Skip to main content
Erschienen in: Clinical Autonomic Research 1/2024

Open Access 23.02.2024 | Review Article

The relevance of the superior cervical ganglion for cardiac autonomic innervation in health and disease: a systematic review

verfasst von: H. Sophia Chen, Lieke van Roon, Yang Ge, Janine M. van Gils, Jan W. Schoones, Marco C. DeRuiter, Katja Zeppenfeld, Monique R. M. Jongbloed

Erschienen in: Clinical Autonomic Research | Ausgabe 1/2024

Abstract

Purpose

The heart receives cervical and thoracic sympathetic contributions. Although the stellate ganglion is considered the main contributor to cardiac sympathetic innervation, the superior cervical ganglia (SCG) is used in many experimental studies. The clinical relevance of the SCG to cardiac innervation is controversial. We investigated current morphological and functional evidence as well as controversies on the contribution of the SCG to cardiac innervation.

Methods

A systematic literature review was conducted in PubMed, Embase, Web of Science, and COCHRANE Library. Included studies received a full/text review and quality appraisal.

Results

Seventy-six eligible studies performed between 1976 and 2023 were identified. In all species studied, morphological evidence of direct or indirect SCG contribution to cardiac innervation was found, but its contribution was limited. Morphologically, SCG sidedness may be relevant. There is indirect functional evidence that the SCG contributes to cardiac innervation as shown by its involvement in sympathetic overdrive reactions in cardiac disease states. A direct functional contribution was not found. Functional data on SCG sidedness was largely unavailable. Information about sex differences and pre- and postnatal differences was lacking.

Conclusion

Current literature mainly supports an indirect involvement of the SCG in cardiac innervation, via other structures and plexuses or via sympathetic overdrive in response to cardiac diseases. Morphological evidence of a direct involvement was found, but its contribution seems limited. The relevance of SCG sidedness, sex, and developmental stage in health and disease remains unclear and warrants further exploration.

Graphical abstract

An overview of the current literature derived from morphological and functional data on the involvement of SCG in cardiac innervation, relevance of sidedness, sex differences, and pre- and postnatal differences in various species. X = Information not available
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1007/​s10286-024-01019-2.

Introduction

A balanced function of the cardiac autonomic nervous system is essential to maintain cardiovascular homeostasis. Cardiac innervation is provided by the autonomic nervous system, which is organized into sympathetic and parasympathetic branches. Balancing sympathetic and parasympathetic tone is mandatory to maintain a regular heartbeat. Parasympathetic innervation of the heart is provided by preganglionic branches of the vagal nerve that synapse close to the target organ, e.g., in intrinsic cardiac ganglia situated in the myocardial wall and on the epicardial surface of the heart [1]. For sympathetic innervation, preganglionic sympathetic axons synapse with sympathetic neurons in the sympathetic chain, after which postganglionic fibers directly innervate either the myocardium or first synapse on the intrinsic cardiac ganglia [1]. In humans, this innervation from the sympathetic chain is likely provided by both cervical and thoracic ganglia, although the exact level of ganglia contributing to the heart is still controversial [2].
Interest in cardiac autonomic innervation has increased in the past decades, as alterations in cardiac innervation, both morphologically and functionally, after cardiac damage have been reported in myriad studies [36]. An especially intriguing phenomenon is the so-called cardiac sympathetic hyperinnervation, which can occur after cardiac damage, such as myocardial infarction (MI) [7, 8]. This hyperinnervation is characterized by an increased amount of sympathetic nerve fibers in the area of damage and has been related to ventricular arrhythmias and sudden cardiac death after MI [6]. Although several excellent mechanistic studies have been performed, the exact underlying relation between the occurrence of sympathetic hyperinnervation and ventricular arrhythmias after MI is still being determined. Apparently sympathetic ganglia, renowned for their limited growth potential after birth, retrieve their potential for fast outgrowth after cardiac damage. These findings have prompted researchers to study cardiac innervation in health and disease.
The stellate ganglia, consisting of the fused inferior cervical ganglion with the first thoracic ganglion, are most renowned for their contribution to cardiac innervation. They are located deeper in the thorax anterior to the first rib (Fig. 1). Although the stellate ganglion is generally accepted to provide a majority of cardiac innervation, other ganglia have been proposed to contribute to health and disease as well, including the thoracic and cervical ganglia [2, 9]. Cervical ganglia have indeed been shown to contribute to cardiac innervation both in animal models as well as in human, however reports in literature differ [2]. These ganglia also contribute to innervation of other structures in the head and neck, including the iris, jaw submandibular gland, the pineal gland, and the carotid body [1012]. One of the cervical ganglia is the superior cervical ganglion (SCG) (Fig. 1), which is in close spatial orientation with the carotid body, a chemoreceptor-sensitive organ that can respond to changes in blood oxygen, carbon dioxide, and pH levels, as well as with the ganglion nodosum, the inferior ganglion of the vagal nerve [13, 14]. From a basic science point of view, the relatively good accessibility of the SCG, which is located at a specific anatomical landmark location, at the bifurcation of the common carotid arteries, provides advantages when using this structure as an experimental model to study cardiac (hyper)innervation. A prerequisite for using the SCG to study cardiac autonomic function is that this structure provides a significant anatomical and/or functional contribution to cardiac innervation.
Consistent differences have been shown in cardiac autonomic regulation between women and men, such as a more pronounced parasympathetic cardiac regulation, higher resting heart rate, and lower baroflex sensitivity in women, although its physiologic usefulness remains largely unknown [15]. Another factor that could possible influence the anatomical and functional contribution to cardiac innervation is the sidedness of the ganglia. The human peripheral cardiac autonomic nervous system shows considerable asymmetry, interindividual variations and regional differences in anatomical, functional, and molecular characteristics [16]. Moreover, MI has been shown to induce morphologic and neurochemical changes in right- and left-sided ganglia [5, 17].
In this review, we aim to systematically investigate current morphological and functional evidence, as well as to expose current controversies and gaps in knowledge, on the contribution of the SCG to cardiac innervation in health and disease in human and other animal models, including the consideration of potentially relevant aspects such as sex and sidedness.

