.
There are data in support of the hypothesis that the synergistic associations between Tau pathology and TDP-43 pathology in LATE-NC may be bidirectional in terms of causation, that is each may promote the occurrence of the other. Relevant clues can be gleaned from an analysis of the anatomical distribution of Tau and TDP-43 pathologies in autopsy cohorts. An early anatomical site of involvement of NFTs in ADNC is the entorhinal cortex (77), whereas an early site of involvement of TDP-43 pathology in some LATE-NC cases is the hippocampal dentate gyrus (43, 78). If tauopathy leads to TDP-43 pathology, then one would predict that early in ADNC, there may be colocalization of Tau and TDP-43 pathology in entorhinal cortical neurons vulnerable to NFTs. These “TDP-43 associated tangles” (as mentioned above) have been reported (45, 48, 79). On the other hand, if TDP-43 pathology promotes tauopathy, one would expect “pure LATE-NC” (lacking substantial ADNC) to have Tau-immunoreactive intraneuronal inclusions, colocalized with TDP-43 NCIs, in the dentate granule cells, well before predicted by classic Braak NFT staging (77). This also has been shown to be true (47, 80, 81). A visual depiction of these phenomena is presented in Fig. 5.
Figure 5.

Aspects of pathologic synergy between Tau pathology and TDP-43 pathology may be bidirectional, and distinctive in different anatomical locations. Shown is a coronal section of a hippocampal formation stained with H&E to indicate the anatomic location of the dentate gyrus (dg), CA1, subiculum (Sub), and entorhinal cortex (ERC). The dg commonly shows TDP-43-immunoreactive NCIs in cases with LATE-NC. In these cases, even with minimal Alzheimer’s disease pathology otherwise, one tends to find Tau-immunoreactive NFTs that resemble NCIs histomorphologically. By contrast, the ERC is a location known to be vulnerable to early Tau NFT development. In this region, one may find TDP-43-immunoreactive inclusions that resemble Tau NFTs. These are circumstantial evidences to support the hypothesis that Tau pathology can promote TDP-43 pathology, and vice versa. Scale bars: H&E = 2 mm; dentate gyrus = 30 µm; entorhinal cortex = 40 µm.
Additional reports have emerged from different experimental systems supporting the idea of bidirectional synergies between Tau and TDP-43 pathologies. Since normal TDP-43 function is partly related to transcript splicing regulation, and the cognate Tau-encoding gene (MAPT) transcript splicing can be linked to Tau pathology, it is both logical and experimentally verified that TDP-43 pathologic changes may promote Tau dysregulation and pathology (82–84). Further, a MAPT mutation has been reported with various comorbid pathologies that include TDP-43 proteinopathy (85). As such, we conclude that there are multiple data in support of the hypothesis that Tau pathology promotes TDP-43 pathology, and vice versa.
Age-related tau astrogliopathy
Another subtype of tauopathy associated with LATE-NC is age-related tau astrogliopathy (ARTAG). Unlike NFTs, which by definition are Tau inclusions in neurons, ARTAG is characterized by Tau pathology in astrocytes (86, 87) (Fig. 6). Tau-immunoreactive astrocyte pathologies in ARTAG may display a range of histomorphologic appearances, including “thorn-shaped” and “granular-fuzzy” astrocytes (as visualized with Tau immunohistochemistry) that may be seen in any anatomic area but particularly in subpial, subependymal, and white matter regions (88–90). ARTAG is likely to occur in brains with comorbid TDP-43 pathologies, including LATE-NC (91–94). For example, Forrest et al showed that ARTAG, and a novel Tau pathology termed age-related tau oligodendrogliopathy (ARTOG), were associated with LATE-NC (94). Sordo et al (74) and Katsumata et al (92) confirmed that ARTAG was associated with LATE-NC. Yokota et al (95) reported that the presence of amygdala granular-fuzzy astrocytes was associated with LATE-NC as well as with Tau-immunoreactive argyrophilic grain pathology. Notably, like some other pathologies discussed herein, ARTAG is strongly associated with advanced age, but unlike some others, is more prevalent in males (74, 96).
Figure 6.
Photomicrographs depict select pathologic features associated with LATE-NC, including Tau NFTs (A), ARTAG (B), and brain arteriolosclerosis (C, D). Panels A and B shows sections stained for phosphorylated Tau protein with immunohistochemistry, and the photomicrographs were taken at the same magnification. Note that the stained structures in (A) depict pathology within neuronal cells, whereas the pathology in (B) shows star-shaped astrocytes (blue arrow), and cells and foot processes that surround a small blood vessel (yellow arrow). (C) A relatively normal-appearing small arteriole in human brain for comparison to (D) that shows a human brain region with moderate-to-severe arteriolosclerosis. Note the rim of inflammatory cells outside the media of the blood vessel (D). Scale bars: A, B = 60 µm; C, D = 30 µm. The pTau antibody used was the PHF-1 clone (254) as reported previously (251).
The mechanisms underlying the pathological synergies or common causes shared by TDP-43 pathology and ARTAG are currently poorly understood. Upstream factors such as traumatic brain injury (TBI) and cerebrovascular pathologies may stimulate both TDP-43 pathology and ARTAG (74, 97, 98). However, there may be actual synergy, wherein TDP-43 dysregulation promotes ARTAG and/or vice versa. Some related insights may be learned by studying rare conditions. For example, a TARDBP mutation was associated with a pathological phenotype that included ARTAG-like glial tau pathology (99). Further, a case was identified with GBE1 mutations leading to both ARTAG-like and TDP-43 pathologies (100). Another FTLD-linked GRN mutation also was correlated with both ARTAG-like and TDP-43 pathologies (101). In a larger autopsy series, ARTAG was relatively common in brains affected by FTLD-TDP (63). These observations again underscore the synergistic interactions and comorbidity of Tau and TDP-43 pathologies, affecting astrocytes as well as other cell types in the brain.
Hippocampal sclerosis (HS) of aging (HS-A)
HS was recognized as a neuropathologic feature associated with dementia in 1994 (102), and the association between HS and TDP-43 proteinopathy was first reported in 2007 (103); both of these advances were accomplished by Dr Dennis Dickson and colleagues at the Mayo Clinic (Fig. 1). Other researchers have helped to gain further insights into the clinical, pathological, and radiographical features of LATE-NC with comorbid HS (104–114). It is also notable that, when multiple variable regression-based statistical models are applied, factoring in TDP-43 proteinopathy and other pathologies, HS is associated with substantial independent cognitive impairment (1, 3, 4, 78, 115–118).
Although the presence of HS heralds an impactful disease process in a clinical sense, the basic definition of HS, and even the term itself, are problematic. According to the consensus-based definition, HS refers to “pyramidal cell loss and gliosis in CA1 and subiculum of the hippocampal formation that is out of proportion to AD neuropathologic change in the same structures” (119). This captures key aspects of the pathology but unfortunately this definition is highly nonspecific and, given the histopathological variability in actual practice, individual neuropathologists must apply their own ad hoc criteria for the many cases that have intermediate levels of cell loss and gliosis (120). It also is widely known that a number of different diseases produce a histopathologic picture that meets the definition. For example, the clinical literature usually refers to a seizure disorder when applying the term HS, and pathology that is labeled HS is also seen in completely different circumstances related to brains affected by anoxia, hypoglycemia, and various infections that target the hippocampus (121–123). We and others have proposed a term that is more specific for HS in the context of LATE-NC, which is “hippocampal sclerosis of aging,” or HS-A (92, 113, 124). The term HS-A is useful and we will apply it subsequently in this present review but it has the drawbacks of not being consensus-based and also is still not specifically defined in terms of the precise histopathologic features other than LATE-NC, cell loss, and gliosis in the hippocampal formation. More rigorous and standardized terminology will be required in the future.
Despite the uncertainties, HS-A is a research focus where data-driven, causal pathoetiologic inferences can be made. This is because the distribution of pathologic findings in multiple autopsy cohorts are relatively consistent and instructive: the great majority of cases with HS in aging have LATE-NC; LATE-NC is approximately twice as common as HS-A; and brains with unilateral HS-A generally have bilateral TDP-43 proteinopathy (75, 78, 111–113, 125–129). These observations lead to the following corollary conclusions: HS-A occurs “downstream” of LATE-NC, and thus LATE-NC-related processes are the major drivers of HS in aging. Given the nonspecific implications of the term HS, the extant data strongly imply that LATE-NC is not only a co-pathology with HS-A. Instead, LATE-NC + HS-A is best considered a severe manifestation or subset of “pure LATE-NC,” with HS-A in LATE-NC analogous to loss of pigmented neurons in Parkinson disease substantia nigra.
Many unanswered questions about HS-A remain. For example, why do only a subset of cases with LATE-NC have HS-A? And, is HS-A influenced by (caused by, or synergistic with) ARTAG and/or cerebrovascular disease? Given that HS in other contexts is associated with vasculopathy and/or ischemia-reperfusion (121), and astrocytes are integral components of the blood-brain barrier, the potential seems high for HS to be exacerbated by astrocyte dysfunction. In animal models, TDP-43 perturbations lead to robust astrocytosis, inflammation, and blood-brain barrier dysfunction (30, 130, 131). Recently, we showed in human brains that hippocampal astrocytosis in LATE-NC+HS-A was more severe than that of ADNC, LATE-NC, or HS alone (132). These data indicate that LATE-NC+HS-A may be tied to TDP-43 dysregulation, astrocytic signaling, blood-brain barrier dysfunction, and/or neuroinflammation.
Cerebrovascular pathology, particularly brain arteriolosclerosis (B-ASC)
Another category of brain pathology that has been associated with LATE-NC is cerebrovascular disease. An early study described cerebrovascular pathologies in a cohort of 13 individuals with HS and dementia (102). Ensuing autopsy series also showed enrichment for cerebrovascular pathology in brains with LATE-NC and HS (Table 1). The subtype of cerebrovascular pathology most often associated with LATE-NC + HS-A was B-ASC, characterized by thickened and/or dysmorphic arterioles in the brain (133) (Fig. 6).
Table 1.
