Skip to main content
Erschienen in: Fluids and Barriers of the CNS 1/2022

Open Access 01.12.2022 | Review

Expression and functions of adenylyl cyclases in the CNS

verfasst von: Karan Devasani, Yao Yao

Erschienen in: Fluids and Barriers of the CNS | Ausgabe 1/2022

Abstract

Adenylyl cyclases (ADCYs), by generating second messenger cAMP, play important roles in various cellular processes. Their expression, regulation and functions in the CNS, however, remain largely unknown. In this review, we first introduce the classification and structure of ADCYs, followed by a discussion of the regulation of mammalian ADCYs (ADCY1-10). Next, the expression and function of each mammalian ADCY isoform are summarized in a region/cell-specific manner. Furthermore, the effects of GPCR-ADCY signaling on blood–brain barrier (BBB) integrity are reviewed. Last, current challenges and future directions are discussed. We aim to provide a succinct review on ADCYs to foster new research in the future.
Hinweise

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
ADCYs
Adenylyl cyclases
ARs
Adenosine receptors
BBB
Blood–brain barrier
cAMP
Cyclic adenosine 3′,5′-monophosphate (cAMP)
CaMKII
Calcium/calmodulin-dependent protein kinase II
CaMKIV
Calcium/calmodulin-dependent protein kinase IV
C5aR
Complement C5a receptor
CBs
Cannabinoid receptors
GLP1R
Glucagon-like peptide-1 receptor
GPCRs
G protein-coupled receptors
GPER-1
G protein-coupled estrogen receptor 1
GWAS
Genome wide analysis studies
HCAR1
Hydrocarboxylic acid receptor 1
LTP
Long-term potentiation
LPARs
Lysophosphatidic acid receptors
OPC
Oligodendrocyte’s precursor cells
PKA
Protein kinase A
PKC
Protein kinase C
RGS2
Regulator of G protein signaling 2
SSTRs
Somatostatin receptors
S1PRs
Sphingosine 1-phosphate receptors

Background

One important mechanism that cells use to sense their environment is via receptor-mediated signaling. Specifically, environmental signals, such as chemokines and neurotransmitters, bind to receptors at plasma membrane and activate key intracellular signaling molecules (e.g., second messengers), transferring information from outside to inside. One ubiquitous second messenger in various cell types is cyclic adenosine 3′,5′-monophosphate (cAMP), which can act via either a kinase-dependent manner to induce protein phosphorylation or a kinase-independent manner to induce protein–protein interactions [1]. cAMP plays a pivotal role in a variety of fundamental cellular processes [2], and thus its level needs to be tightly regulated. Adenylyl cyclases (ADCYs) catalyze the production of cAMP from ATP, while phosphodiesterases (PDEs) degrade cAMP to 5′-AMP [3, 4]. This review summarizes the structure, regulation, expression and functions of ADCYs in the CNS.

Classification of ADCYs

ADCYs are grouped into six different classes (class I-VI) based on their structural and domain organizations [5]. Class I is the gamma-proteobacterial type found mainly in gram-negative bacteria, such as Escherichia coli [6, 7]. Class II exists in pathogens that secrete toxin proteins, including Bordetella pertussis and Bacillus anthracis [8]. Class III is the universal or ancestral class of ADCYs found in both bacteria and eukaryotes. Since many of the class III ADCYs have been identified in higher eukaryotes and most thoroughly studied in mammals, they are also known as mammalian ADCYs. Class IV has been identified in Yersinia pestis and in ruminal bacteria Aeromonas hydrophila [5]. Class V and class VI are found in anaerobic bacterium Prevotella ruminicola and nitrogen fixing bacterium Rhizobium etli, respectively [9, 10]. The last two classes of ADCYs have not yet been structurally characterized. In this review, we focus on class III/mammalian ADCYs.

Mammalian ADCYs

Mammalian ADCYs have ten isoforms: nine transmembrane ADCYs (ADCY1-9) and one soluble ADCY (sADCY/ADCY10). All transmembrane ADCYs have a similar structure, but are different in their length and sequence at amino acids 1080–1353 [11] (Table 1). They consist of two discrete membrane-spanning (M1 and M2) domains with each containing six transmembrane alpha-helices, a single N-terminal cytosolic domain, and two cytoplasmic (C1 and C2) domains (Fig. 1). The C1 domain lies between two transmembrane domains, while the C2 domain is at the large C-terminus of the protein. These C1/C2 domains are subdivided into C1a/C2a and C1b/C2b subdomains (Fig. 1). The C1a and C2a subdomains are the catalytic site and highly conserved: they are structurally identical and homologous among all 9 transmembrane isoforms, while the C1b and C2b subdomains are the regulatory site [11, 12]. Unlike ADCY1-9, ADCY10 does not have clearly defined transmembrane domains (Table 1) [13]. Its catalytic domain is more related to bicarbonate-sensing ADCY from cyanobacteria than that of ADCY1-9.
Table 1
Chromosomal location, structure and length of mammalian ADCYs
Gene name
Chromosome (human)
Chromosome (mouse)
Structure
Length (human)
Length (mouse)
ADCY1
7p12
11A2
https://static-content.springer.com/image/art%3A10.1186%2Fs12987-022-00322-2/MediaObjects/12987_2022_322_Figa_HTML.gif
1119
1118
ADCY2
5p15
13C1
https://static-content.springer.com/image/art%3A10.1186%2Fs12987-022-00322-2/MediaObjects/12987_2022_322_Figb_HTML.gif
1091
1090
ADCY3
2p22-24
12A-B
https://static-content.springer.com/image/art%3A10.1186%2Fs12987-022-00322-2/MediaObjects/12987_2022_322_Figc_HTML.gif
1144
1145
ADCY4
14q11.2
14D3
https://static-content.springer.com/image/art%3A10.1186%2Fs12987-022-00322-2/MediaObjects/12987_2022_322_Figd_HTML.gif
1077
1077
ADCY5
3q13.2-q21
16B5
https://static-content.springer.com/image/art%3A10.1186%2Fs12987-022-00322-2/MediaObjects/12987_2022_322_Fige_HTML.gif
1261
1262
ADCY6
12q12-13
15F
https://static-content.springer.com/image/art%3A10.1186%2Fs12987-022-00322-2/MediaObjects/12987_2022_322_Figf_HTML.gif
1168
1165
ADCY7
16q12-13
8C3-D
https://static-content.springer.com/image/art%3A10.1186%2Fs12987-022-00322-2/MediaObjects/12987_2022_322_Figg_HTML.gif
1080
1099
ADCY8
8q24
15
https://static-content.springer.com/image/art%3A10.1186%2Fs12987-022-00322-2/MediaObjects/12987_2022_322_Figh_HTML.gif
1251
1249
ADCY9
16p13.3
16B1
https://static-content.springer.com/image/art%3A10.1186%2Fs12987-022-00322-2/MediaObjects/12987_2022_322_Figi_HTML.gif
1353
1353
ADCY10
1q24.2
1
https://static-content.springer.com/image/art%3A10.1186%2Fs12987-022-00322-2/MediaObjects/12987_2022_322_Figj_HTML.gif
1610
1614
Genetic studies have shown that ADCY genes are not clustered in the genome: each isoform is coded by a gene localized on a different chromosome [14, 15] (Table 1). This enables isoform-specific regulation of ADCYs.

Regulation of ADCYs

ADCY activity is mainly regulated by G protein-coupled receptors (GPCRs). G protein is a heterotrimer containing α, β and γ subunits. Based on its function, Gα subunit is divided into four major categories: Gαs, Gαi/o, Gαq/11 and Gα12/13 (Fig. 2). Currently, there are five β and 11 γ subunits, which through forming highly active βγ heterodimers participate in the regulation of various biological processes [16]. Upon ligand binding, GPCRs change their confirmation replace GDP with GTP on Gα subunit, leading to dissociation of Gβγ complex. Then Gα and Gβγ independently activate downstream signaling cascades. The system returns to the resting state when ligands are released from GPCRs, which causes hydrolysis of GTP to GDP on Gα subunit and subsequent reassociation of Gβγ with Gα to form heterotrimers.
The effect of GPCRs on ADCY activity is dependent on the type of G protein (Fig. 2). Specifically, Gαs directly activates ADCYs, increasing cAMP production. Gαi/o directly activate or inhibit ADCY activity in an ADCY isoform-specific manner. Gαq/11 indirectly regulates ADCY activity via protein kinase C (PKC) and/or Ca2+, which can activate or inhibit ADCY activity. Gα12/13 does not seem to be involved in the regulation of ADCY activity. The dissociated Gβγ complex can either activate or inhibit ADCY activity depending on its Gα partner and ADCY isoforms. It should be noted that Gβγ can also be generated via modulatory protein GoLoco without activation of GPCRs. Similarly, Gβγ generated this way may have stimulatory or inhibitory effect on ADCY activity.
Based on their signaling properties, transmembrane ADCYs are further divided into four groups (Group I-IV). Group I is composed of ADCY1, ADCY3 and ADCY8; Group II includes ADCY2, ADCY4 and ADCY7; Group III consists of ADCY5 and ADCY6; and Group IV contains ADCY9 only. The unique features of these four groups of transmembrane ADCYs and soluble ADCY are discussed below and summarized in Fig. 3.

Group I

Group I ADCYs are activated by Gαs and Ca2+/calmodulin, and inhibited by Gαi/o and Gβγ. It should be noted that these ADCYs have different sensitivity to the stimuli. For example, ADCY3 and ADCY8 are five-fold less sensitive to Ca2+ than ADCY1 [17]. In addition, the activity of group I ADCYs can also be regulated by phosphorylation. For instance, PKCα activates ADCY1 and ADCY3; whereas calcium/calmodulin-dependent protein kinase IV (CaMKIV) inactivates ADCY1, CaMKII and regulator of G protein signaling 2 (RGS2) inactivate ADCY3, and PKA inactivates ADCY8 [18].

Group II

Group II ADCYs are activated by both Gαs and Gβγ with a higher potency on Gαs, but insensitive to Ca2+/calmodulin. Although group II ADCYs are not inhibited by Gαi/o, it is assumed that Gβγ released from Gαi/o stimulation can synergistically stimulate them [11, 15, 19]. In addition, PKC regulates group II ADCYs in an isoform-specific manner. Specifically, PKCα activates ADCY2 but inactivates ADCY4, and PKCδ activates ADCY7 [18].

Group III

Group III ADCYs are activated by Gαs and Gβγ, but inhibited by Gαi/o and free Ca2+. Although most ADCYs are inhibited by high (non-physiological) concentration of Ca2+, ADCY5 and ADCY6 are inhibited by Ca2+ at sub-micromolar level [17], which may have important physiological implications. In addition, ADCY5 is also activated by PKCα/δ and inhibited by RGS2, PKA and Rica8; whereas ADCY6 is inhibited by RGS2, PKA and PKCδ/ε [18].

Group IV

Group IV ADCY is activated solely by Gαs. Unlike other transmembrane ADCYs, ADCY9 is insensitive to forskolin due to the lack of a key leucine residue in the catalytic cleft [20, 21]. Although ADCY9 is not regulated by Gαi/o or Gβγ, calcineurin and PKC are able to inhibit its activity [18].

sADCY/ADCY10

Unlike transmembrane ADCYs, ADCY10 is not associated with the membrane: it is diffusely distributed in the cytoplasm and nucleus [22]. Although ADCY10 is not regulated by G protein and insensitive to forskolin, it is activated by Ca2+ and bicarbonates [22, 23]. The negative regulators of ADCY10 have yet to be identified and the functions of ADCY10 remain largely unknown. It has been reported that nuclear ADCY10 is involved in gene regulation [24], while mitochondrial ADCY10 moderates oxidative phosphorylation in response to CO2/HCO3 generated by citric acid cycle [25, 26].

Expression and functions of ADCYs in the CNS

ADCYs are found in almost all cells and different cell types express distinct ADCY isoforms. The expression patterns of ADCY isoforms are mainly obtained from RNA-sequencing analyses (at the mRNA level). Currently, ADCY expression profiles at the protein level are limited, possibly due to the lack of isoform-specific antibodies.
ADCYs play a variety of important functions in the CNS, ranging from learning/memory to movements. Abnormal ADCY expression is found in and associated with many neurological disorders, including Alzheimer’s disease and depressive disorders. For example, loss of ADCY1 leads to impaired synaptic plasticity and deficits in spatial learning [27], while overexpression of ADCY1 in the forebrain enhances recognition and memory [28]. ADCY8 exerts similar functions as ADCY1 [2935] and is associated with bipolar disorder [36] and post-traumatic stress disorder [37] in humans. ADCY3 is involved in olfactory-dependent learning and associated with major depressive disorder in humans [38, 39]. Loss of ADCY5 results in Parkinson-like motor dysfunction and locomotor impairment [40]. ADCY7 is linked to familial major depression in both mice and humans [41, 42]. It should be noted that not all ADCYs are well studied. The functions of ADCY2, ADCY4, ADCY6, ADCY9 and ADCY10 in the CNS remain largely unknown. Here, we discuss and summarize the expression (Table 2) and function (Table 3) of each ADCY isoform in the CNS.
Table 2
Region/cell-specific expression of ADCYs in the CNS
Isoforms
Sites of expression
Cell types
ADCY1
Piriform cortex#, CA1-CA2 of hippocampus#, dentate gyrus#, striatum#, amygdala^, thalamus#, and cerebellum#, cerebral cortex^, and olfactory bulb layers*
Neurons#, oligodendrocytes^, microglia^, astrocytes^, fibroblasts* and endothelial cells*
ADCY2
Piriform cortex#, CA1 of hippocampus#, dentate gyrus#, striatum#, CA2-CA3 of hippocampus^, thalamus^, hypothalamus^, olfactory bulb layers^, cerebral cortex^, amygdala*, and cerebellum*
Neurons#, astrocytes#, fibroblasts#, smooth muscle cells#, oligodendrocytes^, microglia*, endothelial cells*, pericytes*
ADCY3
CA1-CA3 of hippocampus#, dentate gyrus#, cerebral cortex^, striatum^, amygdala^, thalamus^, hypothalamus^, cerebellum^, olfactory bulb layers*, piriform cortex*, and choroid plexus
Neurons#, astrocytes#, fibroblasts#, pericytes#, oligodendrocytes^, smooth muscle cells*, microglia*, endothelial cells*, and choroid plexus epithelial cells
ADCY4
Hippocampus* and olfactory epithelium*
Endothelial cells#, neurons*, astrocytes*, fibroblasts*, smooth muscle cells*, oligodendrocytes*, microglia*, and pericytes*
ADCY5
Striatum#, hypothalamus^, olfactory bulb layers^, cerebral cortex*, piriform cortex*, amygdala*, CA1-CA3 of hippocampus*, dentate gyrus*, and thalamus*
Neurons#, oligodendrocytes#, fibroblasts#, smooth muscle cells#, astrocytes*, microglia*, endothelial cells*, and pericytes*
ADCY6
Piriform cortex#, amygdala#, CA1-CA3 of hippocampus#, dentate gyrus#, hypothalamus#, cerebellum#, choroid plexus#, olfactory bulb layers^, cerebral cortex^, striatum^, and thalamus^
Neurons#, astrocytes#, fibroblasts#, smooth muscle cells#, oligodendrocytes#, endothelial cells#, pericytes#, and microglia*
ADCY7
Thalamus^, and hypothalamus^, cerebral cortex*, amygdala*, corpus callosum*, cerebellum*, and olfactory system*
Fibroblasts#, smooth muscle cells#, microglia#, neurons*, astrocytes*, oligodendrocytes*, endothelial cells*, and pericytes*
ADCY8
Piriform cortex#, CA1-CA2 of hippocampus#, dentate gyrus#, thalamus#, hypothalamus#, cerebellum#, olfactory bulb#, cerebral cortex*, and amygdala*
Neurons#, astrocytes#, fibroblasts*, smooth muscle cells*, oligodendrocytes*, microglia*, and endothelial cells*
ADCY9
Olfactory bulb#, cerebral cortex#, piriform cortex#, CA1-CA3 of hippocampus#, dentate gyrus#, cerebellum#, amygdala^, thalamus*, and hypothalamus*
Neurons#, astrocytes#, fibroblasts#, smooth muscle cells#, oligodendrocytes#, microglia#, endothelial cells#, and pericytes#
ADCY10
Visual cortex*, hippocampus*, and cerebellum*, and choroid plexus
Neurons*, astrocytes*, fibroblasts*, smooth muscle cells*, oligodendrocytes*, microglia*, endothelial cells*, pericytes*, and choroid plexus epithelial cells
#High expression, ^Medium expression, *Low expression
Table 3
ADCY functions and their associated diseases
Isoforms
Knockout/knockdown
Overexpression
Potential functions
Associated diseases
References
ADCY1
Yesa
Yes
Learning, memory, LTP, synaptic plasticity, drug dependency, nociception, and pain
Anxiety-like behavior, hearing impairment, sleep deprivation, schizophrenia, bipolar disorder, and autism
[28, 50, 53, 5557]
ADCY2
Synaptic plasticity and neuropsychiatric functions
Bipolar disorder, anxiety, stress-like disorders, Lesch-Nyhan disease, and schizophrenia
[6366]
ADCY3
Yesa,b
Odorant signaling, learning, and memory
Obesity, depression, and inflammatory bowel disease
[19, 38, 68, 7072, 7479, 190]
ADCY4
Yesb
[81]
ADCY5
Yesa,c
Yes
Learning, memory, synaptic plasticity, and extrapyramidal motor functions
Familial dyskinesia and facial myokymia, anxiety, depressive-like disorder, and movement disorders
[8385, 89, 9295]
ADCY6
Yesa
Yes
Metabolic functions and fluid homeostasis in kidney
Axoglial diseases and lethal congenital contracture syndrome
[97, 98, 100, 101]
ADCY7
Yesc
Yes
Alcohol dependency
Depression-like disorder, inflammatory bowel disease, Crohn’s disease, ulcerative colitis, and autoimmune diseases
[42, 108, 109]
ADCY8
Yesa
Yes
Learning, memory, LTP, synaptic plasticity, nociception, and pain
Dissociative amnesia, post-traumatic stress disorder, depression, and bipolar disorder
[29, 33, 37, 52, 112, 113]
ADCY9
Yesa
Yes
Learning and memory, immunological functions, and cardioprotective
Asthma, mood disorders, and bipolar disorder
[116, 119121]
ADCY10
Yesa
Synaptic plasticity, learning and memory, ocular dominance plasticity, and fertilization
Infertility and absorptive hypercalciuria
[126, 132, 133]
aGlobal knockout, bConditional knockout, cKnockdown