Methods

Research questions

To perform a comprehensive systematic review of current knowledge on the contribution of the SCG in cardiac autonomic innervation, our research questions were defined as follows:
1.
What is the morphological and functional evidence that the SCG is involved in cardiac innervation in various species, in health as well as in cardiac disease?
(a)
Is sidedness relevant (e.g., using left- or right-sided SCG) to study cardiac innervation?
 
(b)
Have sex differences been studied and/or encountered?
 
(c)
Are there pre- and postnatal/adult differences?
 
(d)
What is the quality of the included studies?
 
 
2.
Which controversies are encountered, and which questions are potentially unanswered by current data?
 
3.
And finally, derived from these data: Is the use of the SCG in experimental setting an adequate structure to study cardiac innervation in health and disease?
 

Search strategy

This systematic review was conducted in PubMed, Embase, Web of Science, and COCHRANE Library databases up to 4 April 2023 and was in adherence with the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) statement guidelines [18], using the same workflow as in previously published systematic reviews [2, 19, 20]. The search strategy was conducted by using keywords for superior cervical ganglion, heart, innervation, and nerve growth factor. The full search strategy for each respective database can be viewed in the Supplementary Materials (Appendix A).

Selection criteria

Papers were considered eligible to be included in this systematic review when both the SCG and the heart were studied. Both studies in health and in disease models were included. Additionally, all animal species including humans, both prenatal and postnatal/adult studies, were included. Functional studies, in vivo as well as in vitro, were included as long as an interdependent effect of the SCG on the heart or vice versa was studied. Papers in which the presence of a direct/causal link between the SCG and the heart was not studied were excluded, e.g., when the effect of certain substances was studied separately on the heart and SCG or when immunostaining was performed on the SCG and/or heart without an intervention to the opposing tissue. Only original research papers were included; reviews, editorials, and book chapters were excluded. Other reasons to exclude the paper were: non-English-language papers, papers that could not be retrieved after significant effort, and papers that were published in a predatory journal [21]. With regard to the date of publication, articles published before 1975 were excluded, partly owing to the difficulties in retrieving several older papers. The reference lists of included papers were searched for eligible articles that were not identified with the query.

Data extraction and quality appraisal

All records were screened for eligibility by two independent authors (H.S.C. and M.R.M.J.) on the basis of titles and abstracts followed by full-text review where necessary, considering the selection criteria described above.
Selected papers were categorized into (1) morphological, (2) functional in vivo, or (3) functional in vitro. In case multiple categories were applicable, the paper was included in all categories and only data relating to the category were extracted and assessed for quality. Anterograde or retrograde labeling studies were considered as morphological studies.
When available, the following data were extracted independently by two authors (H.S.C. and L.V.R.): author and year of publication; objectives; number; species; strain/breed; genotype; age; weight; sex; study type; data on sidedness; experimental condition; experimental setting; solution and staining or histochemistry; SCG-heart-related outcome; limitations. Discrepancies between the observers’ judgments were resolved by discussion and consensus.
The methodological quality of the morphological studies was scored by two authors (H.S.C. and M.R.M.J.) using the Quality Appraisal for Cadaveric Studies (QUACS) scale [22]. Similarly, the methodological quality of the functional studies was scored by two authors (H.S.C. and L.V.R.) using the Animal Research: Reporting In Vivo Experiments (ARRIVE) guidelines 2.0 [23] for the in vivo studies and an adapted version for the in vitro studies (Supplementary Materials, Appendix B) as no existing quality assessment tool currently covers all critical aspects of in vitro studies [24].
After comparing and discussing the individual scores for each paper, a consensus score was achieved for all papers included in this review.
Note that all extracted data and quality assessments were solely based on aspects concerning the relationship between the SCG and the heart.