Published studies that evaluated the association between cerebrovascular pathologies and LATE-NC/hippocampal sclerosis (HS) phenotypes
| Ref. | Year | Pheno-type(s) | Associated with LATE-NC and/or HS? |
|||||||
|---|---|---|---|---|---|---|---|---|---|---|
| Sample size (total) | Arterio-loscler-osis | Athero-scler-osis | Micro-infarcts | Lacunar infarcts | Large infarcts | CAA | Note | |||
| (102) | 1994 | HS | 13 | Overall high level of cerebrovascular pathologies in HS case series | No control group or type-specific associations | |||||
| (106) | 2002 | HS | 48 | “…medical record review indicated higher frequencies of clinical stroke and neuroradiologic white matter abnormalities in the HS group” | HS pathology was not associated with vascular neuropathology phenotypes | |||||
| (133) | 2014 | HS | 2218 | Yes | No | No | No | No | No | Study included digital pathology methods of vessel geometry in a subset; also included NACC NP data. |
| (235) | 2018 | LATE-NC | 929 | Yes | No | No | No | No | No | NACC study focusing on TDP-43 pathology yes/no |
| (236) | 2020 | HS and LATE-NC | 749 | Yes (post. water-shed only) | No | No | N/A | No | Yes | Rush U./ROS-MAP study. Only posterior watershed brain arteriolosclerosis was associated with LATE-NC |
| (237) | 2020 | LATE-NC | 616 | Yes | No | No | No | No | No | NACC study focusing on TDP-43 pathology yes/no |
| (128) | 2020 | HS and LATE-NC | 359 | N/A | Yes (with HS) | (Infarct groups clustered together) Not associated with either HS or LATE-NC | N/A | U. Pitt ADRC study; HS but not LATE-NC associated with atherosclerosis. | ||
| (238) | 2021 | HS and LATE-NC | 98 | Yes | No | No | N/A | N/A | No | Duke/UNC ADRC study; HS and LATE-NC associated with brain arteriolosclerosis |
| (239) | 2021 | HS | 122 | HS was associated LATE-NC and with “vascular changes in the basal ganglia” but not with large infarcts or CAA or ADNC | U. Antwerp cohort, focusing on FTLD and vascular pathology system of Deramecourt (138) was used | |||||
| (240) | 2022 | HS and LATE-NC | 203 | Yes | No | No | N/A | N/A | No | Duke/UNC ADRC study; LATE-NC associated with brain arteriolosclerosis in young-old and older-old |
| (92) | 2023 | HS and LATE-NC | 408 | Yes | No | “MVL”; No | N/A | N/A | No | UCI 90+Study; readouts here relate to quantitative assessments of HS, not the dichotomous HS± ones. |
| (241) | 2023 | HS and LATE-NC | 4354 | No | No | No | No | No | No | International multicenter study with disparate brain arteriolosclerosis scoring methods used across centers |
ADRC, AD Research Center; CAA, cerebral amyloid angiopathy; MVL, microvascular lesions; N/A, data not available; ROS-MAP, Rush Religious Order Study/Memory-Aging Project; UCI, University of California Irvine; UNC, University of North Carolina; U.Pitt, University of Pittsburgh (Other abbreviations defined elsewhere.)
Like most of the pathological features discussed in this review article, B-ASC is at present incompletely understood, inexactly defined, and not always diagnosed using standardized methods (134–136). This adds a potentially significant confounding factor to scientific inquiry, particularly when trying to integrate the results from cohorts that have divergent operationalization of B-ASC. Readers are referred to prior reviews (134, 137–140). Although B-ASC is associated with diabetes and hypertension (particularly in persons younger than age 80 at death) and B-ASC is also associated with HS, HS has not been shown to be associated with either diabetes or hypertension (117, 133, 134, 136, 141). These data indicate that the pathological phenotype described as B-ASC that is driven by hypertension and/or diabetes is nonidentical (or completely different) in comparison to that which is associated with LATE-NC/HS-A. For the sake of the present article, the term B-ASC should be considered a proxy label for a subset of brain microvasculopathy that is currently in its early stages of being understood. Figure 3B provides a depiction of LATE-NC odds ratios for moderate/severe B-ASC versus none/mild B-ASC stratified by the 4 age at death categories in the NACC Neuropathology Dataset (72, 73).
The neurovascular unit (NVU), which appears to be disrupted in B-ASC (134, 135), comprises a number of different cell types that together regulate blood flow, filter and exchange substances between blood and brain, modulate neuroinflammation, and perform other functions (142, 143). Correspondingly, the influence of TDP-43 pathology may be exerted on behalf of multiple NVU cell types that include astrocytes, endothelial cells, and/or vascular mural cells, that is pericytes and smooth muscle cells. A subset of TDP-43 proteinopathy has been reported within astrocyte foot processes near capillaries (144). These TDP-43-immunoreactive structures were termed Lin bodies (Fig. 1B) and their influence on vascular function is unknown (35, 144). More work is required to characterize the presence and impact TDP-43 pathology that takes place within glial cells in LATE-NC and other conditions. Otherwise, astrocytes in LATE-NC were discussed above in the section related to ARTAG.
Intriguing recent research focusing on endothelial cells has uncovered another mechanism whereby TDP-43 dysregulation may lead to vasculopathy (and/or vice versa). Endothelial cell TDP-43 knockdown or dysregulation in animal models led to impaired endothelial function and attenuated angiogenesis (30, 145, 146). These influences could cascade to other cell types and promote broader levels of dysfunction in LATE-NC. This would lead to increased inflammation, secondary neurodegeneration, and/or impairment of mechanisms of resilience. Since endothelial cells are often involved in cerebral amyloid angiopathy (CAA) (147, 148), it is of interest that most prior studies do not find an association between CAA and LATE-NC (Table 1).
In addition to affecting astrocytes and endothelial cells, LATE-NC may also have deleterious impacts on the NVU’s mural (blood vessel wall) cells. Using microvessel extracts from the parietal cortex of participants of the Rush University Religious Orders Study (149), in a Western blotting analysis, Bourassa et al (150) found that levels of mural markers referent to smooth muscle or pericyte cells were correlated with TDP-43 pathology and protein cleavage products. There is also support for a hypothetical mechanism linking NVU mural cell dysfunction with HS-A through a genetically inherited mechanism, as described below.
Another potentially disease-driving mechanism, although mostly beyond the purview of the current article, is senescence. The prevalence of B-ASC (like LATE-NC, HS-A, and ARTAG), increases with every added year in advanced old age (74, 75, 134, 151, 152). By contrast, the prevalence of pathologies such as ADNC, FTLD-TDP, and others level off and decrease in advanced old age (38, 76, 152). The association between B-ASC and LATE-NC is stronger in the “oldest-old” (Fig. 3B). Note that, whereas Figure 4 shows the proportion with moderate-severe B-ASC, over 80% of individuals beyond age 80 had some identifiable B-ASC (141, 153, 154). Small-vessel pathologic changes have been shown to be a component of senescence, across multiple organs and organisms (155–160). However, the LATE-NC-relevant senescence-driven B-ASC mechanisms are poorly understood and data from other contexts may not correlate perfectly with human brain-specific mechanisms.
Other conditions that may influence and/or resemble the brain changes of LATE-NC
Two additional pathological paradigms with aspects of overlap with LATE-NC are LBDs and the brain changes that are associated with repetitive TBI—chronic traumatic encephalopathy (CTE). The term LBDs indicates the presence of inclusion bodies in neurons and/or glia comprising misfolded α-synuclein. There are several notable phenomena at the nexus of LATE-NC and LBDs. Given the likelihood of each occurring in isolation, a disproportionately large proportion of brains have a combination of pathologies that include LATE-NC, ADNC, and Lewy bodies (52). This “quadruple misfolded protein” (Aβ, Tau, TDP-43, and α-synuclein pathologies in the same brain) phenotype is characterized by a swift and severe dementia syndrome, often with clinical psychotic symptoms (52, 53). The prevalence of this pattern of pathologies indicates that polyproteinopathy-influencing pathways of pathogenesis exist that are imperfectly understood (52, 53, 161). The amygdala has been noted to be an anatomical region where LBD as well as TDP-43 pathology are often involved early in pathogenesis (59, 117). In cases with early LATE-NC that also have amygdala LBD co-pathology, the TDP-43 pathology pattern tends to be different from that seen in cases lacking Lewy bodies (46, 162).
CTE is another clinical-pathological entity with aspects of overlap with LATE-NC. While the pathognomonic features of CTE involve intraneuronal Tau pathology (163, 164), TDP-43 pathology may exist as a co-pathology, even among individuals age 50 and younger. The TDP-43-like pathology in CTE may be localized to the hippocampus, but often it occurs in a brain region that tracks very separately from the stereotypical LATE-NC stages (165) and other TDP-43 pathologic staging systems (7, 43). Intriguingly, CTE may also show HS pathology, even among individuals of an age group (persons younger than age 60 years) who otherwise would be unlikely to have HS-A (165).
Collectively, the multitude of pathologies with overlapping features provides important clues about how homeostatic or deleterious influences in different brain conditions are linked to healthy and disease states. These phenomena also indicate the challenges of clinical-pathological correlation when an incomplete list of neuropathologies are factored in to statistical models—1 pathologic feature may actually be a proxy measure for others. See Figure 7 for a schematic presentation of prior work, including pathology-pathology associations and selected references. So far, we have highlighted the pathologic manifestations that are associated with cognitive impairment. These data help to contextualize parallel studies of upstream factors that influence cognitive impairment or resilience.
Figure 7.

Schematic depiction of TDPopathies (diseases with TDP-43 pathology) including LATE-NC which is most common by far, along with LATE-NC-associated neuropathologic phenotypes. References for the studies that reported these associations shown as lines between subsets of pathology: (a) Refs. (2, 103, 255); (b) Refs. (235, 236, 238); (c) Refs. (74, 94, 95); (d) Refs. (103, 110); (e) Refs. (103, 110); (f) Ref. (74); (g) Ref. (92).
GENETIC ASSOCIATIONS WITH LATE-NC AND HS-A
Genetic studies provide critical insights into disease-related mechanisms. To date, 4 genes have been reported (with replication) to harbor risk alleles associated with LATE-NC: Granulin (GRN), Transmembrane protein 106B (TMEM106B), Apolipoprotein E (APOE), and Sortilin related receptor 1 (SORL1) (111, 166–172). We will discuss separately the association between HS-A and ATP-binding cassette sub-family member 9 (ABCC9) genetic variants. There are a number of other genes that have demonstrated suggestive or otherwise not-yet replicated associations with LATE-NC/HS phenotypes (Table 2).
Table 2.
Genes linked to LATE-NC/hippocampal sclerosis (HS) neuropathologic phenotypes
| Main finding replicated? | Gene (protein) | Disease-related phenotype(s) | Notes | Selected Refs. |
|---|---|---|---|---|
| Replicated | TMEM106B | LATE-NC, FTLD-TDP, pediatric leukodystrophy, viral pathogenesis | TMEM106B C-terminal amyloidogenic fragments form disease-associated “inclusion bodies” | (129, 171, 173, 188, 242, 243) |
| GRN (Progranulin) | LATE-NC, HS, FTLD-TDP, neuronal ceroid lipofuscinosis | GRN is an inflammation-regulating growth factor also implicated in cancers | (167, 170, 244, 245) | |
| APOE | LATE-NC, ADNC, Lewy body diseases, hypercholesterolemia | APOE is prime driver of late-onset AD neuropathologic changes | (111, 129, 171, 205, 236, 246) | |
| SORL1 | LATE-NC+ADNC, clinical AD, clinical FTLD | SORL1 variants are associated with ADNC+LATE-NC pathologic phenotype | (211, 213, 214, 228) | |
| ABCC9 (SUR2) | HS, Cantu syndrome, ABCC9-related intellectual disability and myopathy syndrome, cardiomyopathy and cardiac arrhythmias | ABCC9 variants are also associated with vascular malformations and white matter hyperintensities | (129, 169, 170, 217, 247) | |
| Not yet replicated | KCNMB2 | HS, possibly ALS, Epilepsy | KCNMB2 encodes a potassium channel expressed in hippocampus | (129, 166, 230, 231, 248) |
| WWOX | LATE-NC, HS, arteriolosclerosis, clinical AD, spinocerebellar ataxia, epilepsy | WWOX was a GWAS “hit” for clinical AD and pathological LATE/HS | (169, 225, 226, 229, 249) | |
| AHRGEF28 (RGNEF) | LATE-NC (suggestive), Cri-du-Chat syndrome | ALS lesions have been shown to stain for RGNEF | (233), in press | |
| TPCN1 | LATE-NC (suggestive), clinical AD | TPCN1 is a lysosomal protein linked by GWAS to clinical AD | (213, 228) |
Genetic variants in GRN and TMEM106B were initially demonstrated to be associated with the risk of FTLD-TDP (173–176). These same genes have now also been shown to have genetic variation associated with risk of LATE-NC and HS-A. The GRN and TMEM106B risk-associated alleles exhibit moderate effects on risk/protection. These phenomena provide compelling evidence for partial pathogenetic overlap between FTLD-TDP and LATE-NC.