ADCY1

Expression

ADCY1 mRNA is transiently expressed in trigeminal nerve nuclei, striatum, dorsal thalamus, hippocampal interneurons, retinal ganglion cells and cerebellar Purkinje cells in early postnatal life [43]. In adulthood, however, its expression is confined to olfactory bulb, pineal gland, cortex, dentate gyrus, various thalamic nuclei, CA1 region of hippocampus and granule cells of the cerebellum [43, 44]. Bulk RNA-sequencing analysis showed that ADCY1 expression was high in neurons and moderate in oligodendrocytes, microglia and astrocytes [45]. Single-cell RNA-sequencing study demonstrated high and low levels of ADCY1 in vascular fibroblast-like cells and endothelial cells, respectively [46].

Function

As a Ca2+/calmodulin-sensitive ADCY, ADCY1 exerts important functions in neuronal development, a process that is critically regulated by calcium [47]. Correlation studies showed that hippocampal expression of ADCY1 was reduced during aging and increased during the acquisition of spatial learning [48, 49], highlighting a possible role of ADCY1 in learning/memory. In addition, ADCY1-null mice exhibited reduced long-term potentiation (LTP) in hippocampal mossy fibers, impaired cerebellar LTP, and higher threshold to inflammatory and chronic pain [5052]. Consistent with these findings, overexpression of ADCY1 in forebrain led to elevated LTP, improved memory and decreased social ability via increased extracellular signal-related kinase (ERK1/2) [28]. These results suggest that ADCY1 regulates learning/memory, LTP and nociception.
Interestingly, FMR1-null mice, a rodent model of fragile X syndrome, exhibited increased ADCY1 expression and ADCY1-overexpression-like phenotype, including enhanced autism-related behaviors and increased ERK1/2 activity [53]. Loss of ADCY1 in FMR1-null background, however, reversed these changes [53]. These findings suggest that FMR1 inhibits ADCY1 expression, and that loss of FMR1-dependent suppression of ADCY1 is a cause for eccentric neuronal signaling in fragile X syndrome.
In humans, it has been reported that a nonsense mutation in ADCY1 gene causes hearing impairment, deafness and loss of hair cell function [54, 55]. In addition, genome wide analysis studies (GWAS) showed that ADCY1 polymorphism was associated with sleep deprivation, schizophrenia and bipolar disorder [56, 57].

ADCY2

Expression

ADCY2 mRNA is highly expressed in piriform cortex, hippocampus, dentate gyrus, striatum and thalamus [58]. Bulk RNA-sequencing study found that ADCY2 was mainly expressed in astrocytes, neurons and oligodendrocytes precursor cells (OPCs); and to a lesser extent in oligodendrocytes and microglia [45]. Single-cell RNA-sequencing analysis showed that ADCY2 was expressed at high levels in astrocytes, vascular fibroblast-like cells and smooth muscle cells, and at low levels in endothelial cells and pericytes [46]. At the protein level, ADCY2 expression has been found in mouse hippocampus [59], indicating a possible role in synaptic plasticity.

Function

The function of ADCY2 remains largely unknown. A correlation study found that ADCY2 was down-regulated throughout the hippocampus during the acquisition of spatial learning in mice [49], suggesting that ADCY2 may be involved in spatial learning and memory. In addition, it has been reported that P19 cells (embryonic carcinoma cells) up-regulate ADCY2 during neuronal [60] and mesodermal [61] differentiation, highlighting a possible role of ADCY2 in cell differentiation during development.
A recent GWAS study revealed that ADCY2 polymorphism was associated with neuropsychiatric disorders, including bipolar disorder [62, 63], anxiety and stress-like disorders [64], Lesch-Nyhan disease and schizophrenia [65, 66].

ADCY3

Expression

ADCY3 mRNA is highly expressed in olfactory sensory neurons (OSN), neuronal primary cilia, and dorsal root ganglion [67, 68]. Bulk and single-cell RNA-sequencing studies showed that ADCY3 was highly expressed in neurons, OPCs, astrocytes, pericytes and vascular fibroblast-like cells; and moderately expressed in oligodendrocytes, microglia, endothelial cells and smooth muscle cells [45, 46]. At the protein level, ADCY3 is mainly found in primary cilia on choroid plexus cells and astrocytes [69].

Function

The high expression of ADCY3 in olfactory sensory neurons suggests that it may regulate odor/pheromone detection [67, 68]. Consistent with these results, ablation of ADCY3 leads to defective olfactory sensory neuron maturation and abnormal olfactory-based behavioral responses, including lack of preference for the test odorants in both sand-buried food task and odor-associated passive avoidance learning paradigm, absence of inter-male aggressiveness and male sexual behavior, and defective maternal behaviors [19, 7072]. In addition, ADCY3-null mice also exhibit impaired learning/memory and short-term memory loss [38], highlighting an essential role of ADCY3 in learning/memory. Furthermore, dysregulation of ADCY3-mediated cAMP signaling in choroid plexus epithelial cells has also been suggested to contribute to the onset of hydrocephalus [73].
GWAS studies demonstrated that ADCY3 polymorphism was associated with obesity [7476], depression [77], and inflammatory bowel disease [78, 79].

ADCY4

Expression

ADCY4 mRNA is expressed at extremely low levels in various brain regions, including olfactory bulbs, cerebral cortex, hippocampus, amygdala, basal ganglia, thalamus, hypothalamus, pons, medulla and cerebellum [58, 80]. Bulk and single-cell RNA-sequencing analyses demonstrated that ADCY4 expression was predominantly detected in endothelial cells [45, 46]. One study reported ADCY4 expression in dentate gyrus and hippocampal CA1/CA3 regions at the protein level [59].

Function

The expression of ADCY4 in hippocampus and dentate gyrus suggests a possible role in synaptic plasticity [59]. Although ADCY4 is also detected in olfactory cilia [19], it does not seem to play a role in olfactory perception since it cannot rescue anosmia in ADCY3-null mice [19].
Outside the CNS, ADCY4 is mainly expressed in the kidney. However, loss of ADCY4 in kidney collecting duct principal cells fails to affect vasopressin-stimulated cAMP generation or sodium/water reabsorption [81], highlighting a dispensable role of ADCY4 in these cells.

ADCY5

Expression

ADCY5 mRNA is highly expressed in the olfactory system, piriform cortex and striatum; and weakly expressed in thalamus and hippocampus [58, 80]. Bulk and single-cell RNA-sequencing analyses revealed high levels of ADCY5 in neurons, vascular fibroblast-like cells, smooth muscle cells, OPCs and oligodendrocytes; and low levels of ADCY5 in microglia, astrocytes, pericytes and endothelial cells [45, 46]. Consistent with these findings, ADCY5 mRNA is detected in cholinergic interneurons and GABAergic medium spiny neurons in the striatum [82, 83].

Function

In vitro study showed that P19 cells up-regulated ADCY5 during neuronal differentiation [60], highlighting an important role of ADCY5 in neuronal maturation/function. Echoed with this result, knockdown of ADCY5 in nucleus accumbens decreases cAMP, leading to blood–brain barrier (BBB) disruption, social stress and depression-like behaviors [84]. Similarly, ADCY5-null mice exhibit poor stress-coping responses [85], indicating a critical role of ADCY5 in the regulation of anxiety and stress. In addition, loss of ADCY5 also impairs striatum-dependent learning, corticostriatal plasticity, dopamine signaling and motor activity [83, 86].
Outside the CNS, ADCY5 participates in the regulation of heart function. In vitro study showed that ADCY5 expression correlated with the appearance of beating cardiomyocytes and transcription of MLC1A (myosin light chain 1 atrial isoform) during mesodermal differentiation of P19 cells, highlighting an important role of ADCY5 in early cardiogenesis and cardiomyocyte differentiation [61]. In addition, deletion of ADCY5 improves basal left ventricular function [87, 88], protects the heart against chronic βAR stimulation [89] and age-related cardiomyopathy [90, 91]. These results indicate a detrimental role of ADCY5 in heart function.
ADCY5 polymorphism has been linked to neuropsychiatric disorders. For example, a missense mutation (A726T) has been associated with familial dyskinesia with facial myokymia (FDFM) [92]. In addition, a homozygous missense or heterozygous de novo mutation (p.R418W) results in early onset of motor disability and movement disorder with severe intellectual disability [9395].

ADCY6

Expression

ADCY6 has a similar but broader and higher expression pattern as ADCY5. In addition to the olfactory system, piriform cortex and striatum, ADCY6 mRNA is also highly expressed in the limbic areas, including amygdala, hippocampus, dentate gyrus and hypothalamus [58, 80]. Bulk and single-cell RNA-sequencing analyses showed that ADCY6 was highly expressed in neurons, OPCs, oligodendrocytes, astrocytes, endothelial cells, pericytes and smooth muscle cells [45, 46].

Function

The function of ADCY6 in the CNS remains unknown. Outside the CNS, ADCY6 is involved in the pathogenesis of cardiac and renal disorders. In vitro study showed that P19 cells up-regulated ADCY6 during mesodermal differentiation [61]. Expression of ADCY6 in the left ventricle of pigs with congestive heart failure increases cardiac contractility and ameliorates cardiac failure [96]. Although loss of ADCY6 does not affect basal cAMP level, it greatly reduces βAR-stimulated cAMP production [97, 98]. ADCY6-null mice display increased urine output, decreased urine osmolarity, reduced responsiveness to arginine vasopressin (AVP), and mild Bartter syndrome-like phenotype [99]. Together, these findings highlight important roles of ADCY6 in cardiac and renal functions. In addition, homozygous missense mutation (R1116C) in ADCY6 reduces myelination in peripheral nervous system, contributing to human axoglial diseases [100] and lethal congenital contracture syndrome [101]. ADCY6 has also been identified as a prognostic factor involved in DNA methylation-regulated immune processes in luminal-like breast cancer [102].

ADCY7

Expression

ADCY7 mRNA expression is restricted to thalamus and hypothalamus, with lower expression in cerebral cortex, amygdala, corpus callosum, cerebellum and olfactory bulbs [41, 58, 80]. Bulk and single-cell RNA-sequencing analyses showed that ADCY7 was highly expressed in microglia and vascular fibroblast-like cells [45, 46]. At the protein level, ADCY7 expression is mainly found in hippocampus, cerebellum, caudate-putamen, cerebral cortex and nucleus accumbens [103].

Function

Ethanol-induced GABAergic transmission in central amygdala neurons was ablated in ADCY7+/ brain slices [104], while mutant mice overexpressing human ADCY7 in the brain displayed higher plasma adrenocorticotropin and corticosterone levels after ethanol injection  [105]. These findings suggest that ADCY7 plays an important role in ethanol modulation of presynaptic GABA release, which may underlie ethanol-related behaviors such as anxiety and dependence.
There is also evidence suggesting that ADCY7 is involved in mood regulation and major depressive disorder. It has been reported that overexpression of ADCY7 in female mice increases depression-like behaviors, while ADCY7+/− mice display decreased depression-like symptoms [42]. Consistent with this finding, a tetranucleotide repeat [(AACA)7] polymorphism in ADCY7 is associated with depressive disorders in humans [42, 65]. Postmortem study found increased ADCY7 expression in the amygdala and anterior cingulate cortex of patients with depression [41].
In addition, ADCY7 is a major contributor of cAMP in T and B lymphocytes. Loss of ADCY7 leads to fewer leukocytes and higher mortality upon bacterial infections [106, 107], indicating an essential role of ADCY7 in immune responses. Consistent with these results, ADCY7 polymorphism is associated with inflammatory bowel disease, Crohn’s disease, ulcerative colitis and autoimmune diseases [108, 109].

ADCY8

Expression

During early postnatal life, ADCY8 mRNA is expressed in hippocampal CA1 region, cortex, cerebellum, olfactory bulb, hypothalamus, amygdala and basal ganglia. In adulthood, ADCY8 is found in olfactory bulb, cerebellum, hypothalamus, thalamus, hippocampal CA1 region, habenula, cerebral and piriform cortices [43, 110]. Bulk and single-cell RNA-sequencing analyses showed that ADCY8 was predominantly expressed in neurons, OPCs and astrocytes [45, 46].