Results

Study selection and inclusion

A total of 591 records were identified through database searching (Fig. 2). Of these, 163 records were duplicates and the number of studies excluded by abstract screening was 363; most of these did not study both SCG and the heart or lacked an evident study of the causal relationship between the SCG and cardiac tissues. The remaining 65 records were screened based on the full-text articles. Then, 8 records were excluded after full-text assessment and 19 additional records were found through cited references in the remaining articles. A total of 76 studies were included in this systematic review. As some articles fit multiple categories, 26 studies were categorized as morphological, 25 as functional in vivo, and 32 as functional in vitro. Table 1 provides an overview of the studies included in the morphology and functional categories of this systematic review.
Table 1
Overview of the studies included in the systematic review
Morphology
Functional
 
Author
Year
Species
In vivo
In vitro
No.
Author
Year
Species
No.
Author
Year
Species
1
Kirby et al.
1980
Chick
1
Brandys et al.
1984
Dog
1
Furshpan et al.
1976
Rat
2
Armour and Hopkins
1981
Dog
2
Zhang et al.
2007
Rat
2
Landis
1976
Rat
3
Billman et al.
1982
Nonhuman primate
3
Li et al.**
2010
Rat
3
Chun and Patterson
1977
Rat
4
Hopkins and Armour
1984
Dog
4
Li et al.
2011
Rat
4
King et al.
1978
Rat
5
Shih et al.
1985
Cat
5
Liu et al.*
2013
Rat
5
Schwab and Landis
1981
Rat
6
Janes et al.
1986
Human
6
Kong et al.**
2013
Rat
6
Coughlin et al.
1981
Mouse
7
Wu et al.
1988
Cat
7
Liu et al.**
2014
Rat
7
Coughlin and Kessler
1982
Mouse
8
Pardini et al.
1989
Rat
8
Na et al.
2014
Rat
8
De Ridder and De Potter
1983
Rat
9
Chuang et al.
1992
Nonhuman primate
9
Zhang et al.
2015
Rat
9
Rawdon and Dockray
1983
Mouse
10
Hirakawa et al.
1993
Dog
10
Tu et al.
2016
Rat
10
Kessler et al.
1984
Rat
11
Verberne et al.
1999
Chick/Quail
11
Wu et al.
2016
Rat
11
Uchida and Tomonaga
1985
Mouse
12
Pather et al.
2003
Human
12
Xu et al.
2016
Rat
12
Lahtinen et al.
1986
Rat
13
Chuang et al.
2004
Nonhuman primate
13
Zou et al.
2017
Rat
13
Furshpan et al. (1)
1986
Rat
14
Kawashima and Sasaki
2005
Human
14
Liu et al.
2018
Rat
14
Furshpan et al. (2)
1986
Rat
15
Kawashima
2005
Human
15
Yu et al.*
2018
Rat
15
Potter et al.
1986
Rat
16
Kawashima et al.
2005
Nonhuman primate
16
Ziegler et al.
2018
Mouse
16
Matsumoto et al.
1987
Rat
17
Li et al.
2006
Guinea Pig
17
Cheng et al.**
2018
Rabbit
17
Conforti et al.
1991
Rat
18
Tanaka et al.
2007
Shrew
18
Shi et al.
2019
Rat
18
Rawdon
1991
Mouse
19
Kawashima and Sasaki
2007
Human
19
Prado et al.
2020
Rat
19
Kannan et al.
1994
Rat
20
Kawashima et al.
2008
Nonhuman primate
20
Zhang et al.
2020
Rat
20
Lockhart et al.
1997
Rat
21
Kawashima et al.
2009
Nonhuman primate
21
Zou et al.
2022
Rat
21
Ulupinar et al.
1998
Rat
22
Kawashima and Thorington
2011
Nonhuman primate
22
Ge et al.
2022
Mouse
22
Hasan et al.
2006
Rat
23
Kawashima et al.
2013
Nonhuman primate
23
Cheng et al.**
2023
Rabbit
23
Shcherbakova et al.
2007
Mouse
24
Liu et al.*
2013
Rat
24
Zhang et al.
2023
Rat
24
Li et al.**
2010
Rat
25
Manousiouthakis et al.
2014
Mouse
    
25
Miwa et al.
2010
Rat
26
Yu et al.*
2018
Rat
    
26
Takeuchi et al.
2011
Rat
        
27
Kong et al.**
2013
Rat
        
28
Miwa et al.
2013
Rat
        
29
Liu et al.**
2014
Rat
        
30
Cheng et al.**
2018
Rabbit
        
31
Ge et al.
2020
Mouse
        
32
Cheng et al.**
2023
Rabbit
*Morphological and functional in vivo
**Functional in vivo and in vitro

Quality assessment

Quality scores assessed by QUACS scale ranged from 42% to 65% in morphological studies with a median of 56% (interquartile range 50–61%). In the majority of morphological studies, a thorough description of methods and results was presented, either with or without details on consistency of data with regard to number or percentages of cases in which observations were made (Fig. 3A). In addition, most studies adequately discussed findings in the context of contemporary evidence. Most studies adequately supported their data description with photographs and/or drawings. The relatively low scores could be attributed mainly to deficient data on statistics, education level of dissecting researchers, and the number of observers, which was lacking in all of the included studies. In addition, distinctly indicated study limitations and clinical implications were lacking in most morphological manuscripts.
Applying the ARRIVE guidelines for scoring the functional in vivo studies resulted in quality scores between 43% and 81% with a median of 64% (interquartile range 62–67%). Most detailed the study design, background, objectives, outcome measures, experimental procedures, results, and declaration of interests (Fig. 3B). In contrast to the morphological studies, functional in vivo studies scored high on statistical methods. Similar to the morphological studies, study limitations and clinical implications were lacking in the majority of the functional in vivo manuscripts, affecting the scientific implications and translation items. Furthermore, blinding, protocol registration, and data access were almost completely lacking. Reporting sample size and applying randomization were insufficiently present in the studies. Although ethical statements were present in the majority of the studies, exact details on housing and husbandry conditions and animal care and monitoring were missing.
The range between quality scores of functional in vitro studies assessed by the adjusted ARRIVE guidelines was the greatest (29–68%) with a median of 41% (interquartile range 36–50%). Like the functional in vivo studies, the functional in vitro studies scored high on study design, outcome measures, experimental procedures, background, and objectives (Fig. 3C). Moreover, culture conditions were fairly consistently reported. The low overall scores were mainly owing to the absence of information on the animals that were used to acquire the experimental cells or tissues. The number of cells or tissues used was often lacking or not clearly reported. Some papers were thorough in describing the statistical methods, whereas others lacked information on the software in which the statistical analysis was performed or an explanation of reference to the calculations in case no software was used. Items that were nearly completely lacking in the functional in vivo studies were also insufficient in the functional in vitro studies, such as randomization, blinding, generalizability/translation, protocol registration, and data access. Remarkably, funding was reported in almost all functional in vitro manuscripts, but a declaration of interest was often absent, leading to a moderate score on this item. A detailed quality assessment is presented in the Supplementary Materials (Appendix C).