As a gene containing variants with common and high-impact risk alleles, TMEM106B has a large influence on public health. Aoki et al (168) reported that the frequency of the C-allele of TMEM106B single-nucleotide polymorphism (SNP) rs1990622 in HS/HS-A was lower than that in non-HS controls. There was further replication of increased risk for HS-A associated with each copy of the T-allele of TMEM106B rs1990622 (169, 170, 177, 178). In linkage with this TMEM106B risk-associated SNP, a large haplotype (a set of continuous alleles, inherited together) that incorporates many introns of the TMEM106B gene was associated with differential expression of TMEM106B protein (179, 180). Further, the TMEM106B gene appears to be pleiotropic for multiple diseases (181–184), and the LATE-NC risk allele in TMEM106B may influence healthy brain aging (179, 185). Recently published data from Dr. Rosa Rademakers and colleagues have demonstrated that a carboxy-terminal fragment (CTF) of the TMEM106B polypeptide (amino acid residues 120–254) forms amyloidogenic fibrils that can be visualized with immunohistochemistry as intracellular aggregates in many aged human brains (186–188). The amount of TMEM106B CTF fibrillary aggregates was correlated with the risk allele (they were found in both FTLD-TDP and LATE-NC cases) and they may be highly relevant to LATE-NC pathogenesis, at least in some cases (186, 189–191).
With regard to GRN, HS/HS-A risk in aged individuals was associated with the T-allele of the GRN SNP rs5848 (167, 171). The same genetic risk variant has been associated with increased inflammatory mediators (e.g. AXL and CLU) in cerebrospinal fluid (192). Separate studies have found that GRN gene products (progranulin/granulins) are key mediators in inflammation and wound repair (193, 194). Further, the proteins coded for by both TMEM106B and GRN have been shown to play important roles in endosomal/lysosomal biology, and there is experimental evidence for functional interaction of these proteins (180, 195–200).
Both risk alleles from the TMEM106B and GRN genes are differentially represented when comparing populations of African and European ancestries: in persons with European ancestry, the TMEM106B risk-associated allele is most frequent (thus, more European-ancestry individuals harbor risk genotypes than not), whereas in persons of African ancestry the TMEM106B risk allele is distinctly less frequent, suggesting differing local genetic architectures (201). By contrast, the risk allele for the GRN SNP rs5848 is far more common in persons with African ancestry than in persons of European ancestry (201). Coupled with the substantial associative impact of these alleles in terms of disease risk (129), these observations suggest that future therapeutic strategies for LATE-NC and FTLD-TDP may have some different outcomes based on the patient populations that are being served.
In addition to TMEM106B and GRN, the APOE ε4 allele also has been associated with increased risk for LATE-NC (172, 202–205). Not all studies found evidence of a direct association between APOE genotype and risk for LATE-NC or HS-A (75, 106, 111, 125, 206). Few individuals with the APOE ε4 allele survive into advanced old age without any Aβ plaques (207, 208), and it remains to be seen exactly how the APOE genotype promotes TDP-43 proteinopathy (there may be multiple pathways involved). Nevertheless, the strong ADNC-driving influence of APOE genotypes is well known, and, along with the pathologic observations described above, these data underscore that brain changes in ADNC, like FTLD-TDP, ARTAG, B-ASC, and CTE, have molecular mechanisms that overlap with LATE-NC.
Another pathogenetic clue comes from the associations between SORL1 and the ADNC+LATE-NC phenotype. SORL1 genetic variants were previously linked to AD risk and the SORL1 protein is involved in amyloid-β clearance and endosomal processing of proteins, so it has a highly credible association with ADNC (209–211). SORL1 mutations have also been linked to FTLD (212). We found an association signal between SORL1 variation and LATE-NC (213); more specifically, the SORL1 variant(s) may drive a subtype of LATE-NC that tends to present with comorbid ADNC. Consistent with that hypothesis, a post-hoc analysis indicated that the rs74685827 SORL1 risk allele was associated with the ADNC+LATE-NC phenotype (213). A separate group of investigators discovered a different genetic variant that was linked to a family lineage with clinical dementia and ADNC+LATE-NC co-pathologies (214).
Genetics have also provided early insights into why a subset of cases with LATE-NC are susceptible to developing comorbid HS-A. The ABCC9 gene association with LATE-NC/HS-A was discovered via genome-wide association study (GWAS) (169). The results of subsequent studies were variable and 1 study failed to replicate the association between ABCC9 and either LATE-NC or HS-A (215). Nonetheless, there has been replication; the finding of the associations between ABCC9 gene variants and HS-A (170), and MRI detected brain atrophy (216), were reported in separate samples from the initial GWAS (169). The gene product of ABCC9, the sulfonylurea receptor type 2 (SUR2) protein, is a component of an ATP-sensitive potassium (“KATP”) channel (217). KATP channels serve as metabolic sensors that regulate blood flow that also constitute a molecular substrate for ischemic preconditioning (217–219). The ABCC9 risk genotype also implicates thyroid hormone dysregulation in LATE-NC (220). When both LATE-NC and HS-A outcomes were included in a statistical model, the ABCC9 alleles that were associated with altered gene expression of ABCC9/SUR2 (i.e. were expression quantitative trait loci [eQTLs]) were associated with risk for HS-A, but not specifically LATE-NC (129). It is also notable that ABCC9/SUR2 is known to be highly expressed in pericytes, which are critical mural cells in vascular signaling (217, 221, 222). HS pathology is associated with vasculopathy even in cases lacking LATE-NC (223, 224). Overall, the existing data support the hypothesis that genetic variation at ABCC9 may render some individuals with LATE-NC vulnerable (or resistant) to HS-A.
In addition to the above genetic loci that have been replicated in relation to their association with LATE-NC/HS-A risk, there have been a number of additional genes and loci with suggestive (but not yet replicated) associations with LATE-NC/HS-A pathologic phenotypes (Table 2). Here, we highlight 2 such genetic loci: WWOX (WW Domain Containing Oxidoreductase), and ARHGEF 28 (Rho guanine nucleotide exchange factor 28)/RGNEF. The WWOX gene is on human chromosome 16 and the WWOX protein plays roles in transcription regulation, glucose metabolism, and central nervous system development (225). In a GWAS of clinically assessed AD-type dementia, a SNP in the WWOX locus, rs62039712, was associated with clinical AD (226). Subsequently, this variant was not found to be an eQTL in brain tissues, and later AD dementia-related GWAS did not identify WWOX as a dementia risk gene (227, 228). In a separate GWAS using HS as a phenotype, WWOX returned several suggestive loci, but none were statistically significant after correcting for multiple comparisons (169). However, in a later gene-focused study, several different WWOX genetic variants again showed associations with LATE-NC, HS, and/or brain arteriolosclerosis (229). These associations remained largely unchanged after adjustment for ADNC, suggesting that these associations were independent of ADNC (229). Another gene related to LATE-NC/HS-A risk is ARGHEF28, which encodes the RGNEF protein. ARHGEF28 has been reported as a putative ALS gene (230, 231). Along the same lines, its cognate protein RGNEF has been found to localize to hallmark TDP-43-immunoreactive inclusion bodies in ALS patient spinal cord motor neurons (232, 233). We recently found that the ARHGEF28 SNP rs80190672 was associated with LATE-NC (234). This genotype-phenotype correlation may provide another point of etiologic connection between LATE-NC and the ALS/FTLD disease continuum.
Genetics discoveries to date pertinent to LATE-NC/HS-A should be considered the “low-hanging fruit” that are mostly referent to European-ancestry cohorts. More work is required to elucidate additional associations between genetic factors and LATE-NC/HS-A risk. This research is challenged by the requirement for larger cohorts, broader genotyping (evaluating genetic phenomena beyond SNPs), high-quality neuropathology data, old-age research volunteers, and ethnically and racially diverse samples comprising a broad range of exposures and experiences. Study design improvements in these areas will no doubt enable future discoveries. This research has the potential to aid precision-based health care and perhaps to help guide clinical trial candidacy.
CONCLUSION
Because LATE-NC is associated with different comorbid neuropathologies and with a set of intriguing genetic risk factors, a review of findings from human autopsy studies provides the bases for generating hypotheses about LATE-NC pathogenesis. The patterns of co-pathologies in multiple autopsy series highlight the possible bidirectional importance of TDP-43 pathology along with Tau pathology and B-ASC/vasculopathy. Genetics studies indicate the importance of endolysosomal pathways, ADNC pathways, TMEM106B CTF inclusions, and potassium (“KATP”) channels. A preliminary mechanistic schema is shown in Figure 8. Implications of these data may be far-reaching and clinically relevant. The partial success of anti-Aβ immunotherapies in slowing the progress of ADNC-driven dementia provides new urgency for sharpening our understanding of the underlying mechanisms, and, ultimately, the potential for therapeutic remedies related to non-Aβ contributions to dementia.
Figure 8.

A review of human phenomenology (pathologic and genetics studies) provides preliminary clues about inter-relationships between pathology-driving factors in LATE-NC. TDP-43 pathology is the disease-defining feature of LATE-NC, and often is accompanied by several co-pathologies and/or predicted by genetic risk factors. More specifically, LATE-NC tends to be correlated with Tau pathology and blood-brain barrier dysfunction, which may play added roles in terms of pathologic and, ultimately, clinical manifestations.
ACKNOWLEDGMENTS
We are profoundly grateful to the research participants, caregivers, clinicians, staff, and colleague scientists who contributed to this study.
Contributor Information
Peter T Nelson, Department of Pathology and Laboratory Medicine, University of Kentucky, Lexington, Kentucky, USA; Department of Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA.
David W Fardo, Department of Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA; Department of Biostatistics, University of Kentucky, Lexington, Kentucky, USA.
Xian Wu, Department of Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA; Department of Biostatistics, University of Kentucky, Lexington, Kentucky, USA.
Khine Zin Aung, Department of Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA; Department of Biostatistics, University of Kentucky, Lexington, Kentucky, USA.
Matthew D Cykowski, Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, Texas, USA.
Yuriko Katsumata, Department of Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA; Department of Biostatistics, University of Kentucky, Lexington, Kentucky, USA.
FUNDING
This study was supported by NIH grants numbers R01 AG061111, R01 AG057187, P30 AG072946, RF1 NS118584, RF1 AG082339, P01 AG078116, R01AG082730.
CONFLICT OF INTEREST
The authors have no duality or conflicts of interest to declare.