Function

In vitro study showed that P19 cells up-regulated ADCY8 during neuronal differentiation [60], suggesting a possible role of ADCY8 in neuronal development. In vivo study demonstrated that knockdown of ADCY8 ablated the midline-crossing of retinal neurons in zebrafish, resulting in mis-projections of exons to the ipsilateral tectum [111], highlighting an essential role of ADCY8 in axonal pathfinding. ADCY8-null mice showed defective short-term plasticity, impaired presynaptic/postsynaptic LTP and abnormal anxiety-like behaviors under stress [29, 33, 51]. In addition, ADCY8-null mice exhibited no reduction in allodynia and slightly reduced behavioral nociceptive responses to subcutaneous formalin injection or nerve injury [52]. ADCY1-null and ADCY1/ADCY8 double knockout mice, on the other hand, displayed more dramatic changes in these tests [52]. These findings indicate a relatively less important role of ADCY8 in behavioral responses to inflammation or nerve injury compared to ADCY1.
GWAS studies showed that ADCY8 polymorphism was associated with various neuropsychiatric disorders, including dissociative amnesia, post-traumatic stress disorder, depression and bipolar disorder [37, 112, 113].

ADCY9

Expression

ADCY9 mRNA is broadly expressed in the brain with high levels in olfactory system, neocortex, piriform cortex, hippocampus, dentate gyrus, thalamus, hypothalamus and cerebellum [114, 115] Bulk and single-cell RNA-sequencing analyses showed that ADCY9 was highly expressed in almost all cell types in the CNS, including neurons, OPC, oligodendrocytes, astrocytes, microglia, endothelial cells, pericytes, smooth muscle cells and vascular fibroblast-like cells [45, 46].

Function

Although ADCY9 is abundantly expressed in the brain, its function in the CNS remains largely unknown. Loss of ADCY9 leads to grade 1 ventricular diastolic dysfunction and embryonic lethality [116], preventing investigation of its function in adulthood. A study found reduced expression of ADCY9 in the hippocampus in aged mice [48]. More importantly, ADCY9 was significantly increased in mouse hippocampus after spatial learning and its expression correlated with animal performance in the Morris water maze test [48]. These findings suggest that ADCY9 may regulate cognitive function and learning/memory.
In addition, there is also evidence showing that ADCY9 modulates immune function. For example, it has been reported that ADCY9 regulates the chemotaxis of neutrophils and monocytes [117] as well as T cell function [11, 118].
GWAS studies found that ADCY9 polymorphism was associated with asthma [119, 120], mood disorders [121], and the efficacy of dalcetrapib, an antiatherogenic drug [122].

ADCY10

Expression

Bulk RNA-sequencings analysis showed minimal expression of ADCY10 in neurons, OPCs, oligodendrocytes, astrocytes, microglia and endothelial cells [45]. Single-cell RNA-sequencing study found relatively high expression of ADCY10 in endothelial cells and astrocytes [46]. At the protein level, ADCY10 is found in astrocytes [123], developing neurons [124], and neurons of visual cortex, hippocampus and cerebellum [125, 126]. In addition, ADCY10 expression has also been found in the choroid plexus at both mRNA [127] and protein [128, 129] levels.

Function

ADCY10 activation in astrocytes increases cAMP level, induces glycogenolysis/glycolysis, and provides energy substrate for astrocytes and neurons [123], suggesting an important role in astrocyte-neuron metabolic coupling. Overexpression of ADCY10 in retinal ganglion and dorsal root ganglion cells promotes axonal outgrowth and growth cone elaboration, whereas inhibition of ADCY10 reverses these changes [124, 125], strongly indicating an essential role of ADCY10 in axonal outgrowth. Based on that ADCY10 is expressed in the choroid plexus and CO2 metabolism is linked to cerebrospinal fluid secretion [130], it is hypothesized that ADCY10 regulates cerebrospinal fluid homeostasis. This is evidence showing that increased ADCY10 expression caused by chloral hydrate-induced removal of cilia enhances transcytosis in choroid plexus epithelial cells [131]. Two ADCY10 knockout mouse lines have been generated: C1KO and C2KO, which prevent the expression of C1 and C2 domains, respectively. Both display defective sperm motility due to decreased cAMP production in testis and spermatozoa [126, 132], highlighting a crucial role of ADCY10 in male infertility.
A clinical study revealed that ADCY10 polymorphism is associated with absorptive hypercalciuria and low spinal bone density [133].

Effects of GPCR-ADCY signaling in BBB integrity

The BBB is a unique feature of CNS blood vessels. It is mainly composed of brain endothelial cells, pericytes, astrocytes, microglia, neurons and a non-cellular component—the basal lamina. By tightly regulating what enters/exits the CNS, the BBB maintains brain homeostasis [134, 135].
The effects of GPCR-ADCY signaling in BBB maintenance remain largely unclear, partially due to the complexity of GPCR-ADCY system. There are 10 different ADCY isoforms, which are coupled to distinct GPCRs in different cell types. However, there is evidence suggesting that certain GPCRs may regulate BBB integrity via ADCY activity, although the specific ADCY isoforms involved in each case remain unknown. Below we briefly discuss a few such GPCRs, including sphingosine 1-phosphate receptors (S1PRs), lysophosphatidic acid receptors (LPARs), cannabinoid receptors (CBs), adenosine receptors (ARs), G protein-coupled estrogen receptor 1 (GPER-1), complement C5a receptor (C5aR), somatostatin receptors (SSTRs), glucagon-like peptide-1 receptor (GLP1R), and hydrocarboxylic acid receptor 1 (HCAR1). The expression, G protein subtypes, and functions (in BBB integrity) of these GPCRs are summarized in Table 4.
Table 4
GPCR and ADCY expression and function on BBB integrity
GPCRs
Types of G proteins
BBB integrity
Cell types
References
S1PR1
i/o
Increase
Astrocytes and endothelial cells
[136, 138140]
S1PR2
i/o, Gαq/11, and Gα12/13
Decrease
Pericytes, glia, fibroblasts, and endothelial cells
S1PR3
i/o, Gαq/11, and Gα12/13
Decrease
Astrocytes and endothelial cells
S1PR5
Gαi/o and Gα12/13
Increase
Oligodendrocytes and endothelial cells
LAPR1
i/o, Gαq/11, and Gα12/13
Decrease
Microglia, oligodendrocytes, astrocytes, and endothelial cells
[147150, 153, 155]
LAPR2
i/o, Gαq/11, and Gα12/13
Decrease
Neuron, fetal astrocytes, and endothelial cells
LAPR3
i/o and Gαq/11
Decrease
Microglia, astrocytes, and endothelial cells
LAPR6
Gαs and Gα12/13
Decrease
Microglia, oligodendrocytes, and endothelial cells
CB1
Gαs, Gαi/o, and Gαq
Increase
Microglia and neurons
[156160]
CB2
Gαs, Gαi/o, and Gαq
Increase
Microglia and neurons
AR-A1
i/o
Decrease
Microglia, neurons, oligodendrocytes, astrocytes, and endothelial cells
[139, 162, 163]
AR-A2A
Gαs
Decrease
Microglia, neurons, and astrocytes
GPER-1
Gαs and Gβγ
Increase
Neurons
[166168]
C5aR
i/o
Decrease
Microglia, astrocytes, and neurons
[169, 170]
SSTRs
i/o and Gβγ
Increase
Neurons
[172174]
GLP1R
Gαs
Increase
Microglia, astrocytes, neurons, and endothelial cells
[175177]
HCAR1
i/o
Increase
Astrocytes, neurons, and endothelial cells
[178180]

S1PRs

S1PRs are the receptors for sphingosine 1-phosphate, a signaling sphingolipid with a diverse range of functions. There are 5 subtypes of S1PRs (S1PR1-5), among which four (S1PR1-3 and S1PR5) have been shown to regulate BBB integrity. In the CNS, S1PR1 and S1PR3 are mainly expressed in astrocytes and endothelial cells; S1PR2 is found in pericytes, glial cells, endothelial cells and fibroblasts [136]; and S1PR5 is mainly found in oligodendrocytes and endothelial cells [137, 138]. S1PR1 is coupled to Gαi/o; S1PR2 and S1PR3 are coupled to Gαi/o,q/11 and Gα12/13; and S1PR5 is coupled to Gαi/o and Gα12/13 [139, 140]. Functional studies suggest that S1PR1 regulates BBB integrity. It has been reported that S1PR1/5 agonist siponimod (BAF-312) enhances BBB integrity and increases tight junction protein expression in an in vitro BBB model [141]. Consistent with this finding, S1PR1 functional antagonist (FTY720P) and endothelium-specific knockout of S1PR1 substantially increase BBB permeability to small tracers [142]. There is also evidence showing that S1PR1 can be targeted to facilitate CNS drug delivery. It has been shown that targeting S1PR with S1P and S1PR agonist fingolimod improves CNS drug delivery by reducing basal activity of P-glycoprotein (P-gp), an ATP-driven drug efflux pump, at the BBB and blood–spinal cord barrier, which significantly increases the uptake of radiolabeled P-gp substrates such as verapamil (three-fold), loperamide (five-fold) and paclitaxel (five-fold) [143, 144]. Similarly, pharmacological studies suggest that S1PR2 and S1PR3 function to decrease BBB integrity [145, 146]. S1PR2 antagonist ameliorates oxidative stress-induced cerebrovascular endothelial barrier impairment and reduces BBB leakage after ischemic injury in mice [145]. S1PR3 antagonist CAY10444 attenuates BBB damage by up-regulating tight junction proteins, reduces brain edema, and improves animal behavior in acute intracerebral hemorrhage [146]. S1PR5, on the other hand, seems to promote BBB integrity. It has bene reported that S1PR5-selective agonist improves BBB integrity in vitro and reduces trans-endothelial migration of monocytes. Echoed with these findings, knockdown of S1PR5 compromises BBB integrity and reduces the expression of tight junction proteins, P-gp and BCRP [138]. It should be noted, however, that the specific ADCY isoforms associated with S1PR1-3 and S1PR5 signaling remain unknown.

LPARs

LPARs are the receptors for lysophosphatidic acid (LPA), a bioactive lipid with important functions in physiology and pathology. There are six subtypes of LPARs (LPAR1-6), among which four (LPAR1-3 and LPAR6) have been shown to regulate BBB integrity. LPAR1 is mainly expressed in astrocytes, microglia, oligodendrocytes and endothelial cells; LPAR2 in endothelial cells, neuron and fetal astrocytes; LPAR3 in microglia, astrocytes and endothelial cells; and LPAR6 in microglia, oligodendrocytes, endothelial cells [147, 148]. LPAR1-3 signal through Gαi/o, Gαq/11 and Gα12/13 [147, 149], while LPAR6 signals through Gαs and Gα12/13 [149]. All of these LPARs (LPAR1-3 and LPAR6) function to decrease BBB integrity. It has been shown that LPA decreases tight junction protein expression and transendothelial electrical resistance via LPAR6 in rat brain endothelial cells [150]. Echoed with this finding, intravenous injection of LPA up-regulates LPAR1-3 expression and transiently increases BBB permeability [151]. Additionally, LPA and amitriptyline have been shown to reduce basal P-gp activity through LPAR1 signaling without affecting the activity of MRP2 (multidrug resistance-associated protein 2) or BCRP (breast cancer resistance protein) in both rat brain capillaries and a rat model of amyotrophic lateral sclerosis [152]. Similarly, LPAR ligand gintonin increases tight junction spaces and decreases tight junction protein expression in human brain microvascular endothelial cells. Gintonin has been shown to enter the brain via LPAR1/LPAR3 and enhance BBB permeability to various tracers in vivo [153]. Consistent with these findings, gintonin enhances CNS delivery of donepezil in a time-dependent manner via LPAR1/3 [154]. LPAR inhibitors (HA130, PF8380 and BrP-LPA), on the other hand, reverse BBB damage and enhance tight junction protein expression after ischemic stroke [155].

CBs

CBs have two subtypes (CB1 and CB2), both of which mediate BBB regulation in healthy and injured/diseased conditions. In the CNS, CB1 and CB2 are mainly expressed in neurons and microglia [156, 157]. They can stimulate and/or inhibit various ADCY isoforms independently. It has been shown that activation of CB1 and CB2 by cannabinoid agonists stimulates group II ADCYs (ADCY2, ADCY4 and ADCY7) through Gαs, but inhibits other transmembrane ADCYs (ADCY1, ADCY3, ADCY5, ADCY6, ADCY8 and ADCY9) through Gαi/o and Gαq [158, 159]. In vitro study showed that pharmacological activation of CB1 but not CB2 restored tight junction stability in HIV-1-induced BBB disruption model [160]. Consistent with this finding, CB1-specific cannabinoid agonists inhibited HIV-1 Gp120-mediated BBB damage and prevented down-regulation of tight junction proteins both in vitro and in vivo [160]. Interestingly, CB2-selective agonist O-1966 prevented LPS-induced loss of tight junction proteins in brain microvascular endothelial cells [157]. These findings suggest a protective role of CB1/2 in BBB integrity.

ARs

ARs are the receptors for adenosine, a purine nucleoside released by neurons and glial cells. There are four subtypes of ARs (A1, A2A, A2B and A3), among which AR-A1 and AR-A2A are involved in BBB regulation. In the CNS, AR-A1 and AR-A2A are predominantly expressed in microglia, oligodendrocytes, astrocytes, neurons and endothelial cells [161]. AR-A1 inhibits ADCY activity through Gαi/o, whereas AR-A2A stimulates ADCY activity via Gαs [139, 162]. Although coupled to different G proteins, both ARs function to compromise BBB integrity. It has been shown that activation of AR-A1 and AR-A2A increases BBB permeability and reduces tight junction protein expression [163]. In addition, AR agonists have been used to facilitate the entry of intravenously administered molecules into the brain [164]. For example, AR-A2A agonist lexiscan has been reported to inhibit the expression of P-gp and BCRP and increase the accumulation of the epirubicin, a P-gp substrate and chemotherapeutic drug, in mouse brain [165]. In addition, lexiscan has also been shown to increase paracellular leakage in cultured brain endothelial cells, enabling a wider therapeutic window for therapeutics to enter the brain [162].

GPER-1

GPER-1, also known as GPR30, is a novel estrogen receptor highly expressed in neurons [166]. Activation of GPER-1 stimulates ADCY activity via Gαs and Gβγ [167]. GPER-1 activation has been shown to reduce BBB leakage and increase tight junction proteins after ischemic injury [168], highlighting a protective role in BBB integrity.

C5aR

C5aR is the receptor for C5a, a potent proinflammatory peptide generated during complement system activation. In the CNS, C5aR is constitutively expressed in astrocytes, microglia and neurons. Activation of C5aR inhibits ADCY activity via Gαi/o [169, 170]. In vitro study showed that activation of C5aR increased BBB permeability and decreased tight junction protein expression [171], suggesting a detrimental role of C5aR in BBB integrity.

SSTRs

SSTRs mediate the effect of somatostatin, a neuropeptide with important functions in modulating cortical circuits and cognition. There are five subtypes of SSTRs (SSTR1-5), all of which are mainly expressed in neurons and inhibit ADCY activity via Gαi/o and Gβγ [172, 173]. In vitro study showed that somatostatin and selective SSTR agonists maintained BBB integrity and restored ZO-1 organization in cytokine- and LPS-treated human brain endothelial cells [174], suggesting that SSTR activation protects BBB integrity.

GLP1R

GLP1R mediates the function of glucagon-like peptide-1, a short peptide hormone secreted by intestinal enteroendocrine L cells and certain neurons. In the CNS, GLP1R is mainly expressed in astrocytes, neurons, microglia and endothelial cells [175]. Activation of GLP1R stimulates ADCY activity via Gαs [176]. In vitro study showed that GLP-1 increased tight junction protein expression and decreased paracellular permeability in brain capillary endothelial cells via cAMP-PKA signaling pathway [177], indicating a protective role in BBB integrity.