Morphological evidence of SCG-cardiac innervation per species

A total of 5 studies were performed in human and 21 in other species (Table 2). In many of the included studies, the main aim was to describe the cardiac autonomic nervous system in general, and they were therefore not focused on the superior cervical ganglion in particular (Supplementary Materials, Appendix D).
Table 2
Summary of the key findings in morphological papers
 
Morphological studies
Author
Year
Pre/postnatal
Type of study
N*
Health status
Sex (M/F)
Relevance sex
Both sides SCG included
Relevance sidedness
Direct involvement of SCG in cardiac innervation found
Indirect involvement of SCG in cardiac innervation found
Human
 Janes et al.
1986
Postnatal
Macromorphology
23
Healthy
15/8
NR
Yes
No
No
No
 Pather et al.
2003
Prenatal
Macromorphology
8
Healthy
4/4
NR
Yes
No
Yes, via superior cardiac nerve
Postnatal
21
11/10
 Kawashima and Sasaki
2005
Postnatal
Macromorphology
2
Retroesophageal right subclavian artery (cause of death: case 1, gastric cancer; case 2, duodenal cancer)
0/2
Yes
Yes, left > right
Sometimes, via superior cardiac nerve
Sometimes, via sympathetic trunk
 Kawashima
2005
Postnatal
Macromorphology
18
Clearly abnormal hearts or surrounding vessels excluded
NR/NR
Yes
Yes, left > right
Sometimes, via superior cardiac nerve
Sometimes, via sympathetic trunk
 Kawashima and Sasaki
2007
Postnatal
Macromorphology
1
Anomalous left vertebral artery; normal great arterial branching pattern of the aortic arch
NR/NR
Yes
No
Yes, via superior cardiac nerve
26
Yes
Yes, left > right
Yes, via superior cardiac nerve
Nonhuman primate (specification see text)
 Billman et al.
1982
Postnatal
Macromorphology
10
Healthy
10/0
Yes
No
No
No
 Chuang et al.
1992
Postnatal
Retrograde labeling
15
Healthy
Either sex, number NR
NR
Yes
Yes, depending on injected location
Yes, limited number of traced cells
 Chuang et al.
2004
Postnatal
Retrograde labeling
16
Healthy
Either sex, number NR
NR
Yes
Yes, depending on injected location
Yes, limited number of traced cells
 Kawashima et al.
2005
Postnatal
Macromorphology
11
Healthy
7/4
No
Yes
No
No
Yes, via sympathetic trunk
 Kawashima et al.
2008
Postnatal
Macromorphology
10
Healthy
8/2
No
Yes
Yes, left > right
Sometimes, via superior cardiac nerve
Sometimes, via sympathetic trunk
 Kawashima et al.
2009
Postnatal
Macromorphology
12
Clearly abnormal hearts, surrounding great vessels or a heart position associated with a condition excluded
4/7, 1 NR
No
Yes
Yes, left > right
No
Yes, via sympathetic trunk
 Kawashima and Thorington
2011
Postnatal
Macromorphology
7
Clearly abnormal heart, surrounding great vessels or a heart position associated with a condition excluded
3/4
NR
Yes
NR
No
Yes, via sympathetic trunk
 Kawashima et al.
2013
Postnatal
Macromorphology
3
Healthy
NR/NR
Yes
No
No
Yes, via sympathetic trunk
Dog
 Armour and Hopkins
1981
Postnatal
Retrograde labeling
38
Healthy
Either sex, number NR
NR
Yes
Yes, depending on injected location
Yes, limited number of traced cells
 Hopkins and Armour
1984
Postnatal
Retrograde labeling
27
Healthy
Either sex, number NR
NR
Yes
Yes, depending on injected location
Yes, limited number of traced cells
 Hirakawa et al.
1993
Postnatal
Retrograde labeling
23
Healthy
Either sex, number NR
NR
Yes
Yes, depending on injected location
Yes, limited number of traced cells
Cat
    