REFERENCES
- 1. Nelson PT, Dickson DW, Trojanowski JQ, et al. Limbic-predominant age-related TDP-43 encephalopathy (LATE): Consensus working group report. Brain 2019;142:1503–27 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Nelson PT, Brayne C, Flanagan ME, et al. Frequency of LATE neuropathologic change across the spectrum of Alzheimer’s disease neuropathology: Combined data from 13 community-based or population-based autopsy cohorts. Acta Neuropathol 2022;144:27–44 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Nelson PT, Abner EL, Schmitt FA, et al. Modeling the association between 43 different clinical and pathological variables and the severity of cognitive impairment in a large autopsy cohort of elderly persons. Brain Pathol 2010;20:66–79 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Boyle PA, Yu L, Leurgans SE, et al. Attributable risk of Alzheimer’s dementia attributed to age-related neuropathologies. Ann Neurol 2019;85:114–24 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Flanagan ME, Cholerton B, Latimer CS, et al. TDP-43 neuropathologic associations in the nun study and the Honolulu-Asia Aging Study. J Alzheimers Dis 2018;66:1549–58 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Hiya S, Maldonado-Diaz C, Walker JM, et al. Cognitive symptoms progress with limbic-predominant age-related TDP-43 encephalopathy stage and co-occurrence with Alzheimer disease. J Neuropathol Exp Neurol 2023;83:2–10 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Nag S, Yu L, Boyle PA, et al. TDP-43 pathology in anterior temporal pole cortex in aging and Alzheimer’s disease. Acta Neuropathol Commun 2018;6:33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Robinson JL, Richardson H, Xie SX, et al. The development and convergence of co-pathologies in Alzheimer’s disease. Brain 2021;144:953–62 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Nelson PT, Schneider JA, Jicha GA, et al. When Alzheimer’s is LATE: Why does it matter? Ann Neurol 2023;94:211–22 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Nag S, Schneider JA.. Limbic-predominant age-related TDP43 encephalopathy (LATE) neuropathological change in neurodegenerative diseases. Nat Rev Neurol 2023;19:525–41 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Boyle PA, Yu L, Wilson RS, et al. Person-specific contribution of neuropathologies to cognitive loss in old age. Ann Neurol 2018;83:74–83 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Finch CE, Kulminski AM.. The Alzheimer’s disease exposome. Alzheimers Dement 2019;15:1123–32 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Eyre HA, Stirland LE, Jeste DV, et al. Life-course brain health as a determinant of late-life mental health: American Association for Geriatric Psychiatry Expert Panel Recommendations. Am J Geriatr Psychiatry 2023;31:1017–31 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Jaisa-Aad M, Munoz-Castro C, Serrano-Pozo A.. Update on modifiable risk factors for Alzheimer’s disease and related dementias. Curr Opin Neurol 2024;37:166–81 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Melo Dos Santos LS, Trombetta-Lima M, Eggen B, et al. Cellular senescence in brain aging and neurodegeneration. Ageing Res Rev 2024;93:102141. [DOI] [PubMed] [Google Scholar]
- 16. James BD, Wilson RS, Boyle PA, et al. TDP-43 stage, mixed pathologies, and clinical Alzheimer’s-type dementia. Brain 2016;139:2983–93 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Hase Y, Polvikoski TM, Firbank MJ, et al. Small vessel disease pathological changes in neurodegenerative and vascular dementias concomitant with autonomic dysfunction. Brain Pathol 2020;30:191–202 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Mehta RI, Schneider JA.. What is ‘Alzheimer’s disease’? The neuropathological heterogeneity of clinically defined Alzheimer’s dementia. Curr Opin Neurol 2021;34:237–45 [DOI] [PubMed] [Google Scholar]
- 19. Jellinger KA. Recent update on the heterogeneity of the Alzheimer’s disease spectrum. J Neural Transm 2022;129:1–24 [DOI] [PubMed] [Google Scholar]
- 20. Neumann M, Sampathu DM, Kwong LK, et al. Ubiquitinated TDP-43 in frontotemporal lobar degeneration and amyotrophic lateral sclerosis. Science 2006;314:130–3 [DOI] [PubMed] [Google Scholar]
- 21. Geser F, Lee VM, Trojanowski JQ.. Amyotrophic lateral sclerosis and frontotemporal lobar degeneration: A spectrum of TDP-43 proteinopathies. Neuropathology 2010;30:103–12 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Buee L, Bussiere T, Buee-Scherrer V, et al. Tau protein isoforms, phosphorylation and role in neurodegenerative disorders. Brain Res Brain Res Rev 2000;33:95–130 [DOI] [PubMed] [Google Scholar]
- 23. Goedert M. Tau protein and neurodegeneration. Semin Cell Dev Biol 2004;15:45–9 [DOI] [PubMed] [Google Scholar]
- 24. Chornenkyy Y, Fardo DW, Nelson PT.. Tau and TDP-43 proteinopathies: Kindred pathologic cascades and genetic pleiotropy. Lab Invest 2019;99:993–1007 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Trojanowski JQ, Lee VM. “Fatal attractions” of proteins. A comprehensive hypothetical mechanism underlying Alzheimer’s disease and other neurodegenerative disorders. Ann NY Acad Sci 2000;924:62–7 [DOI] [PubMed] [Google Scholar]
- 26. Irwin KE, Jasin P, Braunstein KE, et al. A fluid biomarker reveals loss of TDP-43 splicing repression in presymptomatic ALS-FTD. Nat Med 2024;30:382–93 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Gao J, Wang L, Huntley ML, et al. Pathomechanisms of TDP-43 in neurodegeneration. J Neurochem 2018;10.1111/jnc.14327 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Wood A, Gurfinkel Y, Polain N, et al. Molecular mechanisms underlying TDP-43 pathology in cellular and animal models of ALS and FTLD. Int J Mol Sci 2021;22:4705. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Jiang L, Ngo ST.. Altered TDP-43 structure and function: Key insights into aberrant RNA, mitochondrial, and cellular and systemic metabolism in amyotrophic lateral sclerosis. Metabolites 2022;12:709. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Cheemala A, Kimble AL, Tyburski JD, et al. Loss of endothelial TDP-43 leads to blood brain barrier defects in mouse models of amyotrophic lateral sclerosis and frontotemporal dementia. bioRxiv 2023; 10.1101/2023.12.13.571184 [Preprint] [Google Scholar]
- 31. White MA, Kim E, Duffy A, et al. TDP-43 gains function due to perturbed autoregulation in a Tardbp knock-in mouse model of ALS-FTD. Nat Neurosci 2018;21:552–63 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Chang K, Ling JP, Redding-Ochoa J, et al. Loss of TDP-43 splicing repression occurs early in the aging population and is associated with Alzheimer’s disease neuropathologic changes and cognitive decline. Acta Neuropathol 2023;147:4. [DOI] [PubMed] [Google Scholar]
- 33. Robinson JL, Porta S, Garrett FG, et al. Limbic-predominant age-related TDP-43 encephalopathy differs from frontotemporal lobar degeneration. Brain 2020;143:2844–57 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Young AL, Vogel JW, Robinson JL, et al. Data-driven neuropathological staging and subtyping of TDP-43 proteinopathies. Brain 2023;146:2975–88 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Nelson PT, Lee EB, Cykowski MD, et al. LATE-NC staging in routine neuropathologic diagnosis: An update. Acta Neuropathol 2023;145:159–73 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Teylan MA, Mock C, Gauthreaux K, et al. Differences in symptomatic presentation and cognitive performance among participants with LATE-NC compared to FTLD-TDP. J Neuropathol Exp Neurol 2021;80:1024–32 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Knopman DS, Roberts RO.. Estimating the number of persons with frontotemporal lobar degeneration in the US population. J Mol Neurosci 2011;45:330–5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Coyle-Gilchrist IT, Dick KM, Patterson K, et al. Prevalence, characteristics, and survival of frontotemporal lobar degeneration syndromes. Neurology 2016;86:1736–43 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Buciuc M, Whitwell JL, Baker MC, et al. Old age genetically confirmed frontotemporal lobar degeneration with TDP-43 has limbic predominant TDP-43 deposition. Neuropathol Appl Neurobiol 2021;47:1050–9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Nelson PT. What to do with unusual TDP-43 proteinopathy cases? Neuropathol Appl Neurobiol 2022;48:e12745. [DOI] [PubMed] [Google Scholar]
- 41. Josephs KA, Mackenzie I, Frosch MP, et al. LATE to the PART-y. Brain 2019;142:e47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Nelson PT, Dickson DW, Trojanowski JQ, et al. Reply: LATE to the PART-y. Brain 2019;142:e48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Josephs KA, Murray ME, Whitwell JL, et al. Updated TDP-43 in Alzheimer’s disease staging scheme. Acta Neuropathol 2016;131:571–85 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Carlos AF, Machulda MM, Rutledge MH, et al. Comparison of clinical, genetic, and pathologic features of limbic and diffuse transactive response DNA-binding protein 43 pathology in Alzheimer’s disease neuropathologic spectrum. J Alzheimers Dis 2023;93:1521–35 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Josephs KA, Murray ME, Tosakulwong N, et al. Pathological, imaging and genetic characteristics support the existence of distinct TDP-43 types in non-FTLD brains. Acta Neuropathol 2019;137:227–38 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Cykowski MD, Arumanayagam AS, Powell SZ, et al. Patterns of amygdala region pathology in LATE-NC: Subtypes that differ with regard to TDP-43 histopathology, genetic risk factors, and comorbid pathologies. Acta Neuropathol 2022;143:531–45 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Smith VD, Bachstetter AD, Ighodaro E, et al. Overlapping but distinct TDP-43 and tau pathologic patterns in aged hippocampi. Brain Pathol 2017;28:264–73 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Josephs KA, Koga S, Tosakulwong N, et al. Molecular fragment characteristics and distribution of tangle associated TDP-43 (TATs) and other TDP-43 lesions in Alzheimer’s disease. Free Neuropathol 2023;4:4–22 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Lee EB, Porta S, Michael Baer G, et al. Expansion of the classification of FTLD-TDP: Distinct pathology associated with rapidly progressive frontotemporal degeneration. Acta Neuropathol 2017;134:65–78 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Hu WT, Josephs KA, Knopman DS, et al. Temporal lobar predominance of TDP-43 neuronal cytoplasmic inclusions in Alzheimer disease. Acta Neuropathol 2008;116:215–20 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Josephs KA, Whitwell JL, Knopman DS, et al. Abnormal TDP-43 immunoreactivity in AD modifies clinicopathologic and radiologic phenotype. Neurology 2008;70:1850–7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Karanth S, Nelson PT, Katsumata Y, et al. Prevalence and clinical phenotype of quadruple misfolded proteins in older adults. JAMA Neurol 2020;77:1299–307 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Katsumata Y, Abner EL, Karanth S, et al. Distinct clinicopathologic clusters of persons with TDP-43 proteinopathy. Acta Neuropathol 2020;140:659–74 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Josephs KA, Dickson DW, Tosakulwong N, et al. Rates of hippocampal atrophy and presence of post-mortem TDP-43 in patients with Alzheimer’s disease: A longitudinal retrospective study. Lancet Neurol 2017;16:917–24 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Kapasi A, Yu L, Boyle PA, et al. Limbic-predominant age-related TDP-43 encephalopathy, ADNC pathology, and cognitive decline in aging. Neurology 2020;95:e1951–62 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Karanth SD, Schmitt FA, Nelson PT, et al. Four common late-life cognitive trajectories patterns associate with replicable underlying neuropathologies. J Alzheimers Dis 2021;82:647–59 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Gal J, Chen J, Katsumata Y, et al. Detergent insoluble proteins and inclusion body-like structures immunoreactive for PRKDC/DNA-PK/DNA-PKcs, FTL, NNT, and AIFM1 in the amygdala of cognitively impaired elderly persons. J Neuropathol Exp Neurol 2018;77:21–39 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Power MC, Mormino E, Soldan A, et al. Combined neuropathological pathways account for age-related risk of dementia. Ann Neurol 2018;84:10–22 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Nelson PT, Abner EL, Patel E, et al. The amygdala as a locus of pathologic misfolding in neurodegenerative diseases. J Neuropathol Exp Neurol 2018;77:2–20 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Tome SO, Gawor K, Thal DR.. LATE-NC in Alzheimer’s disease: Molecular aspects and synergies. Brain Pathol 2023;e13213. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Jadhav VS, Stair JG, Eck RJ, et al. Transcriptomic evaluation of tau and TDP-43 synergism shows tauopathy predominance and reveals potential modulating targets. Neurobiol Dis 2024;193:106441. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Tome SO, Gomes LA, Li X, et al. TDP-43 interacts with pathological tau protein in Alzheimer’s disease. Acta Neuropathol 2021;141:795–9 [DOI] [PubMed] [Google Scholar]
- 63. Koga S, Zhou X, Murakami A, et al. Concurrent tau pathologies in frontotemporal lobar degeneration with TDP-43 pathology. Neuropathol Appl Neurobiol 2022;48:e12778. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Smirnov DS, Salmon DP, Galasko D, et al. TDP-43 pathology exacerbates cognitive decline in primary age-related tauopathy. Ann Neurol 2022;92:425–38 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Braak H, Thal DR, Ghebremedhin E, et al. Stages of the pathologic process in Alzheimer disease: Age categories from 1 to 100 years. J Neuropathol Exp Neurol 2011;70:960–9 [DOI] [PubMed] [Google Scholar]
- 66. Lippa CF, Rosso AL, Stutzbach LD, et al. Transactive response DNA-binding protein 43 burden in familial Alzheimer disease and Down syndrome. Arch Neurol 2009;66:1483–8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. McAleese KE, Walker L, Erskine D, et al. TDP-43 pathology in Alzheimer’s disease, dementia with Lewy bodies and ageing. Brain Pathol 2017;27:472–9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. McAleese KE, Walker L, Erskine D, et al. Concomitant LATE-NC in Alzheimer’s disease is not associated with increased tau or amyloid-beta pathological burden. Neuropathol Appl Neurobiol 2020;46:722–34 [DOI] [PubMed] [Google Scholar]
- 69. Crary JF, Trojanowski JQ, Schneider JA, et al. Primary age-related tauopathy (PART): A common pathology associated with human aging. Acta Neuropathol 2014;128:755–66 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Yokota O, Davidson Y, Bigio EH, et al. Phosphorylated TDP-43 pathology and hippocampal sclerosis in progressive supranuclear palsy. Acta Neuropathol 2010;120:55–66 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Koga S, Sanchez-Contreras M, Josephs KA, et al. Distribution and characteristics of transactive response DNA binding protein 43 kDa pathology in progressive supranuclear palsy. Mov Disord 2017;32:246–55 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Besser LM, Kukull WA, Teylan MA, et al. The revised National Alzheimer’s Coordinating Center’s neuropathology form: Available data and new analyses. J Neuropathol Exp Neurol 2018;77:717–26 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Mock C, Teylan M, Beecham G, et al. The utility of the National Alzheimer’s Coordinating Center’s database for the rapid assessment of evolving neuropathologic conditions. Alzheimer Dis Assoc Disord 2020;34:105–11 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Katsumata Y, Wu X, Aung KZ, et al. Pathologic correlates of aging-related tau astrogliopathy: ARTAG is associated with LATE-NC and cerebrovascular pathologies, but not with ADNC. Neurobiol Dis 2024;191:106412. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Nelson PT, Schmitt FA, Lin Y, et al. Hippocampal sclerosis in advanced age: Clinical and pathological features. Brain 2011;134:1506–18 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Farfel JM, Yu L, Boyle PA, et al. Alzheimer’s disease frequency peaks in the tenth decade and is lower afterwards. Acta Neuropathol Commun 2019;7:104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Braak H, Braak E, Bohl J.. Staging of Alzheimer-related cortical destruction. Eur Neurol 1993;33:403–8 [DOI] [PubMed] [Google Scholar]
- 78. Hokkanen SRK, Hunter S, Polvikoski TM, , et al. ; MRC CFAS and CC75C Study Group. Hippocampal sclerosis, hippocampal neuron loss patterns and TDP-43 in the aged population. Brain Pathol 2018;28:548–59 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Tome SO, Vandenberghe R, Ospitalieri S, et al. Distinct molecular patterns of TDP-43 pathology in Alzheimer’s disease: Relationship with clinical phenotypes. Acta Neuropathol Commun 2020;8:61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Zhang X, Sun B, Wang X, et al. Phosphorylated TDP-43 staging of primary age-related tauopathy. Neurosci Bull 2019;35:183–92 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Minogue G, Kawles A, Zouridakis A, et al. Distinct patterns of hippocampal pathology in Alzheimer’s disease with transactive response DNA-binding protein 43. Ann Neurol 2023;94:1036–47 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. Gu J, Chen F, Iqbal K, et al. Transactive response DNA-binding protein 43 (TDP-43) regulates alternative splicing of tau exon 10: Implications for the pathogenesis of tauopathies. J Biol Chem 2017;292:10600–12 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Gu J, Wang W, Miao S, et al. Protein Phosphatase 1 dephosphorylates TDP-43 and suppresses its function in tau exon 10 inclusion. FEBS Lett 2018;592:402–10 [DOI] [PubMed] [Google Scholar]
- 84. Wu R, Zhou D, Shen X, et al. Phosphorylation of trans-active response DNA-binding protein-of 43 kDa promotes its cytoplasmic aggregation and modulates its function in tau mRNA stability and exon 10 alternative splicing. J Neurochem 2021;158:766–78 [DOI] [PubMed] [Google Scholar]
- 85. Forrest SL, Shepherd CE, McCann H, et al. Globular glial tauopathy with a mutation in MAPT and unusual TDP-43 proteinopathy in a patient with behavioural-variant frontotemporal dementia. Acta Neuropathol 2021;141:791–4 [DOI] [PubMed] [Google Scholar]
- 86. Kovacs GG, Ferrer I, Grinberg LT, et al. Aging-related tau astrogliopathy (ARTAG): Harmonized evaluation strategy. Acta Neuropathol 2016;131:87–102 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87. Kovacs GG, Xie SX, Lee EB, et al. Multisite assessment of aging-related tau astrogliopathy (ARTAG). J Neuropathol Exp Neurol 2017;76:605–19 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Ikeda K, Akiyama H, Kondo H, et al. Thorn-shaped astrocytes: Possibly secondarily induced tau-positive glial fibrillary tangles. Acta Neuropathol 1995;90:620–5 [DOI] [PubMed] [Google Scholar]
- 89. Ikeda K, Akiyama H, Arai T, et al. Glial tau pathology in neurodegenerative diseases: Their nature and comparison with neuronal tangles. Neurobiol Aging 1998;19:S85–91 [DOI] [PubMed] [Google Scholar]
- 90. Schultz C, Ghebremedhin E, Del Tredici K, et al. High prevalence of thorn-shaped astrocytes in the aged human medial temporal lobe. Neurobiol Aging 2004;25:397–405 [DOI] [PubMed] [Google Scholar]
- 91. Prater KE, Latimer CS, Jayadev S.. Glial TDP-43 and TDP-43 induced glial pathology, focus on neurodegenerative proteinopathy syndromes. Glia 2022;70:239–55 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Sordo L, Qian T, Bukhari SA, et al. Characterization of hippocampal sclerosis of aging and its association with other neuropathologic changes and cognitive deficits in the oldest-old. Acta Neuropathol 2023;146:415–32 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. Kovacs GG, Lee VM, Trojanowski JQ.. Protein astrogliopathies in human neurodegenerative diseases and aging. Brain Pathol 2017;27:675–90 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Forrest SL, Wagner S, Kim A, et al. Association of glial tau pathology and LATE-NC in the ageing brain. Neurobiol Aging 2022;119:77–88 [DOI] [PubMed] [Google Scholar]
- 95. Yokota O, Miki T, Nakashima-Yasuda H, et al. Amygdala granular fuzzy astrocytes are independently associated with both LATE neuropathologic change and argyrophilic grains: A study of Japanese series with a low to moderate Braak stage. Acta Neuropathol Commun 2023;11:148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. Kovacs GG, Robinson JL, Xie SX, et al. Evaluating the patterns of aging-related tau astrogliopathy unravels novel insights into brain aging and neurodegenerative diseases. J Neuropathol Exp Neurol 2017;76:270–288 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. Forrest SL, Kril JJ, Wagner S, et al. Chronic traumatic encephalopathy (CTE) is absent from a European community-based aging cohort while cortical aging-related tau astrogliopathy (ARTAG) Is Highly Prevalent. J Neuropathol Exp Neurol 2019;78:398–405 [DOI] [PubMed] [Google Scholar]
- 98. Bachstetter AD, Garrett FG, Jicha GA, et al. Space-occupying brain lesions, trauma-related tau astrogliopathy, and ARTAG: A report of two cases and a literature review. Acta Neuropathol Commun 2021;9:49. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. Gelpi E, van der Zee J, Turon Estrada A, et al. TARDBP mutation p.Ile383Val associated with semantic dementia and complex proteinopathy. Neuropathol Appl Neurobiol 2014;40:225–30 [DOI] [PubMed] [Google Scholar]
- 100. Uemura MT, Suh ER, Robinson JL, et al. Abundant copathologies of polyglucosan bodies, frontotemporal lobar degeneration with TDP-43 inclusions and ageing-related tau astrogliopathy in a family with a GBE1 mutation. Neuropathol Appl Neurobiol 2023;49:e12865. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101. Gomez-Tortosa E, Baradaran-Heravi Y, Gonzalez Alvarez V, et al. Presence of tau astrogliopathy in frontotemporal dementia caused by a novel Grn nonsense (Trp2*) mutation. Neurobiol Aging 2019;76:214.e11–5 [DOI] [PubMed] [Google Scholar]
- 102. Dickson DW, Davies P, Bevona C, et al. Hippocampal sclerosis: A common pathological feature of dementia in very old (> or = 80 years of age) humans. Acta Neuropathol 1994;88:212–21 [DOI] [PubMed] [Google Scholar]
- 103. Amador-Ortiz C, Lin WL, Ahmed Z, et al. TDP-43 immunoreactivity in hippocampal sclerosis and Alzheimer’s disease. Ann Neurol 2007;61:435–45 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Corey-Bloom J, Sabbagh MN, Bondi MW, et al. Hippocampal sclerosis contributes to dementia in the elderly. Neurology 1997;48:154–60 [DOI] [PubMed] [Google Scholar]
- 105. Zabar Y, Carson KA, Troncoso JC, et al. Dementia due to hippocampal sclerosis: Clinical features and comparison to Alzheimer’s disease. Neurology 1998;50:A59–60 [Google Scholar]
- 106. Leverenz JB, Agustin CM, Tsuang D, et al. Clinical and neuropathological characteristics of hippocampal sclerosis: A community-based study. Arch Neurol 2002;59:1099–106 [DOI] [PubMed] [Google Scholar]