HCAR1

HCAR1, also known as GPR81, is expressed in endothelial cells, astrocytes and neurons [178, 179]. It inhibits ADCY activity through Gαi/o [178, 179]. In vitro study showed that LPS reduced the expression of HCAR1 and tight junction proteins and increased BBB permeability in rat brain microvascular endothelial cells [180]. In addition, activation of HCAR1 stimulates mitochondrial biogenesis and regulates monocarboxylate transporter expression in brain endothelial cells, which are crucial for the metabolism and function of the neurovascular unit [181, 182]. These results suggest a possible role of HCAR1 in BBB maintenance.

Conclusions and future directions

Since the identification of cAMP as an important second messenger, substantial progress has been made with respect to the structure, expression, regulation and functions of ADCYs. There are, however, still several key questions that need to be answered in future research.
First, the expression profiles of ADCYs at the protein level remain largely unknown, possibly due to the lack of isoform-specific antibodies. Current knowledge on ADCY expression is mainly at the mRNA level. Future research should focus on addressing this bottleneck by developing isoform-specific antibodies and innovative genetic tools (e.g., reporter mouse lines).
Second, there is a lack of genetic knockout/overexpression models for certain ADCY isoforms. For example, the phenotypes of ADCY2, ADCY4 and ADCY7 global knockout mice as well as ADCY2, ADCY3, ADCY4 and ADCY10 overexpression mice remain unknown. In addition, the cell-specific conditional knockout mice for many ADCY isoforms are still lacking. Furthermore, there are currently few compound knockout mice available, which are useful in dissecting the roles of ADCYs with compensatory/overlapping functions. Generating these genetic tools will enable loss-of-function studies and substantially move the field forward.
Third, there is a lack of isoform-specific pharmacological reagents for ADCYs. Incomplete pharmacological characterization of mammalian ADCYs has resulted in misconceptions/errors in the selectivity of certain compounds [183]. The lack of selectivity and potency of pharmacological reagents has often resulted in inaccurate or even faulty conclusions in ADCY research. Future research should focus on screening and identifying isoform-specific ADCY activators and inhibitors.
Fourth, the association profiles between ADCYs and GPCRs in different cell types remain unknown. Establishing a cell-specific GPCR-ADCY association profile will significantly enrich our knowledge in GPCR-ADCY signaling. Together with the cell-specific expression profiles of GPCRs and ADCYs, this association profile makes it possible to determine the crosstalk between various signaling pathways, promoting more accurate and safer treatments.
Fifth, more efficient and specific detection approaches are needed for ADCY research. Although multiple methods exist to measure cAMP levels in cells, these approaches are mostly end-point assays and unable to reflect cAMP levels in real time or that generated by a specific ADCY isoform [184, 185]. Although fluorescence resonance energy transfer (FRET)-based cAMP biosensors allow the cAMP detection in living cells in real time [186, 187], they usually have low efficiency and sensitivity. More sensitive and rapid approaches are needed. In addition, biosensors that are able to target different subcellular compartments may help study localized cAMP dynamics [188, 189].
Last, the functions of ADCYs are not fully understood. For example, the roles of ADCYs in CNS barriers (e.g., BBB, blood-CSF barrier and brain-CSF barrier) and the underlying molecular mechanisms are only partially understood. In addition, the functions of ADCY isoforms in human diseases and the links between ADCY gene polymorphisms and human diseases remain largely unknown. With the generation of novel tools (isoform-specific antibodies and genetic mouse lines), we expect to determine the functional significance of each ADCY isoform in a cell-specific manner.

Acknowledgements

We thank the Yao lab for helpful discussions. The figures were made using BioRender.

Declarations

Not applicable.
Not applicable.