Healthy
      
 Shih et al.
1985
Postnatal
Retrograde labeling
15
Healthy
Either sex, number NR
NR
Yes
Yes, depending on injected location
Yes, limited number of traced cells
 Wu et al.
1988
Postnatal
Retrograde labeling
28
Healthy
Either sex, number NR
NR
Yes
Yes, depending on injected location
Yes, limited number of traced cells
Guinea pig
 Li et al.
2006
Postnatal
Anterograde labeling
NR
Chemical sympathectomy with 6-OHDA
NR/0
No
Yes, limited number of traced cells
Rat
           
 Pardini et al.
1989
Postnatal
Retrograde labeling
69
Healthy
NR/NR
Yes
No
Yes, limited number of traced cells
 Liu et al.
2013
Postnatal
Retrograde labeling
24
MI by LAD occlusion
NR/NR
No, only left SCG
Yes, limited number of traced cells
 Yu et al.
2018
Postnatal
Retrograde labeling
24
MI by LAD occlusion
24/0
Yes
No
Yes, limited number of traced cells
Mouse
 Manousiouthakis et al.
2014
Prenatal
Histomorphology
Unclear
Healthy
NR/NR
Yes
NR
No
Yes, intermixed with projections from stellate ganglia
Shrew
 Tanaka et al.
2007
Postnatal
Macromorphology
10
Healthy
5/5
NR
Yes
No
No
Yes
Chick
 Kirby et al.
1980
Prenatal
Histomorphology
NR chick
Healthy
NR/NR
Yes
No
No
No
 Verberne et al.
1999
Prenatal
Histomorphology
3 chick, 21 quail-chick chimeras
Healthy
NR/NR
Yes
NR
No
Yes, via carotid nerve
Only n-numbers related to SCG were included in the table. NR Not reported, when health state is not specifically reported, it was considered as healthy

Human (n = 5)

The cardiac nerves are commonly described as nerves connecting to the heart either “with direct connections or connections via the cardiac plexus” [2, 9, 16, 25]. The superior cardiac nerve (SCN) was observed in most studies [9, 2527]. When observed, the SCN arose directly from the SCG (53–100% of cases) or the sympathetic trunk between the SCG and middle cervical ganglia (MCG). The left and right SCG were investigated in all included studies, but there is no consensus about the relevance of sidedness. Some subjects showed no relevance of sidedness [26, 27], while in others a larger contribution of the left than the right SCG was seen [9, 25, 27]. In one study, no cardiopulmonary nerves were found to originate from either the SCG or the sympathetic trunk between the SCG and MCG [28]. The studies included subjects from both sexes [26] or only females [25], or did not report the sex [9, 27]. Only one study also investigated prenatal subjects (8 out of 29 subjects) but did not specifically report separate results on the contribution of the SCG to cardiac innervation in pre- and postnatal/adult subjects [26].

Nonhuman primate (n = 8)

The relation between the SCG and cardiac innervation has been studied in various species of nonhuman primates (arranged by primate evolutionary phylogeny): Lorisiformes (lorises and galagos) [29], tarsiers [30], New World Monkeys [31], Old World Monkeys (e.g., rhesus, macaque, and Taiwan monkey) [3234], gibbons [35], and unspecified [36].
In the Lorisiformes, tarsiers, New World Monkeys, and Old World Monkeys, the SCN was observed, but never originated directly from the SCG. Instead, the SCN was found to originate indirectly from the sympathetic trunk between the SCG and MCG [2931, 33]. Contrastingly, one study performed in Old World Monkeys did not find a visual direct nor indirect connection between the SCG and the heart [32]. However, retrograde labeling with horseradish peroxidase (HRP) supported nerve connection between the SCG and the heart, also in Old World Monkeys [34, 36]. In gibbons, a nonhuman primate that is evolutionarily closed to the human, the SCN was observed to originate from the SCG in 65% and from the sympathetic trunk below the SCG in 50% of cases [35]. Within primates, higher levels in the evolutionary hierarchy seemed to relate to increased prevalence of the superior cardiac nerve and increased observation of a direct connection from the superior cardiac nerve toward the heart (Fig. 4).
Most studies included subjects from both sexes [29, 31, 33, 35], although the exact number per sex was sometimes not reported [36]. One study included only males, and one study did not report on the sex [30, 32]. In case results were specified per sex, no evident differences existed [31, 33, 35]. Both sides of the SCG were included in all studies, but the relevance of sidedness remains dubious, as some reported no differences [30, 32, 33], while others found a dominance of the left SCG over the right SCG, or reported that relevance of sidedness depended on the injected location of HRP [31, 3436]. No prenatal studies in the nonhuman primates investigating the connections between SCG and the heart were found.

Dogs (n = 3)

All studies performed in postnatal dogs used retrograde labeling to trace the connectivity from the heart to the SCG and found an involvement of the SCG in cardiac innervation, although the contribution was minimal [3739]. Only very few neurons could be traced back in the SCG after HRP injection in the heart or cardiac nerves, and in more than half of the SCG, no retrogradely labeled neurons could be traced back at all. The dogs were from either sex, but the number and the results were not reported separately. Both sides of the SCG were included in all studies, but the results were different depending on the location of the HRP injection. Upon HRP injection into the cardiac nerves, only sporadically labeled neurons were found in the caudal pole of the ipsilateral SCG [39]. No labeled neurons were observed on the contralateral side. After injection in the heart, only a small number of labeled cells could be traced back in the bilateral SCG [37, 38].