- 107. Beach TG, Sue L, Scott S, et al. Hippocampal sclerosis dementia with tauopathy. Brain Pathol 2003;13:263–78 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108. Blass DM, Hatanpaa KJ, Brandt J, et al. Dementia in hippocampal sclerosis resembles frontotemporal dementia more than Alzheimer disease. Neurology 2004;63:492–7 [DOI] [PubMed] [Google Scholar]
- 109. Hatanpaa KJ, Blass DM, Pletnikova O, et al. Most cases of dementia with hippocampal sclerosis may represent frontotemporal dementia. Neurology 2004;63:538–42 [DOI] [PubMed] [Google Scholar]
- 110. Zarow C, Sitzer TE, Chui HC.. Understanding hippocampal sclerosis in the elderly: Epidemiology, characterization, and diagnostic issues. Curr Neurol Neurosci Rep 2008;8:363–70 [DOI] [PubMed] [Google Scholar]
- 111. Pao WC, Dickson DW, Crook JE, et al. Hippocampal sclerosis in the elderly: Genetic and pathologic findings, some mimicking Alzheimer disease clinically. Alzheimer Dis Assoc Disord 2011;25:364–8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112. Zarow C, Weiner MW, Ellis WG, et al. Prevalence, laterality, and comorbidity of hippocampal sclerosis in an autopsy sample. Brain Behav 2012;2:435–42 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113. Nelson PT, Smith CD, Abner EL, et al. Hippocampal sclerosis of aging, a prevalent and high-morbidity brain disease. Acta Neuropathol 2013;126:161–77 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114. Hatanpaa KJ, Raisanen JM, Herndon E, et al. Hippocampal sclerosis in dementia, epilepsy, and ischemic injury: Differential vulnerability of hippocampal subfields. J Neuropathol Exp Neurol 2014;73:136–42 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115. Kero M, Raunio A, Polvikoski T, et al. Hippocampal sclerosis in the oldest old: A Finnish population-based study. J Alzheimers Dis 2018;63:263–72 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116. Robinson JL, Corrada MM, Kovacs GG, et al. Non-Alzheimer’s contributions to dementia and cognitive resilience in The 90+ Study. Acta Neuropathol 2018;136:377–88 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117. Gauthreaux KM, Teylan MA, Katsumata Y, et al. Limbic-predominant age-related TDP-43 encephalopathy: Medical and pathologic factors associated with comorbid hippocampal sclerosis. Neurology 2022;98:e1422–33 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118. Montine TJ, Corrada MM, Kawas C, et al. Association of cognition and dementia with neuropathologic changes of Alzheimer disease and other conditions in the oldest-old. Neurology 2022;99:e1067–78 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119. Montine TJ, Phelps CH, Beach TG, Alzheimer’s Association, et al. National Institute on Aging-Alzheimer’s Association guidelines for the neuropathologic assessment of Alzheimer’s disease: A practical approach. Acta Neuropathol 2012;123:1–11 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120. Ighodaro ET, Jicha GA, Schmitt FA, et al. Hippocampal sclerosis of aging can be segmental: Two cases and review of the literature. J Neuropathol Exp Neurol 2015;74:642–52 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121. Kalaria RN, Ferrer I, Love S.. Vascular disease, hypoxia, and related conditions. In: Love S, et al. , eds. Greenfield’s Neuropathology. 9th ed.London: CRC Press; 2015. [Google Scholar]
- 122. Thom M, Sisodiya S, Epilepsy. In: Love S, et al. , eds. Greenfield’s Neuropathology. Vol. 9. New York: CRC Press; 2015:683-739 [Google Scholar]
- 123. Lee EB, Lee VM, Trojanowski JQ, et al. TDP-43 immunoreactivity in anoxic, ischemic and neoplastic lesions of the central nervous system. Acta Neuropathol 2008;115:305–11 [DOI] [PubMed] [Google Scholar]
- 124. Li JX, Nguyen HL, Qian T, et al. ; Alzheimer’s Disease Neuroimaging Initiative. Longitudinal hippocampal atrophy in hippocampal sclerosis of aging. Aging Brain 2023;4:100092. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125. Brenowitz WD, Monsell SE, Schmitt FA, et al. Hippocampal sclerosis of aging is a key Alzheimer’s disease mimic: Clinical-pathologic correlations and comparisons with both Alzheimer’s disease and non-tauopathic frontotemporal lobar degeneration. J Alzheimers Dis 2014;39:691–702 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126. Zarow C, Wang L, Chui HC, et al. MRI shows more severe hippocampal atrophy and shape deformation in hippocampal sclerosis than in Alzheimer’s disease. Int J Alzheimers Dis 2011;2011:483972. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127. Murray ME, Bieniek KF, Banks Greenberg M, et al. Progressive amnestic dementia, hippocampal sclerosis, and mutation in C9ORF72. Acta Neuropathol 2013;126:545–54 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128. Lopez OL, Kofler J, Chang Y, et al. Hippocampal sclerosis, TDP-43, and the duration of the symptoms of dementia of AD patients. Ann Clin Transl Neurol 2020;7:1546–56 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129. Dugan AJ, Nelson PT, Katsumata Y, et al. ; Alzheimer’s Disease Genetics Consortium. Analysis of genes (TMEM106B, GRN, ABCC9, KCNMB2, and APOE) implicated in risk for LATE-NC and hippocampal sclerosis provides pathogenetic insights: A retrospective genetic association study. Acta Neuropathol Commun 2021;9:152. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130. Zamudio F, Loon AR, Smeltzer S, et al. TDP-43 mediated blood-brain barrier permeability and leukocyte infiltration promote neurodegeneration in a low-grade systemic inflammation mouse model. J Neuroinflammation 2020;17:283. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131. Wu LS, Cheng WC, Shen CK.. Targeted depletion of TDP-43 expression in the spinal cord motor neurons leads to the development of amyotrophic lateral sclerosis-like phenotypes in mice. J Biol Chem 2012;287:27335–44 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132. Niedowicz DM, Katsumata Y, Nelson PT.. In severe ADNC, hippocampi with comorbid LATE-NC and hippocampal sclerosis have substantially more astrocytosis than those with LATE-NC or hippocampal sclerosis alone. J Neuropathol Exp Neurol 2023;82:987–94 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133. Neltner JH, Abner EL, Baker S, et al. Arteriolosclerosis that affects multiple brain regions is linked to hippocampal sclerosis of ageing. Brain 2014;137:255–67 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134. Blevins BL, Vinters HV, Love S, et al. Brain arteriolosclerosis. Acta Neuropathol 2020;141:1–24 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135. Hainsworth AH, Markus HS, Schneider JA.. Cerebral small vessel disease, hypertension, and vascular contributions to cognitive impairment and dementia. Hypertension 2024;81:75–86 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136. Arvanitakis Z, Capuano AW, Lamar M, et al. Late-life blood pressure association with cerebrovascular and Alzheimer disease pathology. Neurology 2018;91:e517–25 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137. Skrobot OA, Attems J, Esiri M, et al. Vascular cognitive impairment neuropathology guidelines (VCING): The contribution of cerebrovascular pathology to cognitive impairment. Brain 2016;139:2957–69 [DOI] [PubMed] [Google Scholar]
- 138. Deramecourt V, Slade JY, Oakley AE, et al. Staging and natural history of cerebrovascular pathology in dementia. Neurology 2012;78:1043–50 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139. Craggs LJ, Yamamoto Y, Deramecourt V, et al. Microvascular pathology and morphometrics of sporadic and hereditary small vessel diseases of the brain. Brain Pathol 2014;24:495–509 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140. Kalaria RN. Neuropathological diagnosis of vascular cognitive impairment and vascular dementia with implications for Alzheimer’s disease. Acta Neuropathol 2016;131:659–85 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141. Ighodaro ET, Abner EL, Fardo DW, et al. ; the Alzheimer’s Disease Neuroimaging Initiative (ADNI). Risk factors and global cognitive status related to brain arteriolosclerosis in elderly individuals. J Cereb Blood Flow Metab 2017;37:201–16 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142. Cervos-Navarro J, Gertz HJ, Frydl V.. Cerebral blood vessel changes in old people. Mech Ageing Dev 1987;39:223–31 [DOI] [PubMed] [Google Scholar]
- 143. Netto JP, Iliff J, Stanimirovic D, et al. Neurovascular unit: Basic and clinical imaging with emphasis on advantages of ferumoxytol. Neurosurgery 2018;82:770–80 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144. Lin WL, Castanedes-Casey M, Dickson DW.. Transactivation response DNA-binding protein 43 microvasculopathy in frontotemporal degeneration and familial Lewy body disease. J Neuropathol Exp Neurol 2009;68:1167–76 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145. Hipke K, Pitter B, Hruscha A, et al. Loss of TDP-43 causes ectopic endothelial sprouting and migration defects through increased fibronectin, vcam 1 and integrin alpha4/beta1. Front Cell Dev Biol 2023;11:1169962. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146. Arribas V, Onetti Y, Ramiro-Pareta M, et al. Endothelial TDP-43 controls sprouting angiogenesis and vascular barrier integrity, and its deletion triggers neuroinflammation. JCI Insight 2024;9:e177819. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147. Grinberg LT, Korczyn AD, Heinsen H.. Cerebral amyloid angiopathy impact on endothelium. Exp Gerontol 2012;47:838–42 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148. Nelson PT, Pious NM, Jicha GA, et al. APOE-epsilon2 and APOE-epsilon4 correlate with increased amyloid accumulation in cerebral vasculature. J Neuropathol Exp Neurol 2013;72:708–15 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149. Bennett DA, Schneider JA, Arvanitakis Z, et al. Overview and findings from the religious orders study. Curr Alzheimer Res 2012;9:628–45 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150. Bourassa P, Tremblay C, Schneider JA, et al. Brain mural cell loss in the parietal cortex in Alzheimer’s disease correlates with cognitive decline and TDP-43 pathology. Neuropathol Appl Neurobiol 2020;46:458–77 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151. Carlos AF, Tosakulwong N, Weigand SD, et al. Frequency and distribution of TAR DNA-binding protein 43 (TDP-43) pathology increase linearly with age in a large cohort of older adults with and without dementia. Acta Neuropathol 2022;144:159–60 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152. Nelson PT, Head E, Schmitt FA, et al. Alzheimer’s disease is not “brain aging”: Neuropathological, genetic, and epidemiological human studies. Acta Neuropathol 2011;121:571–87 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153. Ferrer I, Bella R, Serrano MT, et al. Arteriolosclerotic leucoencephalopathy in the elderly and its relation to white matter lesions in Binswanger’s disease, multi-infarct encephalopathy and Alzheimer’s disease. J Neurol Sci 1990;98:37–50 [DOI] [PubMed] [Google Scholar]
- 154. Jellinger KA, Attems J.. Prevalence of dementia disorders in the oldest-old: An autopsy study. Acta Neuropathol 2010;119:421–33 [DOI] [PubMed] [Google Scholar]
- 155. Kalaria RN, Hase Y.. Neurovascular ageing and age-related diseases. Subcell Biochem 2019;91:477–99 [DOI] [PubMed] [Google Scholar]