Competing interests

The authors have no financial or non-financial competing interests.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Kawasaki H, Springett GM, Mochizuki N, Toki S, Nakaya M, Matsuda M, et al. A family of cAMP-binding proteins that directly activate Rap1. Science. 1998;282:2275–9.PubMedCrossRef Kawasaki H, Springett GM, Mochizuki N, Toki S, Nakaya M, Matsuda M, et al. A family of cAMP-binding proteins that directly activate Rap1. Science. 1998;282:2275–9.PubMedCrossRef
2.
3.
Zurück zum Zitat Serezani CH, Ballinger MN, Aronoff DM, Peters-Golden M. Cyclic AMP: Master regulator of innate immune cell function. Am J Respir Cell Mol Biol. 2008;39:127–32.PubMedPubMedCentralCrossRef Serezani CH, Ballinger MN, Aronoff DM, Peters-Golden M. Cyclic AMP: Master regulator of innate immune cell function. Am J Respir Cell Mol Biol. 2008;39:127–32.PubMedPubMedCentralCrossRef
4.
Zurück zum Zitat Kleuss C. Adenylate cyclase. Pharm Compr Pharmacol Ref. 2007;1–12. Kleuss C. Adenylate cyclase. Pharm Compr Pharmacol Ref. 2007;1–12.
5.
Zurück zum Zitat Linder JU, Schultz JE. The class III adenylyl cyclases: multi-purpose signalling modules. Cell Signal. 2003;15:1081–9.PubMedCrossRef Linder JU, Schultz JE. The class III adenylyl cyclases: multi-purpose signalling modules. Cell Signal. 2003;15:1081–9.PubMedCrossRef
7.
Zurück zum Zitat Shimada T, Fujita N, Yamamoto K, Ishihama A. Novel roles of camp receptor protein (CRP) in regulation of transport and metabolism of carbon sources. PLoS ONE. 2011;6:e20081.PubMedPubMedCentralCrossRef Shimada T, Fujita N, Yamamoto K, Ishihama A. Novel roles of camp receptor protein (CRP) in regulation of transport and metabolism of carbon sources. PLoS ONE. 2011;6:e20081.PubMedPubMedCentralCrossRef
8.
Zurück zum Zitat Ahuja N, Kumar P, Bhatnagar R. The adenylate cyclase toxins. Crit Rev Microbiol. 2004;30:187–96.PubMedCrossRef Ahuja N, Kumar P, Bhatnagar R. The adenylate cyclase toxins. Crit Rev Microbiol. 2004;30:187–96.PubMedCrossRef
9.
Zurück zum Zitat Gallagher DT, Smith NN, Kim SK, Heroux A, Robinson H, Reddy PT. Structure of the class IV adenylyl cyclase reveals a novel fold. J Mol Biol. 2006;362:114–22.PubMedCrossRef Gallagher DT, Smith NN, Kim SK, Heroux A, Robinson H, Reddy PT. Structure of the class IV adenylyl cyclase reveals a novel fold. J Mol Biol. 2006;362:114–22.PubMedCrossRef
10.
Zurück zum Zitat Téllez-Sosa J, Soberón N, Vega-Segura A, Torres-Márquez ME, Cevallos MA. The Rhizobium etli cyaC product: characterization of a novel adenylate cyclase class. J Bacteriol. 2002;184:3560–8.PubMedPubMedCentralCrossRef Téllez-Sosa J, Soberón N, Vega-Segura A, Torres-Márquez ME, Cevallos MA. The Rhizobium etli cyaC product: characterization of a novel adenylate cyclase class. J Bacteriol. 2002;184:3560–8.PubMedPubMedCentralCrossRef
11.
Zurück zum Zitat Dessauer CW, Watts VJ, Ostrom RS, Conti M, Dove S, Seifert R. International union of basic and clinical pharmacology. CI. structures and small molecule modulators of mammalian adenylyl cyclases. Pharmacol Rev. 2017;69:96–139.CrossRef Dessauer CW, Watts VJ, Ostrom RS, Conti M, Dove S, Seifert R. International union of basic and clinical pharmacology. CI. structures and small molecule modulators of mammalian adenylyl cyclases. Pharmacol Rev. 2017;69:96–139.CrossRef
12.
Zurück zum Zitat Brust TF. Second messengers | adenylyl cyclases. Encycl Biol Chem. 2021;III:496–502. Brust TF. Second messengers | adenylyl cyclases. Encycl Biol Chem. 2021;III:496–502.
13.
Zurück zum Zitat Kleinboelting S, Diaz A, Moniot S, Van Den Heuvel J, Weyand M, Levin LR, et al. Crystal structures of human soluble adenylyl cyclase reveal mechanisms of catalysis and of its activation through bicarbonate. Proc Natl Acad Sci USA. 2014;111:3727–32.PubMedPubMedCentralCrossRef Kleinboelting S, Diaz A, Moniot S, Van Den Heuvel J, Weyand M, Levin LR, et al. Crystal structures of human soluble adenylyl cyclase reveal mechanisms of catalysis and of its activation through bicarbonate. Proc Natl Acad Sci USA. 2014;111:3727–32.PubMedPubMedCentralCrossRef
14.
Zurück zum Zitat Hanoune J, Pouille Y, Tzavara E, Shen T, Lipskaya L, Miyamoto N, et al. Adenylyl cyclases: structure, regulation and function in an enzyme superfamily. Mol Cell Endocrinol. 1997;128:179–94.PubMedCrossRef Hanoune J, Pouille Y, Tzavara E, Shen T, Lipskaya L, Miyamoto N, et al. Adenylyl cyclases: structure, regulation and function in an enzyme superfamily. Mol Cell Endocrinol. 1997;128:179–94.PubMedCrossRef
15.
Zurück zum Zitat Defer N, Best-Belpomme M, Hanoune J. Tissue specificity and physiological relevance of various isoforms of adenylyl cyclase. Am J Physiol Physiol. 2000;279:F400–16.CrossRef Defer N, Best-Belpomme M, Hanoune J. Tissue specificity and physiological relevance of various isoforms of adenylyl cyclase. Am J Physiol Physiol. 2000;279:F400–16.CrossRef
17.
Zurück zum Zitat Sadana R, Dessauer CW. Physiological roles for G protein-regulated adenylyl cyclase isoforms: insights from knockout and overexpression studies. Neurosignals. 2009;17:5–22.PubMedCrossRef Sadana R, Dessauer CW. Physiological roles for G protein-regulated adenylyl cyclase isoforms: insights from knockout and overexpression studies. Neurosignals. 2009;17:5–22.PubMedCrossRef
19.
Zurück zum Zitat Wong ST, Trinh K, Hacker B, Chan GCK, Lowe G, Gaggar A, et al. Disruption of the type III adenylyl cyclase gene leads to peripheral and behavioral anosmia in transgenic mice. Neuron. 2000;27:487–97.PubMedCrossRef Wong ST, Trinh K, Hacker B, Chan GCK, Lowe G, Gaggar A, et al. Disruption of the type III adenylyl cyclase gene leads to peripheral and behavioral anosmia in transgenic mice. Neuron. 2000;27:487–97.PubMedCrossRef
20.
Zurück zum Zitat Yan SZ, Huang ZH, Andrews RK, Tang WJ. Conversion of forskolin-insensitive to forskolin-sensitive (mouse-type IX) adenylyl cyclase. Mol Pharmacol. 1998;53:182–7.PubMedCrossRef Yan SZ, Huang ZH, Andrews RK, Tang WJ. Conversion of forskolin-insensitive to forskolin-sensitive (mouse-type IX) adenylyl cyclase. Mol Pharmacol. 1998;53:182–7.PubMedCrossRef
23.
Zurück zum Zitat Jaiswal BS, Conti M. Calcium regulation of the soluble adenylyl cyclase expressed in mammalian spermatozoa. Proc Natl Acad Sci USA. 2003;100:10676–81.PubMedPubMedCentralCrossRef Jaiswal BS, Conti M. Calcium regulation of the soluble adenylyl cyclase expressed in mammalian spermatozoa. Proc Natl Acad Sci USA. 2003;100:10676–81.PubMedPubMedCentralCrossRef
24.
Zurück zum Zitat Zippin JH, Farrell J, Huron D, Kamenetsky M, Hess KC, Fischman DA, et al. Bicarbonate-responsive “soluble” adenylyl cyclase defines a nuclear cAMP microdomain. J Cell Biol. 2004;164:527–34.PubMedPubMedCentralCrossRef Zippin JH, Farrell J, Huron D, Kamenetsky M, Hess KC, Fischman DA, et al. Bicarbonate-responsive “soluble” adenylyl cyclase defines a nuclear cAMP microdomain. J Cell Biol. 2004;164:527–34.PubMedPubMedCentralCrossRef
25.
Zurück zum Zitat Acin-Perez R, Salazar E, Kamenetsky M, Buck J, Levin LR, Manfredi G. Cyclic AMP produced inside mitochondria regulates oxidative phosphorylation. Cell Metab. 2009;9:265–76.PubMedPubMedCentralCrossRef Acin-Perez R, Salazar E, Kamenetsky M, Buck J, Levin LR, Manfredi G. Cyclic AMP produced inside mitochondria regulates oxidative phosphorylation. Cell Metab. 2009;9:265–76.PubMedPubMedCentralCrossRef
26.
Zurück zum Zitat Acin-Perez R, Salazar E, Brosel S, Yang H, Schon EA, Manfredi G. Modulation of mitochondrial protein phosphorylation by soluble adenylyl cyclase ameliorates cytochrome oxidase defects. EMBO Mol Med. 2009;1:392–406.PubMedPubMedCentralCrossRef Acin-Perez R, Salazar E, Brosel S, Yang H, Schon EA, Manfredi G. Modulation of mitochondrial protein phosphorylation by soluble adenylyl cyclase ameliorates cytochrome oxidase defects. EMBO Mol Med. 2009;1:392–406.PubMedPubMedCentralCrossRef
27.
Zurück zum Zitat Wu ZL, Thomas SA, Villacres EC, Xia Z, Simmons ML, Chavkin C, et al. Altered behavior and long-term potentiation in type I adenylyl cyclase mutant mice. Proc Natl Acad Sci USA. 1995;92:220–4.PubMedPubMedCentralCrossRef Wu ZL, Thomas SA, Villacres EC, Xia Z, Simmons ML, Chavkin C, et al. Altered behavior and long-term potentiation in type I adenylyl cyclase mutant mice. Proc Natl Acad Sci USA. 1995;92:220–4.PubMedPubMedCentralCrossRef
28.
Zurück zum Zitat Wang H, Ferguson GD, Pineda VV, Cundiff PE, Storm DR. Overexpression of type-1 adenylyl cyclase in mouse forebrain enhances recognition memory and LTP. Nat Neurosci. 2004;7:635–42.PubMedCrossRef Wang H, Ferguson GD, Pineda VV, Cundiff PE, Storm DR. Overexpression of type-1 adenylyl cyclase in mouse forebrain enhances recognition memory and LTP. Nat Neurosci. 2004;7:635–42.PubMedCrossRef
29.
Zurück zum Zitat Wang H, Pineda VV, Chan GCK, Wong ST, Muglia LJ, Storm DR. Type 8 adenylyl cyclase is targeted to excitatory synapses and required for mossy fiber long-term potentiation. J Neurosci. 2003;23:9710–8.PubMedPubMedCentralCrossRef Wang H, Pineda VV, Chan GCK, Wong ST, Muglia LJ, Storm DR. Type 8 adenylyl cyclase is targeted to excitatory synapses and required for mossy fiber long-term potentiation. J Neurosci. 2003;23:9710–8.PubMedPubMedCentralCrossRef
30.
Zurück zum Zitat Wong ST, Athos J, Figueroa XA, Pineda VV, Schaefer ML, Chavkin CC, et al. Calcium-stimulated adenylyl cyclase activity is critical for hippocampus-dependent long-term memory and late phase LTP. Neuron. 1999;23:787–98.PubMedCrossRef Wong ST, Athos J, Figueroa XA, Pineda VV, Schaefer ML, Chavkin CC, et al. Calcium-stimulated adenylyl cyclase activity is critical for hippocampus-dependent long-term memory and late phase LTP. Neuron. 1999;23:787–98.PubMedCrossRef
31.
Zurück zum Zitat Zheng F, Zhang M, Ding Q, Sethna F, Yan L, Moon C, et al. Voluntary running depreciates the requirement of Ca2+-stimulated cAMP signaling in synaptic potentiation and memory formation. Learn Mem. 2016;23:442–9.PubMedPubMedCentralCrossRef Zheng F, Zhang M, Ding Q, Sethna F, Yan L, Moon C, et al. Voluntary running depreciates the requirement of Ca2+-stimulated cAMP signaling in synaptic potentiation and memory formation. Learn Mem. 2016;23:442–9.PubMedPubMedCentralCrossRef
32.
Zurück zum Zitat Yamamoto M, Götz ME, Ozawa H, Luckhaus C, Saito T, Rösler M, et al. Hippocampal level of neural specific adenylyl cyclase type I is decreased in Alzheimer’s disease. Biochim Biophys Acta - Mol Basis Dis. 2000;1535:60–8.CrossRef Yamamoto M, Götz ME, Ozawa H, Luckhaus C, Saito T, Rösler M, et al. Hippocampal level of neural specific adenylyl cyclase type I is decreased in Alzheimer’s disease. Biochim Biophys Acta - Mol Basis Dis. 2000;1535:60–8.CrossRef
33.
Zurück zum Zitat Schaefer ML, Wong ST, Wozniak DF, Muglia LM, Liauw JA, Zhuo M, et al. Altered stress-induced anxiety in adenylyl cyclase type VIII-deficient mice. J Neurosci. 2000;20:4809–20.PubMedPubMedCentralCrossRef Schaefer ML, Wong ST, Wozniak DF, Muglia LM, Liauw JA, Zhuo M, et al. Altered stress-induced anxiety in adenylyl cyclase type VIII-deficient mice. J Neurosci. 2000;20:4809–20.PubMedPubMedCentralCrossRef
35.
Zurück zum Zitat Razzoli M, Andreoli M, Maraia G, Di Francesco C, Arban R. Functional role of calcium-stimulated adenylyl cyclase 8 in adaptations to psychological stressors in the mouse: implications for mood disorders. Neuroscience. 2010;170:429–40.PubMedCrossRef Razzoli M, Andreoli M, Maraia G, Di Francesco C, Arban R. Functional role of calcium-stimulated adenylyl cyclase 8 in adaptations to psychological stressors in the mouse: implications for mood disorders. Neuroscience. 2010;170:429–40.PubMedCrossRef
36.
Zurück zum Zitat Zhang P, Xiang N, Chen Y, Śliwerska E, McInnis MG, Burmeister M, et al. Family-based association analysis to finemap bipolar linkage peak on chromosome 8q24 using 2,500 genotyped SNPs and 15,000 imputed SNPs. Bipolar Disord. 2010;12:786–92.PubMedPubMedCentralCrossRef Zhang P, Xiang N, Chen Y, Śliwerska E, McInnis MG, Burmeister M, et al. Family-based association analysis to finemap bipolar linkage peak on chromosome 8q24 using 2,500 genotyped SNPs and 15,000 imputed SNPs. Bipolar Disord. 2010;12:786–92.PubMedPubMedCentralCrossRef
38.
Zurück zum Zitat Wang Z, Phan T, Storm DR. The type 3 adenylyl cyclase is required for novel object learning and extinction of contextual memory: role of camp signaling in primary cilia. J Neurosci. 2011;31:5557–61.PubMedPubMedCentralCrossRef Wang Z, Phan T, Storm DR. The type 3 adenylyl cyclase is required for novel object learning and extinction of contextual memory: role of camp signaling in primary cilia. J Neurosci. 2011;31:5557–61.PubMedPubMedCentralCrossRef
39.
Zurück zum Zitat Chen X, Luo J, Leng Y, Yang Y, Zweifel LS, Palmiter RD, et al. Ablation of type III adenylyl cyclase in mice causes reduced neuronal activity, altered sleep pattern, and depression-like phenotypes. Biol Psychiatry. 2016;80:836–48.PubMedCrossRef Chen X, Luo J, Leng Y, Yang Y, Zweifel LS, Palmiter RD, et al. Ablation of type III adenylyl cyclase in mice causes reduced neuronal activity, altered sleep pattern, and depression-like phenotypes. Biol Psychiatry. 2016;80:836–48.PubMedCrossRef
41.
Zurück zum Zitat Joeyen-Waldorf J, Nikolova YS, Edgar N, Walsh C, Kota R, Lewis DA, et al. Adenylate cyclase 7 is implicated in the biology of depression and modulation of affective neural circuitry. Biol Psychiatry. 2012;71:627–32.PubMedPubMedCentralCrossRef Joeyen-Waldorf J, Nikolova YS, Edgar N, Walsh C, Kota R, Lewis DA, et al. Adenylate cyclase 7 is implicated in the biology of depression and modulation of affective neural circuitry. Biol Psychiatry. 2012;71:627–32.PubMedPubMedCentralCrossRef
42.
Zurück zum Zitat Hines LM, Hoffman PL, Bhave S, Saba L, Kaiser A, Snell L, et al. A sex-specific role of type VII adenylyl cyclase in depression. J Neurosci. 2006;26:12609–19.PubMedPubMedCentralCrossRef Hines LM, Hoffman PL, Bhave S, Saba L, Kaiser A, Snell L, et al. A sex-specific role of type VII adenylyl cyclase in depression. J Neurosci. 2006;26:12609–19.PubMedPubMedCentralCrossRef
43.
Zurück zum Zitat Nicol X, Muzerelle A, Bachy I, Ravary A, Gaspar P. Spatiotemporal localization of the calcium-stimulated adenylate cyclases, AC1 and AC8, during mouse brain development. J Comp Neurol. 2005;486:281–94.PubMedCrossRef Nicol X, Muzerelle A, Bachy I, Ravary A, Gaspar P. Spatiotemporal localization of the calcium-stimulated adenylate cyclases, AC1 and AC8, during mouse brain development. J Comp Neurol. 2005;486:281–94.PubMedCrossRef
44.
Zurück zum Zitat Xia Z, Choi E-J, Wang F, Blazynski C, Storm DR. Type I calmodulin-sensitive adenylyl cyclase is neural specific. J Neurochem. 1993;60:305–11.PubMedCrossRef Xia Z, Choi E-J, Wang F, Blazynski C, Storm DR. Type I calmodulin-sensitive adenylyl cyclase is neural specific. J Neurochem. 1993;60:305–11.PubMedCrossRef
47.
Zurück zum Zitat Konur S, Ghosh A. Calcium signaling and the control of dendritic development. Neuron. 2005;46:401–5.PubMedCrossRef Konur S, Ghosh A. Calcium signaling and the control of dendritic development. Neuron. 2005;46:401–5.PubMedCrossRef
48.
Zurück zum Zitat Mons N, Segu L, Nogues X, Buhot MC. Effects of age and spatial learning on adenylyl cyclase mRNA expression in the mouse hippocampus. Neurobiol Aging. 2004;25:1095–106.PubMedCrossRef Mons N, Segu L, Nogues X, Buhot MC. Effects of age and spatial learning on adenylyl cyclase mRNA expression in the mouse hippocampus. Neurobiol Aging. 2004;25:1095–106.PubMedCrossRef
49.
Zurück zum Zitat Mons N, Guillou JL, Decorte L, Jaffard R. Spatial learning induces differential changes in calcium/calmodulin-stimulated (ACI) and calcium-insensitive (ACII) adenylyl cyclases in the mouse hippocampus. Neurobiol Learn Mem. 2003;79:226–35.PubMedCrossRef Mons N, Guillou JL, Decorte L, Jaffard R. Spatial learning induces differential changes in calcium/calmodulin-stimulated (ACI) and calcium-insensitive (ACII) adenylyl cyclases in the mouse hippocampus. Neurobiol Learn Mem. 2003;79:226–35.PubMedCrossRef
50.
Zurück zum Zitat Storm DR, Hansel C, Hacker B, Parent A, Linden DJ. Impaired cerebellar long-term potentiation in type I adenylyl cyclase mutant mice. Neuron. 1998;20:1199–210.PubMedCrossRef Storm DR, Hansel C, Hacker B, Parent A, Linden DJ. Impaired cerebellar long-term potentiation in type I adenylyl cyclase mutant mice. Neuron. 1998;20:1199–210.PubMedCrossRef
52.
Zurück zum Zitat Wei F, Qiu CS, Kim SJ, Muglia L, Maas JW, Pineda VV, et al. Genetic elimination of behavioral sensitization in mice lacking calmodulin-stimulated adenylyl cyclases. Neuron. 2002;36:713–26.PubMedCrossRef Wei F, Qiu CS, Kim SJ, Muglia L, Maas JW, Pineda VV, et al. Genetic elimination of behavioral sensitization in mice lacking calmodulin-stimulated adenylyl cyclases. Neuron. 2002;36:713–26.PubMedCrossRef
53.
Zurück zum Zitat Sethna F, Feng W, Ding Q, Robison AJ, Feng Y, Wang H. Enhanced expression of ADCY1 underlies aberrant neuronal signalling and behaviour in a syndromic autism model. Nat Commun. 2017;8:14359.PubMedPubMedCentralCrossRef Sethna F, Feng W, Ding Q, Robison AJ, Feng Y, Wang H. Enhanced expression of ADCY1 underlies aberrant neuronal signalling and behaviour in a syndromic autism model. Nat Commun. 2017;8:14359.PubMedPubMedCentralCrossRef
54.
Zurück zum Zitat Ansar M, Chahrour MH, AminudDin M, Arshad M, Haque S, Pham TL, et al. DFNB44, a novel autosomal recessive non-syndromic hearing impairment locus, maps to chromosome 7p14.1-q11.22. Hum Hered. 2004;57:195–9.PubMedCrossRef Ansar M, Chahrour MH, AminudDin M, Arshad M, Haque S, Pham TL, et al. DFNB44, a novel autosomal recessive non-syndromic hearing impairment locus, maps to chromosome 7p14.1-q11.22. Hum Hered. 2004;57:195–9.PubMedCrossRef
56.
Zurück zum Zitat Amare AT, Schubert KO, Hou L, Clark SR, Papiol S, Heilbronner U, et al. Association of polygenic score for schizophrenia and HLA antigen and inflammation genes with response to lithium in bipolar affective disorder: a genome-wide association study. JAMA Psychiat. 2018;75:65–74. Amare AT, Schubert KO, Hou L, Clark SR, Papiol S, Heilbronner U, et al. Association of polygenic score for schizophrenia and HLA antigen and inflammation genes with response to lithium in bipolar affective disorder: a genome-wide association study. JAMA Psychiat. 2018;75:65–74.
59.
Zurück zum Zitat Baker LP, Nielsen MD, Impey S, Hacker BM, Poser SW, Chan MYM, et al. Regulation and immunohistochemical localization of βγ-stimulated adenylyl cyclases in mouse hippocampus. J Neurosci. 1999;19:180–92.PubMedPubMedCentralCrossRef Baker LP, Nielsen MD, Impey S, Hacker BM, Poser SW, Chan MYM, et al. Regulation and immunohistochemical localization of βγ-stimulated adenylyl cyclases in mouse hippocampus. J Neurosci. 1999;19:180–92.PubMedPubMedCentralCrossRef
61.
Zurück zum Zitat Lipskaia L, Grépin C, Defer N, Hanoune J. Adenylyl cyclase activity and gene expression during mesodermal differentiation of the P19 embryonal carcinoma cells. J Cell Physiol. 1998;176:50–6.PubMedCrossRef Lipskaia L, Grépin C, Defer N, Hanoune J. Adenylyl cyclase activity and gene expression during mesodermal differentiation of the P19 embryonal carcinoma cells. J Cell Physiol. 1998;176:50–6.PubMedCrossRef
62.
Zurück zum Zitat McCarthy MJ, Liang S, Spadoni AD, Kelsoe JR, Simmons AN. Whole brain expression of bipolar disorder associated genes: structural and genetic analyses. PLoS ONE. 2014;9:e100204.PubMedPubMedCentralCrossRef McCarthy MJ, Liang S, Spadoni AD, Kelsoe JR, Simmons AN. Whole brain expression of bipolar disorder associated genes: structural and genetic analyses. PLoS ONE. 2014;9:e100204.PubMedPubMedCentralCrossRef