Cats (n = 2)

In both morphological studies performed in postnatal cats, retrograde labeling was used to study the relation between the heart and SCG [40, 41]. Data in the cat show similar results as in dogs, with no or only very few cells that could be traced back into the SCG after injection of HRP into the heart, depending on the injected location. Likewise, both SCG sides were included and its relevance depended on the site of HRP injection. Subjects were from either sex, but the exact number and the results were again not specified for each sex.

Guinea pigs (n = 1)

Only one study focused on the morphology of SCG in cardiac innervation in postnatal guinea pigs using anterograde labeling [42]. The tracer biotinylated dextran amine (BDA) was injected into the SCG, and a distribution of anterogradely traced sympathetic axons and varicosities was observed in the heart. No conclusions can be drawn regarding sex and SCG sidedness, as only males were included in the study and the SCG side was not reported.

Rats (n = 3)

All papers investigated the morphological relevance of the SCG in cardiac innervation in postnatal rats using retrograde labeling [4345]. Again, a small morphological contribution of the SCG in cardiac innervation seems to be present. Only a small minority of labeled cells were found bilaterally in the SCG after injection of the heart (3% left ventricle, 1% right ventricle). Interestingly, HRP injected into the cardiac apex several days after MI revealed more HRP staining in the SCG from MI rats compared with control or sham [43, 45]. One study was performed in male rats [45], while the sex was not reported in the other studies [43, 44]. In the two studies in which both sides of the SCG were included, no evident differences were found between the sides [44, 45].

Mice (n = 1)

Through murine whole-mount visualization of sympathetic nerve morphology using tyrosine hydroxylase (TH) staining, it was observed that a portion of nerves arising from the SCG project toward the heart [46]. These nerves intermix on the ventral side of the heart with the ventral projections from the stellate ganglia. Surprisingly, the SCG nerves did not project to the dorsal side of the heart. Both sides of the SCG were included, but the results were not specified per side. The sex was also not reported. In contrast to the previously described species, only prenatal mice were studied.

Shrew (n = 1)

Macromorphological connections between SCG and heart were present in the postnatal shrew [47]. The nerve originating from both the left and right SCG descended to reach the aortic arch and formed nerve plexuses supplying nerves to the ventral wall of the ventricle. An equal number of male and female shrews were studied, but the results were not specified per sex.

Chick (n = 2)

Histomorphological techniques (catecholamine histofluorescence and silver preparations) were used to study the developing sympathetic innervation in chick embryos [48]. All nerve branches from the SCG were directed cranially, and no fluorescent postganglionic fibers could be traced from the SCG to the heart. In contrast, in another study, no direct connection between the SCG and the heart was found, but the SCG seemed to innervate the heart indirectly via the carotid nerve, which joins the nodose ganglion of the vagal nerve whose branches enter the arterial and venous pole of the heart [49]. Both sides of the SCG were included in both studies, but the results were not separately reported. Additionally, the sex was not reported.

Summary

  • In all species, morphological evidence that the SCG innervates the heart either directly or indirectly was found, but its contribution is likely limited as only very few neurons could be retrogradely traced.
  • SCG sidedness may be relevant, and if so, often the left SCG is macromorphologically considered to contribute more than the right SCG to cardiac innervation. Of note, findings depended on the site of injection in the heart in retrograde labeling studies.
  • There is not enough morphological information about sex differences.
  • Prenatal studies were found to be underrepresented in morphological studies.