- 156. Ungvari Z, Tarantini S, Donato AJ, et al. Mechanisms of vascular aging. Circ Res 2018;123:849–67 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157. Mistriotis P, Andreadis ST.. Vascular aging: Molecular mechanisms and potential treatments for vascular rejuvenation. Ageing Res Rev 2017;37:94–116 [DOI] [PubMed] [Google Scholar]
- 158. Tesauro M, Mauriello A, Rovella V, et al. Arterial ageing: From endothelial dysfunction to vascular calcification. J Intern Med 2017;281:471–82 [DOI] [PubMed] [Google Scholar]
- 159. Shindo A, Ishikawa H, Ii Y, et al. Clinical features and experimental models of cerebral small vessel disease. Front Aging Neurosci 2020;12:109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160. Hainsworth AH, Allan SM, Boltze J, et al. Translational models for vascular cognitive impairment: A review including larger species. BMC Med 2017;15:16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161. Walker L, Attems J.. Prevalence of concomitant pathologies in Parkinson’s disease: Implications for prognosis, diagnosis, and insights into common pathogenic mechanisms. J Parkinsons Dis 2024;14:35–52 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162. Uemura MT, Robinson JL, Cousins KAQ, et al. Distinct characteristics of limbic-predominant age-related TDP-43 encephalopathy in Lewy body disease. Acta Neuropathol 2022;143:15–31 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163. McKee AC, Stein TD, Huber BR, et al. Chronic traumatic encephalopathy (CTE): Criteria for neuropathological diagnosis and relationship to repetitive head impacts. Acta Neuropathol 2023;145:371–94 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164. Butler M, Dixon E, Stein TD, et al. Tau pathology in chronic traumatic encephalopathy is primarily neuronal. J Neuropathol Exp Neurol 2022;81:773–80 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165. Nicks R, Clement NF, Alvarez VE, et al. Repetitive head impacts and chronic traumatic encephalopathy are associated with TDP-43 inclusions and hippocampal sclerosis. Acta Neuropathol 2023;145:395–408 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166. Beecham GW, Hamilton K, Naj AC, et al. ; Alzheimer’s Disease Genetics Consortium (ADGC). Genome-wide association meta-analysis of neuropathologic features of Alzheimer’s disease and related dementias. PLoS Genet 2014;10:e1004606. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167. Dickson DW, Baker M, Rademakers R.. Common variant in GRN is a genetic risk factor for hippocampal sclerosis in the elderly. Neurodegener Dis 2010;7:170–4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168. Aoki N, Murray ME, Ogaki K, et al. Hippocampal sclerosis in Lewy body disease is a TDP-43 proteinopathy similar to FTLD-TDP type A. Acta Neuropathol 2015;129:53–64 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169. Nelson PT, Estus S, Abner EL, et al. ; Alzheimer’ Disease Genetic Consortium. ABCC9 gene polymorphism is associated with hippocampal sclerosis of aging pathology. Acta Neuropathol 2014;127:825–43 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170. Nelson PT, Wang WX, Partch AB, et al. Reassessment of risk genotypes (GRN, TMEM106B, and ABCC9 variants) associated with hippocampal sclerosis of aging pathology. J Neuropathol Exp Neurol 2015;74:75–84 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171. Murray ME, Cannon A, Graff-Radford NR, et al. Differential clinicopathologic and genetic features of late-onset amnestic dementias. Acta Neuropathol 2014;128:411–21 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172. Yang HS, Yu L, White CC, et al. Evaluation of TDP-43 proteinopathy and hippocampal sclerosis in relation to APOE epsilon4 haplotype status: A community-based cohort study. Lancet Neurol 2018;17:773–781 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173. Van Deerlin VM, Sleiman PM, Martinez-Lage M, et al. Common variants at 7p21 are associated with frontotemporal lobar degeneration with TDP-43 inclusions. Nat Genet 2010;42:234–9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174. Baker M, Mackenzie IR, Pickering-Brown SM, et al. Mutations in progranulin cause tau-negative frontotemporal dementia linked to chromosome 17. Nature 2006;442:916–9 [DOI] [PubMed] [Google Scholar]
- 175. Boeve BF, Baker M, Dickson DW, et al. Frontotemporal dementia and parkinsonism associated with the IVS1 + 1G->A mutation in progranulin: A clinicopathologic study. Brain 2006;129:3103–14 [DOI] [PubMed] [Google Scholar]
- 176. Cruts M, Gijselinck I, van der Zee J, et al. Null mutations in progranulin cause ubiquitin-positive frontotemporal dementia linked to chromosome 17q21. Nature 2006;442:920–4 [DOI] [PubMed] [Google Scholar]
- 177. Dickson DW, Rademakers R, Nicholson AM.. The TMEM106B locus and TDP-43 pathology in older persons without FTLD. Neurology 2015;85:1354–55 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178. Yu L, De Jager PL, Yang J, et al. The TMEM106B locus and TDP-43 pathology in older persons without FTLD. Neurology 2015;84:927–34 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179. Ren Y, van Blitterswijk M, Allen M, et al. TMEM106B haplotypes have distinct gene expression patterns in aged brain. Mol Neurodegener 2018;13:35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180. Nicholson AM, Rademakers R.. What we know about TMEM106B in neurodegeneration. Acta Neuropathol 2016;132:639–51 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181. Hsiao SJ, Karajannis MA, Diolaiti D, et al. A novel, potentially targetable TMEM106B-BRAF fusion in pleomorphic xanthoastrocytoma. Cold Spring Harb Mol Case Stud 2017;3:a001396. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182. Ou SH, Chalmers ZR, Azada MC, et al. Identification of a novel TMEM106B-ROS1 fusion variant in lung adenocarcinoma by comprehensive genomic profiling. Lung Cancer 2015;88:352–4 [DOI] [PubMed] [Google Scholar]
- 183. Cherry JD, Mez J, Crary JF, et al. Variation in TMEM106B in chronic traumatic encephalopathy. Acta Neuropathol Commun 2018;6:115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184. Gallagher MD, Suh E, Grossman M, et al. TMEM106B is a genetic modifier of frontotemporal lobar degeneration with C9orf72 hexanucleotide repeat expansions. Acta Neuropathol 2014;127:407–18 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185. Rhinn H, Abeliovich A.. Differential aging analysis in human cerebral cortex identifies variants in TMEM106B and GRN that regulate aging phenotypes. Cell Syst 2017;4:404–15.e5 [DOI] [PubMed] [Google Scholar]
- 186. Perneel J, Neumann M, Heeman B, et al. Accumulation of TMEM106B C-terminal fragments in neurodegenerative disease and aging. Acta Neuropathol 2023;145:285–302 [DOI] [PubMed] [Google Scholar]
- 187. Chang A, Xiang X, Wang J, et al. Homotypic fibrillization of TMEM106B across diverse neurodegenerative diseases. Cell 2022;185:1346–55.e15 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188. Perneel J, Rademakers R.. Identification of TMEM106B amyloid fibrils provides an updated view of TMEM106B biology in health and disease. Acta Neuropathol 2022;144:807–19 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189. Neumann M, Perneel J, Cheung S, et al. Limbic-predominant age-related TDP-43 proteinopathy (LATE-NC) is associated with abundant TMEM106B pathology. Acta Neuropathol 2023;146:163–6 [DOI] [PubMed] [Google Scholar]
- 190. C TV, Perneel J, Wynants S, et al. C-terminal TMEM106B fragments in human brain correlate with disease-associated TMEM106B haplotypes. Brain 2023;146:4055–64 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191. Lee JY, Harney D, Kwok J, et al. The major TMEM106B dementia risk allele affects TMEM106B protein levels and myelin lipid homeostasis in the ageing human hippocampus. Mol Neurodegener 2023;18:163. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192. Fardo DW, Katsumata Y, Kauwe JS, et al. ; Alzheimer’s Disease Neuroimaging Initiative. CSF protein changes associated with hippocampal sclerosis risk gene variants highlight impact of GRN/PGRN. Exp Gerontol 2017;90:83–9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193. Ahmed Z, Mackenzie IR, Hutton ML, et al. Progranulin in frontotemporal lobar degeneration and neuroinflammation. J Neuroinflammation 2007;4:7–20 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194. Miller ZA, Rankin KP, Graff-Radford NR, et al. TDP-43 frontotemporal lobar degeneration and autoimmune disease. J Neurol Neurosurg Psychiatry 2013;84:956–62 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195. Paushter DH, Du H, Feng T, et al. The lysosomal function of progranulin, a guardian against neurodegeneration. Acta Neuropathol 2018;136:1–17 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196. Klein ZA, Takahashi H, Ma M, et al. Loss of TMEM106B ameliorates lysosomal and frontotemporal dementia-related phenotypes in progranulin-deficient mice. Neuron 2017;95:281–96.e6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197. Zhou X, Sun L, Brady OA, et al. Elevated TMEM106B levels exaggerate lipofuscin accumulation and lysosomal dysfunction in aged mice with progranulin deficiency. Acta Neuropathol Commun 2017;5:9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198. Chen-Plotkin AS, Unger TL, Gallagher MD, et al. TMEM106B, the risk gene for frontotemporal dementia, is regulated by the microRNA-132/212 cluster and affects progranulin pathways. J Neurosci 2012;32:11213–27 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199. Yang HS, White CC, Klein HU, et al. Genetics of gene expression in the aging human brain reveal TDP-43 proteinopathy pathophysiology. Neuron 2020;107:496–508.e6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200. Schweighauser M, Arseni D, Bacioglu M, et al. Age-dependent formation of TMEM106B amyloid filaments in human brains. Nature 2022;605:310–4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201. Katsumata Y, Fardo DW, Shade LMP, et al. ; Alzheimer’s Disease Genetics Consortium. LATE-NC risk alleles (in TMEM106B, GRN, and ABCC9 genes) among persons with African ancestry. J Neuropathol Exp Neurol 2023;82:760–8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202. Robinson JL, Lee EB, Xie SX, et al. Neurodegenerative disease concomitant proteinopathies are prevalent, age-related and APOE4-associated. Brain 2018;141:2181–93 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203. Wennberg AM, Tosakulwong N, Lesnick TG, et al. Association of apolipoprotein E epsilon4 with transactive response DNA-binding protein 43. JAMA Neurol 2018;75:1347–54 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 204. Farfel JM, Yu L, Buchman AS, et al. Relation of genomic variants for Alzheimer disease dementia to common neuropathologies. Neurology 2016;87:489–96 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205. Walker JM, Richardson TE.. Cognitive resistance to and resilience against multiple comorbid neurodegenerative pathologies and the impact of APOE status. J Neuropathol Exp Neurol 2023;82:110–9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 206. Troncoso JC, Kawas CH, Chang CK, et al. Lack of association of the apoE4 allele with hippocampal sclerosis dementia. Neurosci Lett 1996;204:138–40 [DOI] [PubMed] [Google Scholar]
- 207. Saunders AM, Strittmatter WJ, Schmechel D, et al. Association of apolipoprotein E allele epsilon 4 with late-onset familial and sporadic Alzheimer’s disease. Neurology 1993;43:1467–72 [DOI] [PubMed] [Google Scholar]