63.
Zurück zum Zitat Mullins N, Forstner AJ, O’Connell KS, Coombes B, Coleman JRI, Qiao Z, et al. Genome-wide association study of more than 40,000 bipolar disorder cases provides new insights into the underlying biology. Nat Genet. 2021;53:817–29.PubMedPubMedCentralCrossRef Mullins N, Forstner AJ, O’Connell KS, Coombes B, Coleman JRI, Qiao Z, et al. Genome-wide association study of more than 40,000 bipolar disorder cases provides new insights into the underlying biology. Nat Genet. 2021;53:817–29.PubMedPubMedCentralCrossRef
64.
Zurück zum Zitat Meier SM, Trontti K, Purves KL, Als TD, Grove J, Laine M, et al. Genetic variants associated with anxiety and stress-related disorders: a genome-wide association study and mouse-model study. JAMA Psychiat. 2019;76:924–32.CrossRef Meier SM, Trontti K, Purves KL, Als TD, Grove J, Laine M, et al. Genetic variants associated with anxiety and stress-related disorders: a genome-wide association study and mouse-model study. JAMA Psychiat. 2019;76:924–32.CrossRef
66.
Zurück zum Zitat Peyrot WJ, Price AL. Identifying loci with different allele frequencies among cases of eight psychiatric disorders using CC-GWAS. Nat Genet. 2021;53:445–54.PubMedPubMedCentralCrossRef Peyrot WJ, Price AL. Identifying loci with different allele frequencies among cases of eight psychiatric disorders using CC-GWAS. Nat Genet. 2021;53:445–54.PubMedPubMedCentralCrossRef
67.
Zurück zum Zitat Zhou Y, Qiu L, Sterpka A, Wang H, Chu F, Chen X. Comparative phosphoproteomic profiling of type III adenylyl cyclase knockout and control, male, and female mice. Front Cell Neurosci. 2019;13:1–21.CrossRef Zhou Y, Qiu L, Sterpka A, Wang H, Chu F, Chen X. Comparative phosphoproteomic profiling of type III adenylyl cyclase knockout and control, male, and female mice. Front Cell Neurosci. 2019;13:1–21.CrossRef
68.
Zurück zum Zitat Hu ML, Zhang WW, Cao H, Zhang YQ. Expression pattern of type 3 adenylyl cyclase in rodent dorsal root ganglion and its primary afferent terminals. Neurosci Lett. 2019;692:16–22.PubMedCrossRef Hu ML, Zhang WW, Cao H, Zhang YQ. Expression pattern of type 3 adenylyl cyclase in rodent dorsal root ganglion and its primary afferent terminals. Neurosci Lett. 2019;692:16–22.PubMedCrossRef
70.
Zurück zum Zitat Zhang Z, Yang D, Zhan M, Zhu N, Zhou Y, Storm DR, et al. Deletion of type 3 adenylyl cyclase perturbs the postnatal maturation of olfactory sensory neurons and olfactory cilium ultrastructure in mice. Front Cell Neurosci. 2017;11:1–15.PubMedPubMedCentralCrossRef Zhang Z, Yang D, Zhan M, Zhu N, Zhou Y, Storm DR, et al. Deletion of type 3 adenylyl cyclase perturbs the postnatal maturation of olfactory sensory neurons and olfactory cilium ultrastructure in mice. Front Cell Neurosci. 2017;11:1–15.PubMedPubMedCentralCrossRef
71.
Zurück zum Zitat Wang Z, Sindreu CB, Li V, Nudelman A, Chan GCK, Storm DR. Pheromone detection in male mice depends on signaling through the type 3 adenylyl cyclase in the main olfactory epithelium. J Neurosci. 2006;26:7375–9.PubMedPubMedCentralCrossRef Wang Z, Sindreu CB, Li V, Nudelman A, Chan GCK, Storm DR. Pheromone detection in male mice depends on signaling through the type 3 adenylyl cyclase in the main olfactory epithelium. J Neurosci. 2006;26:7375–9.PubMedPubMedCentralCrossRef
73.
Zurück zum Zitat Banizs B, Pike MM, Millican CL, Ferguson WB, Komlosi P, Sheetz J, et al. Dysfunctional cilia lead to altered ependyma and choroid plexus function, and result in the formation of hydrocephalus. Development. 2005;132:5329–39.PubMedCrossRef Banizs B, Pike MM, Millican CL, Ferguson WB, Komlosi P, Sheetz J, et al. Dysfunctional cilia lead to altered ependyma and choroid plexus function, and result in the formation of hydrocephalus. Development. 2005;132:5329–39.PubMedCrossRef
75.
Zurück zum Zitat Warrington NM, Howe LD, Paternoster L, Kaakinen M, Herrala S, Huikari V, et al. A genome-wide association study of body mass index across early life and childhood. Int J Epidemiol. 2015;44:700–12.PubMedPubMedCentralCrossRef Warrington NM, Howe LD, Paternoster L, Kaakinen M, Herrala S, Huikari V, et al. A genome-wide association study of body mass index across early life and childhood. Int J Epidemiol. 2015;44:700–12.PubMedPubMedCentralCrossRef
76.
Zurück zum Zitat Stergiakouli E, Gaillard R, Tavaré JM, Balthasar N, Loos RJ, Taal HR, et al. Genome-wide association study of height-adjusted BMI in childhood identifies functional variant in ADCY3. Obesity. 2014;22:2252–9.PubMedCrossRef Stergiakouli E, Gaillard R, Tavaré JM, Balthasar N, Loos RJ, Taal HR, et al. Genome-wide association study of height-adjusted BMI in childhood identifies functional variant in ADCY3. Obesity. 2014;22:2252–9.PubMedCrossRef
78.
Zurück zum Zitat Liu JZ, van Sommeren S, Huang H, Ng SC, Alberts R, Takahashi A, et al. Association analyses identify 38 susceptibility loci for inflammatory bowel disease and highlight shared genetic risk across populations. Nat Genet. 2015;47:979–86.PubMedPubMedCentralCrossRef Liu JZ, van Sommeren S, Huang H, Ng SC, Alberts R, Takahashi A, et al. Association analyses identify 38 susceptibility loci for inflammatory bowel disease and highlight shared genetic risk across populations. Nat Genet. 2015;47:979–86.PubMedPubMedCentralCrossRef
79.
Zurück zum Zitat Hulur I, Gamazon ER, Skol AD, Xicola RM, Llor X, Onel K, et al. Enrichment of inflammatory bowel disease and colorectal cancer risk variants in colon expression quantitative trait loci. BMC Genomics. 2015;16:1–15.CrossRef Hulur I, Gamazon ER, Skol AD, Xicola RM, Llor X, Onel K, et al. Enrichment of inflammatory bowel disease and colorectal cancer risk variants in colon expression quantitative trait loci. BMC Genomics. 2015;16:1–15.CrossRef
81.
Zurück zum Zitat Kittikulsuth W, Stuart D, Kohan DE. Adenylyl cyclase 4 does not regulate collecting duct water and sodium handling. Physiol Rep. 2014;2:1–9.CrossRef Kittikulsuth W, Stuart D, Kohan DE. Adenylyl cyclase 4 does not regulate collecting duct water and sodium handling. Physiol Rep. 2014;2:1–9.CrossRef
83.
Zurück zum Zitat Kheirbek MA, Britt JP, Beeler JA, Ishikawa Y, McGehee DS, Zhuang X. Adenylyl cyclase type 5 contributes to corticostriatal plasticity and striatum-dependent learning. J Neurosci. 2009;29:12115–24.PubMedPubMedCentralCrossRef Kheirbek MA, Britt JP, Beeler JA, Ishikawa Y, McGehee DS, Zhuang X. Adenylyl cyclase type 5 contributes to corticostriatal plasticity and striatum-dependent learning. J Neurosci. 2009;29:12115–24.PubMedPubMedCentralCrossRef
86.
Zurück zum Zitat Lee KW, Hong JH, Choi IY, Che Y, Lee JK, Yang SD, et al. Impaired D2 dopamine receptor function in mice lacking type 5 adenylyl cyclase. J Neurosci. 2002;22:7931–40.PubMedPubMedCentralCrossRef Lee KW, Hong JH, Choi IY, Che Y, Lee JK, Yang SD, et al. Impaired D2 dopamine receptor function in mice lacking type 5 adenylyl cyclase. J Neurosci. 2002;22:7931–40.PubMedPubMedCentralCrossRef
88.
Zurück zum Zitat Okumura S, Kawabe JI, Yatani A, Takagi G, Lee MC, Hong C, et al. Type 5 adenylyl cyclase disruption alters not only sympathetic but also parasympathetic and calcium-mediated cardiac regulation. Circ Res. 2003;93:364–71.PubMedCrossRef Okumura S, Kawabe JI, Yatani A, Takagi G, Lee MC, Hong C, et al. Type 5 adenylyl cyclase disruption alters not only sympathetic but also parasympathetic and calcium-mediated cardiac regulation. Circ Res. 2003;93:364–71.PubMedCrossRef
89.
Zurück zum Zitat Okumura S, Vatner DE, Kurotani R, Bai Y, Gao S, Yuan Z, et al. Disruption of type 5 adenylyl cyclase enhances desensitization of cyclic adenosine monophosphate signal and increases Akt signal with chronic catecholamine stress. Circulation. 2007;116:1776–83.PubMedCrossRef Okumura S, Vatner DE, Kurotani R, Bai Y, Gao S, Yuan Z, et al. Disruption of type 5 adenylyl cyclase enhances desensitization of cyclic adenosine monophosphate signal and increases Akt signal with chronic catecholamine stress. Circulation. 2007;116:1776–83.PubMedCrossRef
90.
Zurück zum Zitat Yan L, Vatner DE, O’Connor JP, Ivessa A, Ge H, Chen W, et al. Type 5 adenylyl cyclase disruption increases longevity and protects against stress. Cell. 2007;130:247–58.PubMedCrossRef Yan L, Vatner DE, O’Connor JP, Ivessa A, Ge H, Chen W, et al. Type 5 adenylyl cyclase disruption increases longevity and protects against stress. Cell. 2007;130:247–58.PubMedCrossRef
91.
Zurück zum Zitat Vatner SF, Pachon RE, Vatner DE. Inhibition of adenylyl cyclase type 5 increases longevity and healthful aging through oxidative stress protection. Oxid Med Cell Longev. 2015;2015:250310.PubMedPubMedCentralCrossRef Vatner SF, Pachon RE, Vatner DE. Inhibition of adenylyl cyclase type 5 increases longevity and healthful aging through oxidative stress protection. Oxid Med Cell Longev. 2015;2015:250310.PubMedPubMedCentralCrossRef
93.
Zurück zum Zitat Okamoto N, Miya F, Kitai Y, Tsunoda T, Kato M, Saitoh S, et al. Homozygous ADCY5 mutation causes early-onset movement disorder with severe intellectual disability. Neurol Sci. 2021;42:2975–8.PubMedCrossRef Okamoto N, Miya F, Kitai Y, Tsunoda T, Kato M, Saitoh S, et al. Homozygous ADCY5 mutation causes early-onset movement disorder with severe intellectual disability. Neurol Sci. 2021;42:2975–8.PubMedCrossRef
94.
Zurück zum Zitat Ferrini A, Steel D, Barwick K, Kurian MA. An update on the phenotype, genotype and neurobiology of ADCY5-related disease. Mov Disord. 2021;36:1104–14.PubMedCrossRef Ferrini A, Steel D, Barwick K, Kurian MA. An update on the phenotype, genotype and neurobiology of ADCY5-related disease. Mov Disord. 2021;36:1104–14.PubMedCrossRef
96.
Zurück zum Zitat Lai NC, Roth DM, Gao MH, Tang T, Dalton N, Lai YY, et al. Intracoronary adenovirus encoding adenylyl cyclase VI increases left ventricular function in heart failure. Circulation. 2004;110:330–6.PubMedCrossRef Lai NC, Roth DM, Gao MH, Tang T, Dalton N, Lai YY, et al. Intracoronary adenovirus encoding adenylyl cyclase VI increases left ventricular function in heart failure. Circulation. 2004;110:330–6.PubMedCrossRef
97.
Zurück zum Zitat Tang T, Gao MH, Lai NC, Firth AL, Takahashi T, Guo T, et al. Adenylyl cyclase type 6 deletion decreases left ventricular function via impaired calcium handling. Circulation. 2008;117:61–9.PubMedCrossRef Tang T, Gao MH, Lai NC, Firth AL, Takahashi T, Guo T, et al. Adenylyl cyclase type 6 deletion decreases left ventricular function via impaired calcium handling. Circulation. 2008;117:61–9.PubMedCrossRef
100.
Zurück zum Zitat Laquérriere A, Maluenda J, Camus A, Fontenas L, Dieterich K, Nolent F, et al. Mutations in CNTNAP1 and ADCY6 are responsible for severe arthrogryposis multiplex congenita with axoglial defects. Hum Mol Genet. 2014;23:2279–89.PubMedCrossRef Laquérriere A, Maluenda J, Camus A, Fontenas L, Dieterich K, Nolent F, et al. Mutations in CNTNAP1 and ADCY6 are responsible for severe arthrogryposis multiplex congenita with axoglial defects. Hum Mol Genet. 2014;23:2279–89.PubMedCrossRef
101.
Zurück zum Zitat Agolini E, Cherchi C, Bellacchio E, Martinelli D, Cocciadiferro D, Cutrera R, et al. Expanding the clinical and molecular spectrum of lethal congenital contracture syndrome 8 associated with biallelic variants of ADCY6. Clin Genet. 2020;97:649–54.PubMedCrossRef Agolini E, Cherchi C, Bellacchio E, Martinelli D, Cocciadiferro D, Cutrera R, et al. Expanding the clinical and molecular spectrum of lethal congenital contracture syndrome 8 associated with biallelic variants of ADCY6. Clin Genet. 2020;97:649–54.PubMedCrossRef
103.
Zurück zum Zitat Mons N, Yoshimura M, Ikeda H, Hoffman PL, Tabakoff B. Immunological assessment of the distribution of Type VII adenylyl cyclase in brain. Brain Res. 1998;788:251–61.PubMedCrossRef Mons N, Yoshimura M, Ikeda H, Hoffman PL, Tabakoff B. Immunological assessment of the distribution of Type VII adenylyl cyclase in brain. Brain Res. 1998;788:251–61.PubMedCrossRef
104.
Zurück zum Zitat Cruz MT, Bajo M, Maragnoli ME, Tabakoff B, Siggins GR, Roberto M. Type 7 adenylyl cyclase is involved in the ethanol and CRF sensitivity of GABAergic synapses in mouse central amygdala. Front Neurosci. 2011;4:1–7.CrossRef Cruz MT, Bajo M, Maragnoli ME, Tabakoff B, Siggins GR, Roberto M. Type 7 adenylyl cyclase is involved in the ethanol and CRF sensitivity of GABAergic synapses in mouse central amygdala. Front Neurosci. 2011;4:1–7.CrossRef
105.
Zurück zum Zitat Pronko SP, Saba LM, Hoffman PL, Tabakoff B. Type 7 adenylyl cyclase-mediated hypothalamic-pituitary-adrenal axis responsiveness: influence of ethanol and sex. J Pharmacol Exp Ther. 2010;334:44–52.PubMedPubMedCentralCrossRef Pronko SP, Saba LM, Hoffman PL, Tabakoff B. Type 7 adenylyl cyclase-mediated hypothalamic-pituitary-adrenal axis responsiveness: influence of ethanol and sex. J Pharmacol Exp Ther. 2010;334:44–52.PubMedPubMedCentralCrossRef
106.
Zurück zum Zitat Jiang LI, Sternweis PC, Wang JE. Zymosan activates protein kinase A via adenylyl cyclase VII to modulate innate immune responses during inflammation. Mol Immunol. 2013;54:14–22.PubMedCrossRef Jiang LI, Sternweis PC, Wang JE. Zymosan activates protein kinase A via adenylyl cyclase VII to modulate innate immune responses during inflammation. Mol Immunol. 2013;54:14–22.PubMedCrossRef
108.
Zurück zum Zitat Luo Y, De Lange KM, Jostins L, Moutsianas L, Randall J, Kennedy NA, et al. Exploring the genetic architecture of inflammatory bowel disease by whole-genome sequencing identifies association at ADCY7. Nat Genet. 2017;49:186–92.PubMedPubMedCentralCrossRef Luo Y, De Lange KM, Jostins L, Moutsianas L, Randall J, Kennedy NA, et al. Exploring the genetic architecture of inflammatory bowel disease by whole-genome sequencing identifies association at ADCY7. Nat Genet. 2017;49:186–92.PubMedPubMedCentralCrossRef
109.
Zurück zum Zitat Li YR, Li J, Zhao SD, Bradfield JP, Mentch FD, Maggadottir SM, et al. Meta-analysis of shared genetic architecture across ten pediatric autoimmune diseases. Nat Med. 2015;21:1018–27.PubMedPubMedCentralCrossRef Li YR, Li J, Zhao SD, Bradfield JP, Mentch FD, Maggadottir SM, et al. Meta-analysis of shared genetic architecture across ten pediatric autoimmune diseases. Nat Med. 2015;21:1018–27.PubMedPubMedCentralCrossRef
110.
Zurück zum Zitat Wieczorek L, Maas JW, Muglia LM, Vogt SK, Muglia LJ. Temporal and regional regulation of gene expression by calcium-stimulated adenylyl cyclase activity during fear memory. PLoS ONE. 2010;5:e13385.PubMedPubMedCentralCrossRef Wieczorek L, Maas JW, Muglia LM, Vogt SK, Muglia LJ. Temporal and regional regulation of gene expression by calcium-stimulated adenylyl cyclase activity during fear memory. PLoS ONE. 2010;5:e13385.PubMedPubMedCentralCrossRef
111.
Zurück zum Zitat Xu H, Leinwand SG, Dell AL, Fried-Cassorla E, Raper JA. The calmodulin-stimulated adenylate cyclase ADCY8 sets the sensitivity of zebrafish retinal axons to midline repellents and is required for normal midline crossing. J Neurosci. 2010;30:7423–33.PubMedPubMedCentralCrossRef Xu H, Leinwand SG, Dell AL, Fried-Cassorla E, Raper JA. The calmodulin-stimulated adenylate cyclase ADCY8 sets the sensitivity of zebrafish retinal axons to midline repellents and is required for normal midline crossing. J Neurosci. 2010;30:7423–33.PubMedPubMedCentralCrossRef
112.
Zurück zum Zitat Naß J, Efferth T. Pharmacogenetics and pharmacotherapy of military personnel suffering from post-traumatic stress disorder. Curr Neuropharmacol. 2016;15:831–60. Naß J, Efferth T. Pharmacogenetics and pharmacotherapy of military personnel suffering from post-traumatic stress disorder. Curr Neuropharmacol. 2016;15:831–60.
114.
Zurück zum Zitat Antoni FA, Palkovits M, Simpson J, Smith SM, Leitch AL, Rosie R, et al. Ca2+/calcineurin-lnhibited adenylyl cyclase, highly abundant in forebrain regions, is important for learning and memory. J Neurosci. 1998;18:9650–61.PubMedPubMedCentralCrossRef Antoni FA, Palkovits M, Simpson J, Smith SM, Leitch AL, Rosie R, et al. Ca2+/calcineurin-lnhibited adenylyl cyclase, highly abundant in forebrain regions, is important for learning and memory. J Neurosci. 1998;18:9650–61.PubMedPubMedCentralCrossRef
116.
Zurück zum Zitat Li Y, Baldwin TA, Wang Y, Subramaniam J, Carbajal AG, Brand CS, et al. Loss of type 9 adenylyl cyclase triggers reduced phosphorylation of Hsp20 and diastolic dysfunction. Sci Rep. 2017;7:1–11. Li Y, Baldwin TA, Wang Y, Subramaniam J, Carbajal AG, Brand CS, et al. Loss of type 9 adenylyl cyclase triggers reduced phosphorylation of Hsp20 and diastolic dysfunction. Sci Rep. 2017;7:1–11.
118.
Zurück zum Zitat Huang B, Zhao J, Lei Z, Shen S, Li D, Shen GX, et al. miR-142-3p restricts cAMP production in CD4+CD25- T cells and CD4+CD25+ TREG cells by targeting AC9 mRNA. EMBO Rep. 2009;10:180–5.PubMedCrossRef Huang B, Zhao J, Lei Z, Shen S, Li D, Shen GX, et al. miR-142-3p restricts cAMP production in CD4+CD25- T cells and CD4+CD25+ TREG cells by targeting AC9 mRNA. EMBO Rep. 2009;10:180–5.PubMedCrossRef
120.
Zurück zum Zitat Kim SH, Ye YM, Lee HY, Sin HJ, Park HS. Combined pharmacogenetic effect of ADCY9 and ADRB2 gene polymorphisms on the bronchodilator response to inhaled combination therapy. J Clin Pharm Ther. 2011;36:399–405.PubMedCrossRef Kim SH, Ye YM, Lee HY, Sin HJ, Park HS. Combined pharmacogenetic effect of ADCY9 and ADRB2 gene polymorphisms on the bronchodilator response to inhaled combination therapy. J Clin Pharm Ther. 2011;36:399–405.PubMedCrossRef
121.
Zurück zum Zitat Toyota T, Hattori E, Meerabux J, Yamada K, Saito K, Shibuya H, et al. Molecular analysis, mutation screening, and association study of adenylate cyclase type 9 gene (ADCY9) in mood disorders. Am J Med Genet. 2002;114:84–92.PubMedCrossRef Toyota T, Hattori E, Meerabux J, Yamada K, Saito K, Shibuya H, et al. Molecular analysis, mutation screening, and association study of adenylate cyclase type 9 gene (ADCY9) in mood disorders. Am J Med Genet. 2002;114:84–92.PubMedCrossRef
123.
Zurück zum Zitat Choi HB, Gordon GRJ, Zhou N, Tai C, Rungta RL, Martinez J, et al. Metabolic communication between astrocytes and neurons via bicarbonate-responsive soluble adenylyl cyclase. Neuron. 2012;75:1094–104.PubMedPubMedCentralCrossRef Choi HB, Gordon GRJ, Zhou N, Tai C, Rungta RL, Martinez J, et al. Metabolic communication between astrocytes and neurons via bicarbonate-responsive soluble adenylyl cyclase. Neuron. 2012;75:1094–104.PubMedPubMedCentralCrossRef
124.
Zurück zum Zitat Wu KY, Zippin JH, Huron DR, Kamenetsky M, Hengst U, Buck J, et al. Soluble adenylyl cyclase is required for netrin-1 signaling in nerve growth cones. Nat Neurosci. 2006;9:1257–64.PubMedPubMedCentralCrossRef Wu KY, Zippin JH, Huron DR, Kamenetsky M, Hengst U, Buck J, et al. Soluble adenylyl cyclase is required for netrin-1 signaling in nerve growth cones. Nat Neurosci. 2006;9:1257–64.PubMedPubMedCentralCrossRef
125.
Zurück zum Zitat Corredor RG, Trakhtenberg EF, Pita-Thomas W, Jin X, Hu Y, Goldberg JL. Soluble adenylyl cyclase activity is necessary for retinal ganglion cell survival and axon growth. J Neurosci. 2012;32:7734–44.PubMedPubMedCentralCrossRef Corredor RG, Trakhtenberg EF, Pita-Thomas W, Jin X, Hu Y, Goldberg JL. Soluble adenylyl cyclase activity is necessary for retinal ganglion cell survival and axon growth. J Neurosci. 2012;32:7734–44.PubMedPubMedCentralCrossRef
126.