Functional in vivo evidence of SCG-cardiac innervation per species

Studies in the functional in vivo category were predominantly performed in rats (Table 3). All studies only included the postnatal/adult stage. The functional relationship between SCG and the heart in vivo was often part of the primary aim (Supplementary Materials, Appendix E).
Table 3
Summary of the key findings in functional in vivo papers
Functional—in vivo studies
Author
Year
Pre/postnatal
N*
Strain/breed
Condition
Sex (M/F)
Relevance sex
Both sides SCG included
Relevance sidedness
Substances tested/outcome measures
Any direct involvement of SCG in cardiac innervation found
Any indirect involvement of SCG in cardiac innervation found
Dog
 Brandys et al.
1984
Postnatal
4
Mongrel
Hexamethonium administration
Either sex, number NR
NR
Yes
No
Heart rate, atrial contraction force, intramyocardial pressure, ECG
No
Rabbit
 Cheng et al.
2018
Postnatal
10
NR
Isoproterenol-induced MI
Either sex, number NR
NR
NR
Fluvastatin
Yes
 Cheng et al.
2023
Postnatal
18
NR
Isoproterenol-induced MI
18/0
NR
P2Y12, TH, ECG, echo
Yes
Rat
 Zhang et al.
2007
Postnatal
Unclear
Sprague–Dawley
Isoproterenol-induced MI
Unclear/0
NR
A-317491, P2X3, behavior
Yes
 Li et al.
2010
Postnatal
Unclear
Sprague–Dawley
Isoproterenol-induced MI
Either sex, number NR
NR
NR
A-317491, P2X2, P2X3, TH, blood pressure, heart rate, respiration
Yes
 Li et al.
2011
Postnatal
Unclear
Sprague–Dawley
Isoproterenol-induced MI
Either sex, number NR
NR
NR
Oximatrine, NE, P2X3, blood pressure, heart rate
Yes
 Liu et al.
2013
Postnatal
64
Sprague–Dawley
LAD occlusion-induced MI
NR
NR
OxATP, TNFα, IL-6, CK-MB, CK, LDH, cardiac troponin I, glutamine synthetase, ERK, TH, substance P, neuronal nuclei, ECG (Q wave), blood pressure, heart rate
Yes
 Kong et al.
2013
Postnatal
56
Sprague–Dawley
LAD occlusion-induced MI
56/0
NR
BBG, P2X7, (p-)ERK1/2
Yes
 Liu et al.
2014
Postnatal
30
Sprague–Dawley
Isoproterenol-induced MI
Either sex, number NR
NR
NR
Puerarin, P2X3, TH, blood pressure, heart rate
Yes
 Na et al.
2014
Postnatal
30
Sprague–Dawley
Monocrotaline-induced pulmonary hypertension with left SCG block
30/0
No, only left
Nitrite, superoxide dismutase, RV systolic pressure, RV/LV mass ratio, heart rate variability
Yes
 Zhang et al.
2015
Postnatal
25
Sprague–Dawley
LAD occlusion-induced MI
25/0
NR
Baicalin, P2X3, CK-MB, cardiac troponin T, epinephrine, ATP, cardiac hypertrophy, Q wave (ECG), blood pressure, heart rate,
Yes
 Tu et al.
2016
Postnatal
56
Sprague–Dawley
LAD occlusion-induced MI
Either sex, number NR
NR
NR
NONRATT021972 siRNA, baicalin, P2X7, GAP43, TH, ECG (Q wave), blood pressure, heart rate
Yes
 Wu et al.
2016
Postnatal
24
Sprague–Dawley
Diabetes mellitus
24/0
NR
Uc.48 + siRNA, P2X2, P2X3, P2X5, P2X7, (p-)ERK1/2
Yes
 Xu et al.
2016
Postnatal
24
Sprague–Dawley
Diabetes mellitus
24/0
NR
NONRATT021972 siRNA, TNFα, insulin receptor substrate 1, neuronal nuclei, heart rate variability
Yes
 Zou et al.
2017
Postnatal
40
Sprague–Dawley
LAD occlusion-induced MI
NR
NR
P2Y12 shRNA, P2Y12, GFAP, TNFα, (p-)P38 mitogen-activated protein kinase, myocardial fiber structure, blood pressure, heart rate
Yes
 Liu et al.
2018
Postnatal
40
Sprague–Dawley
Β-aminopropionitrile-induced aortic dissection with SCG removal
40/0
Yes, bilateral
–*
Matrix metalloproteinase-9, aortic wall thickening, heart rate, aortic dissection incidence
Yes
 Yu et al.*
2018
Postnatal
24
Sprague–Dawley
LAD occlusion-induced MI
24/0
NR
Oxytonergic receptor, TH
Yes
 Shi et al.
2019
Postnatal
Unclear
Sprague–Dawley
LAD occlusion-induced MI
Unclear/0
NR
TH, GAD65/67, GABAARβ2, P2X7, intracellular Ca2+, NE, renal sympathetic nerve activity
Yes
 Prado et al.
2020
Postnatal
Unclear
Wistar
MI with reperfusion with SCG removal (Langendorff-setting)
Unclear/0
Yes, bilateral
–*
Melatonin receptor, SERCA2A, K-ATP channels, connexin-43, TNFα, nitrotyrosine, TGFβ, vimentin, ECG (ventricular arrhythmias, QRS, QT, QTc, PR), epicardial action potential, early after depolarizations, heart rate
Yes
 Zhang et al.
2020
Postnatal
104
Sprague–Dawley
LAD occlusion-induced MI
104/0
NR
SCG10 (marker for axonal regeneration), TH
Yes
 Zou
2022
Postnatal
72
Sprague–Dawley
LAD occlusion-induced MI
72/0
NR
Uc.48 + lncRNA/shRNA, clopidogrel, heart rate, blood pressure
Yes
 Zhang
2023
Postnatal
40
Sprague–Dawley
LAD occlusion-induced MI
40/0
NR
P2X7, GS, ECG, sympathetic nerve discharge (SND), blood pressure
Yes
Mouse
 Ziegler et al.
2018
Postnatal
22
C57BL6/N
LAD occlusion-induced MI with SCG removal
22/0
Yes, bilateral
–*
Sirius Red, high-sensitivity cardiac troponin T, pro-NGF, TH, CD68, Cx3cr1, TNFα, cardiac hypertrophy, cardiomyocyte hypertrophy, cardiac function
Yes
 Ge et al.
2022
Postnatal
20
C57BL/6 J
LAD occlusion-induced MI
20/0
Yes
No
NGF, BDNF, GAP43, TrK, TH, CHAT, neuron size, beta-III-tubulin
Yes
HRV heart rate variability; ISH in situ hybridization, NR not reported, SCGx superior cervical ganglionectomy, A-317491 selective P2X2/3 receptor antagonist, oxATP: P2X7 receptor antagonist, BBG Brilliant Blue G: P2X7 receptor antagonist, puerarin: major active ingredient extracted from the traditional Chinese plant medicine Ge-gen, Baicalin: compound purified from the dry roots of Scutellaria baicalensis
*Owing to bilateral SCGx, the relevance of sidedness could not be studied

Dogs (n = 1)

The direct role of the SCG in cardiac innervation was investigated by measuring cardiac responses to stimulation of sympathetic ganglia in dogs from either sex (not further specified) [50]. When the SCG were stimulated bilaterally in a variety of regions, none of the stimulations produced any detectable cardiac responses. These data indicate that, although morphologically a small amount of nerves can be traced between heart and SCG, functionally this is insufficient to achieve significant cardiac stimulation.