- 208. Schmechel DE, Saunders AM, Strittmatter WJ, et al. Increased amyloid beta-peptide deposition in cerebral cortex as a consequence of apolipoprotein E genotype in late-onset Alzheimer disease. Proc Natl Acad Sci USA 1993;90:9649–53 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 209. Reitz C, Cheng R, Rogaeva E, et al. ; Genetic and Environmental Risk in Alzheimer Disease 1 Consortium. Meta-analysis of the association between variants in SORL1 and Alzheimer disease. Arch Neurol 2011;68:99–106 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 210. Miyashita A, Koike A, Jun G, et al. ; Alzheimer Disease Genetics Consortium. SORL1 is genetically associated with late-onset Alzheimer’s disease in Japanese, Koreans and Caucasians. PLoS One 2013;8:e58618. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 211. Rogaeva E, Meng Y, Lee JH, et al. The neuronal sortilin-related receptor SORL1 is genetically associated with Alzheimer disease. Nat Genet 2007;39:168–77 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 212. Benussi L, Longobardi A, Kocoglu C, et al. Investigating the endo-lysosomal system in major neurocognitive disorders due to Alzheimer’s disease, frontotemporal lobar degeneration and lewy body disease: Evidence for SORL1 as a cross-disease gene. Int J Mol Sci 2021;22:13633. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 213. Katsumata Y, Shade LM, Hohman TJ, et al. ; Alzheimer’s Disease Genetics Consortium. Multiple gene variants linked to Alzheimer’s-type clinical dementia via GWAS are also associated with non-Alzheimer’s neuropathologic entities. Neurobiol Dis 2022;174:105880. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 214. Fazeli E, Child DD, Bucks SA, et al. A familial missense variant in the Alzheimer’s disease gene SORL1 impairs its maturation and endosomal sorting. Acta Neuropathol 2024;147:20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 215. Hokkanen SRK, Kero M, Kaivola K, et al. ; EClipSE Collaboration. Putative risk alleles for LATE-NC with hippocampal sclerosis in population-representative autopsy cohorts. Brain Pathol 2020;30:364–72 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 216. Nho K, Saykin AJ, Nelson PT, et al. ; Disease Neuroimaging Initiative As. Hippocampal sclerosis of aging, a common Alzheimer’s disease ‘mimic’: Risk genotypes are associated with brain atrophy outside the temporal lobe. J Alzheimers Dis 2016;52:373–83 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 217. Nelson PT, Jicha GA, Wang WX, et al. ABCC9/SUR2 in the brain: Implications for hippocampal sclerosis of aging and a potential therapeutic target. Ageing Res Rev 2015;24:111–25 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 218. Nichols CG, Singh GK, Grange DK.. KATP channels and cardiovascular disease: Suddenly a syndrome. Circ Res 2013;112:1059–72 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 219. Aggarwal S, Randhawa PK, Singh N, et al. Role of ATP-sensitive potassium channels in remote ischemic preconditioning induced tissue protection. J Cardiovasc Pharmacol Ther 2017;22:467–75 [DOI] [PubMed] [Google Scholar]
- 220. Nelson PT, Katsumata Y, Nho K, et al. ; Alzheimer’s Disease Neuroimaging Initiative (ADNI). Genomics and CSF analyses implicate thyroid hormone in hippocampal sclerosis of aging. Acta Neuropathol 2016;132:841–58 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 221. Bondjers C, He L, Takemoto M, et al. Microarray analysis of blood microvessels from PDGF-B and PDGF-Rbeta mutant mice identifies novel markers for brain pericytes. Faseb J 2006;20:1703–5 [DOI] [PubMed] [Google Scholar]
- 222. Kirkwood PM, Gibson DA, Smith JR, et al. Single-cell RNA sequencing redefines the mesenchymal cell landscape of mouse endometrium. Faseb J 2021;35:e21285. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 223. Jellinger KA. The pathology of “vascular dementia”: A critical update. J Alzheimers Dis 2008;14:107–23 [DOI] [PubMed] [Google Scholar]
- 224. Ferrer I, Vidal N.. Neuropathology of cerebrovascular diseases. Handb Clin Neurol 2017;145:79–114 [DOI] [PubMed] [Google Scholar]
- 225. Kośla K, Kałuzińska Ż, Bednarek AK.. The WWOX gene in brain development and pathology. Exp Biol Med (Maywood) 2020;245:1122–9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 226. Kunkle BW, Grenier-Boley B, Sims R, et al. ; Genetic and Environmental Risk in AD/Defining Genetic, Polygenic and Environmental Risk for Alzheimer’s Disease Consortium (GERAD/PERADES). Genetic meta-analysis of diagnosed Alzheimer’s disease identifies new risk loci and implicates Abeta, tau, immunity and lipid processing. Nat Genet 2019;51:414–30 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 227. Wightman DP, Jansen IE, Savage JE, et al. ; 23andMe Research Team. A genome-wide association study with 1,126,563 individuals identifies new risk loci for Alzheimer’s disease. Nat Genet 2021;53:1276–82 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 228. Bellenguez C, Kucukali F, Jansen IE, et al. ; CHARGE. New insights into the genetic etiology of Alzheimer’s disease and related dementias. Nat Genet 2022;54:412–36 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 229. Dugan AJ, Nelson PT, Katsumata Y, et al. ; Alzheimer’s Disease Genetics Consortium. Association between WWOX/MAF variants and dementia-related neuropathologic endophenotypes. Neurobiol Aging 2022;111:95–106 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 230. Song Y, Lin F, Ye CH, et al. Rare, low-frequency and common coding variants of ARHGEF28 gene and their association with sporadic amyotrophic lateral sclerosis. Neurobiol Aging 2020;87:138.e1–e6 [DOI] [PubMed] [Google Scholar]
- 231. Ma Y, Tang L, Chen L, et al. ARHGEF28 gene exon 6/intron 6 junction mutations in Chinese amyotrophic lateral sclerosis cohort. Amyotroph Lateral Scler Frontotemporal Degener 2014;15:309–11 [DOI] [PubMed] [Google Scholar]
- 232. Droppelmann CA, Keller BA, Campos-Melo D, et al. Rho guanine nucleotide exchange factor is an NFL mRNA destabilizing factor that forms cytoplasmic inclusions in amyotrophic lateral sclerosis. Neurobiol Aging 2013;34:248–62 [DOI] [PubMed] [Google Scholar]
- 233. Keller BA, Volkening K, Droppelmann CA, et al. Co-aggregation of RNA binding proteins in ALS spinal motor neurons: Evidence of a common pathogenic mechanism. Acta Neuropathol 2012;124:733–47 [DOI] [PubMed] [Google Scholar]
- 234. Katsumata Y, Fardo DW, Shade LMP, et al. ; National Alzheimer’s Coordinating Center. Genetic associations with dementia-related proteinopathy: Application of item response theory. Alzheimers Dement 2024. doi: 10.1002/alz.13741 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 235. Katsumata Y, Fardo DW, Kukull WA, et al. Dichotomous scoring of TDP-43 proteinopathy from specific brain regions in 27 academic research centers: Associations with Alzheimer’s disease and cerebrovascular disease pathologies. Acta Neuropathol Commun 2018;6:142. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 236. Agrawal S, Yu L, Kapasi A, et al. Limbic-predominant age-related TDP-43 encephalopathy neuropathologic change and microvascular pathologies in community-dwelling older persons. Brain Pathol 2021;31:e12939. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 237. Besser LM, Teylan MA, Nelson PT.. Limbic predominant age-related TDP-43 encephalopathy (LATE): Clinical and neuropathological associations. J Neuropathol Exp Neurol 2020;79:305–13 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 238. Harrison WT, Lusk JB, Liu B, et al. Limbic-predominant age-related TDP-43 encephalopathy neuropathological change (LATE-NC) is independently associated with dementia and strongly associated with arteriolosclerosis in the oldest-old. Acta Neuropathol 2021;142:917–9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 239. Sieben A, Van Langenhove T, Vermeiren Y, et al. Hippocampal sclerosis in frontotemporal dementia: When vascular pathology meets neurodegeneration. J Neuropathol Exp Neurol 2021;80:313–24 [DOI] [PubMed] [Google Scholar]
- 240. Wang SJ, Guo Y, Ervin JF, et al. Neuropathological associations of limbic-predominant age-related TDP-43 encephalopathy neuropathological change (LATE-NC) differ between the oldest-old and younger-old. Acta Neuropathol 2022;144:45–57 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 241. Nichols E, Merrick R, Hay SI, et al. The prevalence, correlation, and co-occurrence of neuropathology in old age: Harmonisation of 12 measures across six community-based autopsy studies of dementia. Lancet Healthy Longev 2023;4:e115–25 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 242. Vass R, Ashbridge E, Geser F, et al. Risk genotypes at TMEM106B are associated with cognitive impairment in amyotrophic lateral sclerosis. Acta Neuropathol 2010;121:373–80 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 243. Rutherford NJ, Carrasquillo MM, Li M, et al. TMEM106B risk variant is implicated in the pathologic presentation of Alzheimer disease. Neurology 2012;79:717–8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 244. Gass J, Cannon A, Mackenzie IR, et al. Mutations in progranulin are a major cause of ubiquitin-positive frontotemporal lobar degeneration. Hum Mol Genet 2006;15:2988–3001 [DOI] [PubMed] [Google Scholar]
- 245. van Swieten JC, Heutink P.. Mutations in progranulin (GRN) within the spectrum of clinical and pathological phenotypes of frontotemporal dementia. Lancet Neurol 2008;7:965–74 [DOI] [PubMed] [Google Scholar]
- 246. Corder EH, Saunders AM, Strittmatter WJ, et al. Gene dose of apolipoprotein E type 4 allele and the risk of Alzheimer’s disease in late onset families. Science 1993;261:921–3 [DOI] [PubMed] [Google Scholar]
- 247. Efthymiou S, Scala M, Nagaraj V, et al. Novel loss-of-function variants expand ABCC9-related intellectual disability and myopathy syndrome. Brain 2024;awae010.doi: 10.1093/brain/awae010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 248. Yu M, Guo L, Li N, et al. Overexpression of Kcnmb2 in dorsal CA1 of offspring mice rescues hippocampal dysfunction caused by a methyl donor-rich paternal diet. Front Cell Neurosci 2018;12:360. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 249. Piard J, Hawkes L, Milh M, et al. ; DDD Study. The phenotypic spectrum of WWOX-related disorders: 20 additional cases of WOREE syndrome and review of the literature. Genet Med 2019;21:1308–18 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 250. Neumann M, Kwong LK, Lee EB, et al. Phosphorylation of S409/410 of TDP-43 is a consistent feature in all sporadic and familial forms of TDP-43 proteinopathies. Acta Neuropathol 2009;117:137–49 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 251. Nelson RS, Abner EL, Jicha GA, et al. Neurodegenerative pathologies associated with behavioral and psychological symptoms of dementia in a community-based autopsy cohort. Acta Neuropathol Commun 2023;11:89. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 252. Gauthreaux K, Kukull WA, Nelson KB, et al. Different cohort, disparate results: Selection bias is a key factor in autopsy cohorts. Alzheimers Dement 2024;20:266–77 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 253. Schmitt FA, Nelson PT, Abner E, et al. University of Kentucky Sanders-Brown healthy brain aging volunteers: Donor characteristics, procedures, and neuropathology. Curr Alzheimer Res 2012;9:724–33 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 254. Greenberg SG, Davies P, Schein JD, et al. Hydrofluoric acid-treated tau PHF proteins display the same biochemical properties as normal tau. J Biol Chem 1992;267:564–9 [PubMed] [Google Scholar]
- 255. Wilson AC, Dugger BN, Dickson DW, et al. TDP-43 in aging and Alzheimer’s disease—A review. Int J Clin Exp Pathol 2011;4:147–55 [PMC free article] [PubMed] [Google Scholar]