Zurück zum Zitat Chen J, Martinez J, Milner TA, Buck J, Levin LR. Neuronal expression of soluble adenylyl cyclase in the mammalian brain. Brain Res. 2013;1518:1–8.PubMedPubMedCentralCrossRef Chen J, Martinez J, Milner TA, Buck J, Levin LR. Neuronal expression of soluble adenylyl cyclase in the mammalian brain. Brain Res. 2013;1518:1–8.PubMedPubMedCentralCrossRef
127.
Zurück zum Zitat Lein ES, Hawrylycz MJ, Ao N, Ayres M, Bensinger A, Bernard A, et al. Genome-wide atlas of gene expression in the adult mouse brain. Nature. 2007;445:168–76.PubMedCrossRef Lein ES, Hawrylycz MJ, Ao N, Ayres M, Bensinger A, Bernard A, et al. Genome-wide atlas of gene expression in the adult mouse brain. Nature. 2007;445:168–76.PubMedCrossRef
130.
Zurück zum Zitat Ames A, Higashi K, Nesbett FB. Effects of Pco2 acetazolamide and ouabain on volume and composition of choroid-plexus fluid. J Physiol. 1965;181:516–24.PubMedPubMedCentralCrossRef Ames A, Higashi K, Nesbett FB. Effects of Pco2 acetazolamide and ouabain on volume and composition of choroid-plexus fluid. J Physiol. 1965;181:516–24.PubMedPubMedCentralCrossRef
131.
Zurück zum Zitat Narita K, Kawate T, Kakinuma N, Takeda S. Multiple primary cilia modulate the fluid transcytosis in choroid plexus epithelium. Traffic. 2010;11:287–301.PubMedCrossRef Narita K, Kawate T, Kakinuma N, Takeda S. Multiple primary cilia modulate the fluid transcytosis in choroid plexus epithelium. Traffic. 2010;11:287–301.PubMedCrossRef
133.
Zurück zum Zitat Reed BY, Gitomer WL, Heller HJ, Ming CH, Lemke M, Padalino P, et al. Identification and characterization of a gene with base substitutions associated with the absorptive hypercalciuria phenotype and low spinal bone density. J Clin Endocrinol Metab. 2002;87:1476–85.PubMedCrossRef Reed BY, Gitomer WL, Heller HJ, Ming CH, Lemke M, Padalino P, et al. Identification and characterization of a gene with base substitutions associated with the absorptive hypercalciuria phenotype and low spinal bone density. J Clin Endocrinol Metab. 2002;87:1476–85.PubMedCrossRef
136.
Zurück zum Zitat Pluimer BR, Colt M, Zhao Z. G Protein-coupled receptors in the mammalian blood-brain barrier. Front Cell Neurosci. 2020;14:1–9.CrossRef Pluimer BR, Colt M, Zhao Z. G Protein-coupled receptors in the mammalian blood-brain barrier. Front Cell Neurosci. 2020;14:1–9.CrossRef
138.
Zurück zum Zitat van Doorn R, Lopes Pinheiro MA, Kooij G, Lakeman K, van het Hof B, van der Pol SMA, et al. Sphingosine 1-phosphate receptor 5 mediates the immune quiescence of the human brain endothelial barrier. J Neuroinflammation. 2012;9:133.PubMedPubMedCentralCrossRef van Doorn R, Lopes Pinheiro MA, Kooij G, Lakeman K, van het Hof B, van der Pol SMA, et al. Sphingosine 1-phosphate receptor 5 mediates the immune quiescence of the human brain endothelial barrier. J Neuroinflammation. 2012;9:133.PubMedPubMedCentralCrossRef
139.
Zurück zum Zitat González-Mariscal L, Raya-Sandino A, González-González L, Hernández-Guzmán C. Relationship between G proteins coupled receptors and tight junctions. Tissue Barriers. 2018;6:1–37.CrossRef González-Mariscal L, Raya-Sandino A, González-González L, Hernández-Guzmán C. Relationship between G proteins coupled receptors and tight junctions. Tissue Barriers. 2018;6:1–37.CrossRef
140.
Zurück zum Zitat Lucaciu A, Brunkhorst R, Pfeilschifter JM, Pfeilschifter W, Subburayalu J. The S1P–S1PR axis in neurological disorders-insights into current and future therapeutic perspectives. Cells. 2020;9:1–37.CrossRef Lucaciu A, Brunkhorst R, Pfeilschifter JM, Pfeilschifter W, Subburayalu J. The S1P–S1PR axis in neurological disorders-insights into current and future therapeutic perspectives. Cells. 2020;9:1–37.CrossRef
141.
Zurück zum Zitat Spampinato SF, Merlo S, Sano Y, Kanda T, Sortino MA. Protective effect of the sphingosine-1 phosphate receptor agonist siponimodon disrupted blood brain barrier function. Biochem Pharmacol. 2021;186:114465.PubMedCrossRef Spampinato SF, Merlo S, Sano Y, Kanda T, Sortino MA. Protective effect of the sphingosine-1 phosphate receptor agonist siponimodon disrupted blood brain barrier function. Biochem Pharmacol. 2021;186:114465.PubMedCrossRef
142.
Zurück zum Zitat Yanagida K, Liu CH, Faraco G, Galvani S, Smith HK, Burg N, et al. Size-selective opening of the blood–brain barrier by targeting endothelial sphingosine 1-phosphate receptor 1. Proc Natl Acad Sci USA. 2017;114:4531–6.PubMedPubMedCentralCrossRef Yanagida K, Liu CH, Faraco G, Galvani S, Smith HK, Burg N, et al. Size-selective opening of the blood–brain barrier by targeting endothelial sphingosine 1-phosphate receptor 1. Proc Natl Acad Sci USA. 2017;114:4531–6.PubMedPubMedCentralCrossRef
143.
Zurück zum Zitat Cannon RE, Peart JC, Hawkins BT, Campos CR, Miller DS. Targeting blood-brain barrier sphingolipid signaling reduces basal P-glycoprotein activity and improves drug delivery to the brain. Proc Natl Acad Sci USA. 2012;109:15930–5.PubMedPubMedCentralCrossRef Cannon RE, Peart JC, Hawkins BT, Campos CR, Miller DS. Targeting blood-brain barrier sphingolipid signaling reduces basal P-glycoprotein activity and improves drug delivery to the brain. Proc Natl Acad Sci USA. 2012;109:15930–5.PubMedPubMedCentralCrossRef
144.
Zurück zum Zitat Cartwright TA, Campos CR, Cannon RE, Miller DS. Mrp1 is essential for sphingolipid signaling to p-glycoprotein in mouse blood-brain and blood-spinal cord barriers. J Cereb Blood Flow Metab. 2013;13:381–8.CrossRef Cartwright TA, Campos CR, Cannon RE, Miller DS. Mrp1 is essential for sphingolipid signaling to p-glycoprotein in mouse blood-brain and blood-spinal cord barriers. J Cereb Blood Flow Metab. 2013;13:381–8.CrossRef
146.
Zurück zum Zitat Xu D, Gao Q, Wang F, Peng Q, Wang G, Wei Q, et al. Sphingosine-1-phosphate receptor 3 is implicated in BBB injury via the CCL2-CCR2 axis following acute intracerebral hemorrhage. CNS Neurosci Ther. 2021;27:674–86.PubMedPubMedCentralCrossRef Xu D, Gao Q, Wang F, Peng Q, Wang G, Wei Q, et al. Sphingosine-1-phosphate receptor 3 is implicated in BBB injury via the CCL2-CCR2 axis following acute intracerebral hemorrhage. CNS Neurosci Ther. 2021;27:674–86.PubMedPubMedCentralCrossRef
148.
Zurück zum Zitat Zhang Y, Chen K, Sloan SA, Bennett ML, Scholze AR, O’Keeffe S, et al. Brain RNA-Seq. An RNA-sequencing Transcr. splicing database glia, neurons, Vasc Cells Cereb Cortex. 2014, p. 11929–47. https://brainrnaseq.org/. Accessed 28 Dec 2021. Zhang Y, Chen K, Sloan SA, Bennett ML, Scholze AR, O’Keeffe S, et al. Brain RNA-Seq. An RNA-sequencing Transcr. splicing database glia, neurons, Vasc Cells Cereb Cortex. 2014, p. 11929–47. https://​brainrnaseq.​org/​. Accessed 28 Dec 2021.
150.
Zurück zum Zitat Masago K, Kihara Y, Yanagida K, Hamano F, Nakagawa S, Niwa M, et al. Lysophosphatidic acid receptor, LPA6, regulates endothelial blood-brain barrier function: implication for hepatic encephalopathy. Biochem Biophys Res Commun. 2018;501:1048–54.PubMedPubMedCentralCrossRef Masago K, Kihara Y, Yanagida K, Hamano F, Nakagawa S, Niwa M, et al. Lysophosphatidic acid receptor, LPA6, regulates endothelial blood-brain barrier function: implication for hepatic encephalopathy. Biochem Biophys Res Commun. 2018;501:1048–54.PubMedPubMedCentralCrossRef
151.
Zurück zum Zitat On NH, Savant S, Toews M, Miller DW. Rapid and reversible enhancement of blood–brain barrier permeability using lysophosphatidic acid. J Cereb Blood Flow Metab. 2013;33:1944–54.PubMedPubMedCentralCrossRef On NH, Savant S, Toews M, Miller DW. Rapid and reversible enhancement of blood–brain barrier permeability using lysophosphatidic acid. J Cereb Blood Flow Metab. 2013;33:1944–54.PubMedPubMedCentralCrossRef
152.
Zurück zum Zitat Banks DB, Chan GNY, Evans RA, Miller DS, Cannon RE. Lysophosphatidic acid and amitriptyline signal through LPA1R to reduce P-glycoprotein transport at the blood–brain barrier. J Cereb Blood Flow Metab. 2018;38:857–68.PubMedCrossRef Banks DB, Chan GNY, Evans RA, Miller DS, Cannon RE. Lysophosphatidic acid and amitriptyline signal through LPA1R to reduce P-glycoprotein transport at the blood–brain barrier. J Cereb Blood Flow Metab. 2018;38:857–68.PubMedCrossRef
155.
Zurück zum Zitat Bhattarai S, Sharma S, Ara H, Subedi U, Sun G, Li C, et al. Disrupted blood-brain barrier and mitochondrial impairment by autotaxin–lysophosphatidic acid axis in postischemic stroke. J Am Heart Assoc. 2021;10:e021511.PubMedPubMedCentralCrossRef Bhattarai S, Sharma S, Ara H, Subedi U, Sun G, Li C, et al. Disrupted blood-brain barrier and mitochondrial impairment by autotaxin–lysophosphatidic acid axis in postischemic stroke. J Am Heart Assoc. 2021;10:e021511.PubMedPubMedCentralCrossRef
157.
Zurück zum Zitat Ramirez SH, Haskó J, Skuba A, Fan S, Dykstra H, McCormick R, et al. Activation of cannabinoid receptor 2 attenuates leukocyte-endothelial cell interactions and blood-brain barrier dysfunction under inflammatory conditions. J Neurosci. 2012;32:4004–16.PubMedPubMedCentralCrossRef Ramirez SH, Haskó J, Skuba A, Fan S, Dykstra H, McCormick R, et al. Activation of cannabinoid receptor 2 attenuates leukocyte-endothelial cell interactions and blood-brain barrier dysfunction under inflammatory conditions. J Neurosci. 2012;32:4004–16.PubMedPubMedCentralCrossRef
158.
Zurück zum Zitat Rhee MH, Bayewitch M, Avidor-Reiss T, Levy R, Vogel Z. Cannabinoid receptor activation differentially regulates the various adenylyl cyclase isozymes. J Neurochem. 1998;71:1525–34.PubMedCrossRef Rhee MH, Bayewitch M, Avidor-Reiss T, Levy R, Vogel Z. Cannabinoid receptor activation differentially regulates the various adenylyl cyclase isozymes. J Neurochem. 1998;71:1525–34.PubMedCrossRef
159.
Zurück zum Zitat Bonhaus DW, Chang LK, Kwan J, Martin GR. Dual activation and inhibition of adenylyl cyclase by cannabinoid receptor agonists: evidence for agonist-specific trafficking of intracellular responses. J Pharmacol Exp Ther. 1998;287:884–8.PubMed Bonhaus DW, Chang LK, Kwan J, Martin GR. Dual activation and inhibition of adenylyl cyclase by cannabinoid receptor agonists: evidence for agonist-specific trafficking of intracellular responses. J Pharmacol Exp Ther. 1998;287:884–8.PubMed
161.
162.
Zurück zum Zitat Bynoe MS, Viret C, Yan A, Kim DG. Adenosine receptor signaling: a key to opening the blood–brain door. Fluids Barriers CNS BioMed Central. 2015;12:1–12. Bynoe MS, Viret C, Yan A, Kim DG. Adenosine receptor signaling: a key to opening the blood–brain door. Fluids Barriers CNS BioMed Central. 2015;12:1–12.
163.
Zurück zum Zitat Carman AJ, Mills JH, Krenz A, Kim DG, Bynoe MS. Adenosine receptor signaling modulates permeability of the blood–brain barrier. J Neurosci. 2011;31:13272–80.PubMedPubMedCentralCrossRef Carman AJ, Mills JH, Krenz A, Kim DG, Bynoe MS. Adenosine receptor signaling modulates permeability of the blood–brain barrier. J Neurosci. 2011;31:13272–80.PubMedPubMedCentralCrossRef
164.
Zurück zum Zitat Gao X, Qian J, Zheng S, Changyi Y, Zhang J, Ju S, et al. Overcoming the blood-brain barrier for delivering drugs into the brain by using adenosine receptor nanoagonist. ACS Nano. 2014;8:3678–89.PubMedCrossRef Gao X, Qian J, Zheng S, Changyi Y, Zhang J, Ju S, et al. Overcoming the blood-brain barrier for delivering drugs into the brain by using adenosine receptor nanoagonist. ACS Nano. 2014;8:3678–89.PubMedCrossRef
165.
Zurück zum Zitat Kim DG, Bynoe MS. A2A adenosine receptor modulates drug efflux transporter P-glycoprotein at the blood-brain barrier. J Clin Invest. 2016;126:1717–33.PubMedPubMedCentralCrossRef Kim DG, Bynoe MS. A2A adenosine receptor modulates drug efflux transporter P-glycoprotein at the blood-brain barrier. J Clin Invest. 2016;126:1717–33.PubMedPubMedCentralCrossRef
166.
Zurück zum Zitat Isensee J, Meoli L, Zazzu V, Nabzdyk C, Witt H, Soewarto D, et al. Expression pattern of G protein-coupled receptor 30 in LacZ reporter mice. Endocrinology. 2009;150:1722–3.PubMedCrossRef Isensee J, Meoli L, Zazzu V, Nabzdyk C, Witt H, Soewarto D, et al. Expression pattern of G protein-coupled receptor 30 in LacZ reporter mice. Endocrinology. 2009;150:1722–3.PubMedCrossRef
167.
Zurück zum Zitat Wei W, Chen ZJ, Zhang KS, Yang XL, Wu YM, Chen XH, et al. The activation of G protein-coupled receptor 30 (GPR30) inhibits proliferation of estrogen receptornegative breast cancer cells in vitro and in vivo. Cell Death Dis. 2014;5:e1428.PubMedPubMedCentralCrossRef Wei W, Chen ZJ, Zhang KS, Yang XL, Wu YM, Chen XH, et al. The activation of G protein-coupled receptor 30 (GPR30) inhibits proliferation of estrogen receptornegative breast cancer cells in vitro and in vivo. Cell Death Dis. 2014;5:e1428.PubMedPubMedCentralCrossRef
169.
Zurück zum Zitat Sayah S, Jauneau AC, Patte C, Tonon MC, Vaudry H, Fontaine M. Two different transduction pathways are activated by C3a and C5a anaphylatoxins on astrocytes. Mol Brain Res. 2003;112:53–60.PubMedCrossRef Sayah S, Jauneau AC, Patte C, Tonon MC, Vaudry H, Fontaine M. Two different transduction pathways are activated by C3a and C5a anaphylatoxins on astrocytes. Mol Brain Res. 2003;112:53–60.PubMedCrossRef
170.
Zurück zum Zitat Brandolini L, Grannonico M, Bianchini G, Colanardi A, Sebastiani P, Paladini A, et al. The novel C5aR antagonist DF3016A protects neurons against ischemic neuroinflammatory injury. Neurotox Res. 2019;36:163–74.PubMedPubMedCentralCrossRef Brandolini L, Grannonico M, Bianchini G, Colanardi A, Sebastiani P, Paladini A, et al. The novel C5aR antagonist DF3016A protects neurons against ischemic neuroinflammatory injury. Neurotox Res. 2019;36:163–74.PubMedPubMedCentralCrossRef
171.
Zurück zum Zitat Mahajan SD, Parikh NU, Woodruff TM, Jarvis JN, Lopez M, Hennon T, et al. C5a alters blood-brain barrier integrity in a human in vitro model of systemic lupus erythematosus. Immunology. 2015;146:130–43.PubMedPubMedCentralCrossRef Mahajan SD, Parikh NU, Woodruff TM, Jarvis JN, Lopez M, Hennon T, et al. C5a alters blood-brain barrier integrity in a human in vitro model of systemic lupus erythematosus. Immunology. 2015;146:130–43.PubMedPubMedCentralCrossRef
172.
Zurück zum Zitat Cakir M, Dworakowska D, Grossman A. Somatostatin receptor biology in neuroendocrine and pituitary tumours: Part 1—molecular pathways. J Cell Mol Med. 2010;14:2570–84.PubMedPubMedCentralCrossRef Cakir M, Dworakowska D, Grossman A. Somatostatin receptor biology in neuroendocrine and pituitary tumours: Part 1—molecular pathways. J Cell Mol Med. 2010;14:2570–84.PubMedPubMedCentralCrossRef
175.
Zurück zum Zitat Shan Y, Tan S, Lin Y, Liao S, Zhang B, Chen X, et al. The glucagon-like peptide-1 receptor agonist reduces inflammation and blood–brain barrier breakdown in an astrocyte-dependent manner in experimental stroke. J Neuroinflamm J. 2019;16:1–20. Shan Y, Tan S, Lin Y, Liao S, Zhang B, Chen X, et al. The glucagon-like peptide-1 receptor agonist reduces inflammation and blood–brain barrier breakdown in an astrocyte-dependent manner in experimental stroke. J Neuroinflamm J. 2019;16:1–20.
179.
Zurück zum Zitat Morland C, Lauritzen KH, Puchades M, Holm-Hansen S, Andersson K, Gjedde A, et al. The lactate receptor, G-protein-coupled receptor 81/hydroxycarboxylic acid receptor 1: expression and action in brain. J Neurosci Res. 2015;93:1045–55.PubMedCrossRef Morland C, Lauritzen KH, Puchades M, Holm-Hansen S, Andersson K, Gjedde A, et al. The lactate receptor, G-protein-coupled receptor 81/hydroxycarboxylic acid receptor 1: expression and action in brain. J Neurosci Res. 2015;93:1045–55.PubMedCrossRef
180.
Zurück zum Zitat Boitsova EB, Morgun AV, Osipova ED, Pozhilenkova EA, Martinova GP, Frolova OV, et al. The inhibitory effect of LPS on the expression of GPR81 lactate receptor in blood–brain barrier model in vitro. J Neuroinflamm. 2018;15:1–7.CrossRef Boitsova EB, Morgun AV, Osipova ED, Pozhilenkova EA, Martinova GP, Frolova OV, et al. The inhibitory effect of LPS on the expression of GPR81 lactate receptor in blood–brain barrier model in vitro. J Neuroinflamm. 2018;15:1–7.CrossRef
182.
Zurück zum Zitat Khilazheva ED, Pisareva NV, Morgun AV, Boitsova EB, Taranushenko TE, Frolova OV, et al. Activation of GPR81 lactate receptors stimulates mitochondrial biogenesis in cerebral microvessel endothelial cells. Ann Clin Exp Neurol. 2017;1:34–9. Khilazheva ED, Pisareva NV, Morgun AV, Boitsova EB, Taranushenko TE, Frolova OV, et al. Activation of GPR81 lactate receptors stimulates mitochondrial biogenesis in cerebral microvessel endothelial cells. Ann Clin Exp Neurol. 2017;1:34–9.
183.
Zurück zum Zitat Bravo CA, Vatner DE, Pachon R, Zhang J, Vatner SF. A food and drug administration-approved antiviral agent that inhibits adenylyl cyclase type 5 protects the ischemic heart even when administered after reperfusion. J Pharmacol Exp Ther. 2016;357:331–6.PubMedPubMedCentralCrossRef Bravo CA, Vatner DE, Pachon R, Zhang J, Vatner SF. A food and drug administration-approved antiviral agent that inhibits adenylyl cyclase type 5 protects the ischemic heart even when administered after reperfusion. J Pharmacol Exp Ther. 2016;357:331–6.PubMedPubMedCentralCrossRef
186.
Zurück zum Zitat Zaccolo M, De Giorgi F, Cho CY, Feng L, Knapp T, Negulescu PA, et al. A genetically encoded, flourescent indicator for cyclic AMP in living cells. Nat Cell Biol. 2000;2:25–9.PubMedCrossRef Zaccolo M, De Giorgi F, Cho CY, Feng L, Knapp T, Negulescu PA, et al. A genetically encoded, flourescent indicator for cyclic AMP in living cells. Nat Cell Biol. 2000;2:25–9.PubMedCrossRef
187.
Zurück zum Zitat Ponsioen B, Zhao J, Riedl J, Zwartkruis F, van der Krogt G, Zaccolo M, et al. Detecting cAMP-induced Epac activation by fluorescence resonance energy transfer: Epac as a novel cAMP indicator. EMBO Rep. 2004;5:1176–80.PubMedPubMedCentralCrossRef Ponsioen B, Zhao J, Riedl J, Zwartkruis F, van der Krogt G, Zaccolo M, et al. Detecting cAMP-induced Epac activation by fluorescence resonance energy transfer: Epac as a novel cAMP indicator. EMBO Rep. 2004;5:1176–80.PubMedPubMedCentralCrossRef
188.
Zurück zum Zitat Agarwal SR, Fiore C, Miyashiro K, Ostrom RS, Harvey RD. Effect of adenylyl cyclase type 6 on localized production of cAMP by b-2 adrenoceptors in human airway smooth-muscle cells. J Pharmacol Exp Ther. 2019;370:104–10.PubMedPubMedCentralCrossRef Agarwal SR, Fiore C, Miyashiro K, Ostrom RS, Harvey RD. Effect of adenylyl cyclase type 6 on localized production of cAMP by b-2 adrenoceptors in human airway smooth-muscle cells. J Pharmacol Exp Ther. 2019;370:104–10.PubMedPubMedCentralCrossRef
189.
Zurück zum Zitat Agarwal SR, Miyashiro K, Latt H, Ostrom RS, Harvey RD. Compartmentalized cAMP responses to prostaglandin EP2 receptor activation in human airway smooth muscle cells. Br J Pharmacol. 2017;174:2784–96.PubMedPubMedCentralCrossRef Agarwal SR, Miyashiro K, Latt H, Ostrom RS, Harvey RD. Compartmentalized cAMP responses to prostaglandin EP2 receptor activation in human airway smooth muscle cells. Br J Pharmacol. 2017;174:2784–96.PubMedPubMedCentralCrossRef
190.
Zurück zum Zitat Grarup N, Moltke I, Andersen MK, Dalby M, Vitting-Seerup K, Kern T, et al. Loss-of-function variants in ADCY3 increase risk of obesity and type 2 diabetes. Nat Genet. 2018;50:172–4.PubMedPubMedCentralCrossRef Grarup N, Moltke I, Andersen MK, Dalby M, Vitting-Seerup K, Kern T, et al. Loss-of-function variants in ADCY3 increase risk of obesity and type 2 diabetes. Nat Genet. 2018;50:172–4.PubMedPubMedCentralCrossRef
Metadaten
Titel
Expression and functions of adenylyl cyclases in the CNS
verfasst von
Karan Devasani
Yao Yao
Publikationsdatum
01.12.2022
Verlag
BioMed Central
Erschienen in
Fluids and Barriers of the CNS / Ausgabe 1/2022
Elektronische ISSN: 2045-8118
DOI
https://doi.org/10.1186/s12987-022-00322-2

Weitere Artikel der Ausgabe 1/2022

Fluids and Barriers of the CNS 1/2022 Zur Ausgabe

Leitlinien kompakt für die Neurologie

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Nicht Creutzfeldt Jakob, sondern Abführtee-Vergiftung

29.05.2024 Hyponatriämie Nachrichten

Eine ältere Frau trinkt regelmäßig Sennesblättertee gegen ihre Verstopfung. Der scheint plötzlich gut zu wirken. Auf Durchfall und Erbrechen folgt allerdings eine Hyponatriämie. Nach deren Korrektur kommt es plötzlich zu progredienten Kognitions- und Verhaltensstörungen.

Schutz der Synapsen bei Alzheimer

29.05.2024 Morbus Alzheimer Nachrichten

Mit einem Neurotrophin-Rezeptor-Modulator lässt sich möglicherweise eine bestehende Alzheimerdemenz etwas abschwächen: Erste Phase-2-Daten deuten auf einen verbesserten Synapsenschutz.

Sozialer Aufstieg verringert Demenzgefahr

24.05.2024 Demenz Nachrichten

Ein hohes soziales Niveau ist mit die beste Versicherung gegen eine Demenz. Noch geringer ist das Demenzrisiko für Menschen, die sozial aufsteigen: Sie gewinnen fast zwei demenzfreie Lebensjahre. Umgekehrt steigt die Demenzgefahr beim sozialen Abstieg.

Hirnblutung unter DOAK und VKA ähnlich bedrohlich

17.05.2024 Direkte orale Antikoagulanzien Nachrichten

Kommt es zu einer nichttraumatischen Hirnblutung, spielt es keine große Rolle, ob die Betroffenen zuvor direkt wirksame orale Antikoagulanzien oder Marcumar bekommen haben: Die Prognose ist ähnlich schlecht.

Update Neurologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.