Rabbits (n = 2)

After the induction of MI in rabbits, ion channel characteristics of SCG neurons were studied. It was found that various channel proteins were increased in the SCG neurons after MI, indicating that a functional link between the SCG and heart exists in vivo [51]. The rabbits were reported to be from either sex, but these results were not reported separately. Additionally, which SCG side was included in the experiments was not reported. In another study of male rabbits, MI led to alterations in the activation and inactivation characteristics of the sodium channels accompanied by increase expression of P2Y12, a purinergic receptor, in SCGs [52]. Of note, these changes in the SCG post-MI do not necessarily show a direct connection, but could be indirect owing to alterations in SCG function through reflex pathways.

Rats (n = 21)

A vast amount of functional in vivo evidence for the indirect contribution of the SCG to cardiac innervation can be attributed to the multitude of studies performed in rats. Different functional methods were used, such as the measurement of blood pressure and heart rate, combined with (immuno)histochemistry, immunofluorescence, Western Blot, (q)RT-PCR, enzyme-linked immunosorbent assay (ELISA), and in situ hybridization. All studies found a functional in vivo connection between the SCG and the heart. This link could be observed in various pathological conditions, including MI (induced by isoproterenol or LAD occlusion), diabetes mellitus, aortic dissection, and pulmonary hypertension (Table 3). Many changes in the SCG after MI were related to purinergic signaling [43, 5363]. The studies included male rats or rats from either sex, but in the latter case the exact number per sex was not reported. Studies only performed in female rats did not exist. In nearly all studies, the side of the studied SCG was not reported.

Mice (n = 2)

Both studies were performed in male adult C57BL6 mice with MI induced by LAD occlusion [64, 65]. Interestingly, removal of the SCG led to almost entire loss of myocardial sympathetic innervation in the left anterior wall (devoid of immunoreactivity) and the left and right SCG comparably contributed to the innervation of the left anterior wall [64]. Neuronal remodeling toward an increased adrenergic phenotype was observed in the SCG after MI, and these changes did not seem to differ between the left and right SCG [65]. When bilaterally removing the SCG during MI, positive effects were seen regarding cardiac function, inflammation, and hyperinnervation [64].

Summary

  • Functional in vivo evidence indicates that the SCG indirectly contributes to cardiac innervation and could be attributed to involvement in sympathetic overdrive reactions in response to cardiac diseases. Most data are derived from studies in disease models. A study performed in dogs failed to demonstrate a direct contribution.
  • Information on the relevance of SCG sidedness is largely unavailable.
  • The female sex is highly underrepresented in functional in vivo studies, and no information on the relevance of sex exists.
  • All studies were performed in the postnatal stage. No functional in vivo studies were found in the prenatal stage.

Functional in vitro evidence of SCG-cardiac innervation per species

Similar to the functional in vivo category, functional in vitro studies were predominantly performed in rats (Table 4). The assessment of a functional relationship between SCG and the heart in vitro was often part of the primary aim (Supplementary Materials, Appendix F).
Table 4
Summary of the key findings in functional in vitro papers
Functional—in vitro studies
Author
Year
Prenatal/postnatal/mixed
N*
Strain/breed
Condition
Sex (M/F)
Relevance sex
Both sides SCG included
Relevance sidedness
Evaluation methods
Substances of interest
Experimental setting
Any direct involvement of SCG in cardiac innervation found
Any indirect involvement of SCG in cardiac innervation found
Rabbit
 Cheng et al.
2018
Postnatal
70
NR
Isoproterenol-induced MI
Either sex, number NR
NR
NR
Electrophysiological recordings
Fluvastatin
Damage to the myocardium, isolated SCG neurons studied
Yes
 Cheng et al.
2023
Postnatal
NR
NR
Isoproterenol-induced MI
NR
NR
Electrophysiological recordings
P2Y12
Damage to the myocardium, isolated SCG neurons studied
Yes
Rat
 Furshpan et al.
1976
Postnatal
180
NR
Healthy
NR
NR
Electrophysiological recordings
Atropine, propranolol, hexamethonium
Co-culture
Yes*
 Landis
1976
Postnatal
NR
NR
Healthy
NR
NR
Electron microscopy
Co-culture
Yes*
 Chun and Patterson
1977
Postnatal
NR
NR
Healthy
NR
NR
Phase-contrast microscopy, isotopic assay
NGF
Co-culture
Yes
 King et al.
1978
Postnatal
NR
Wistar
Healthy
NR
NR
Optical recordings
Tyramine, norepinephrine
Co-culture
Yes
 Schwab et al.
1981
Postnatal
Unclear
NR
Healthy
NR
NR
Electron microscopy, fluorescence microscopy
Lectins, toxins, HCM
Culture of SCG neurons with HCM
Yes*
 De Ridder and De Potter
1983
Prenatal
NR
Sprague–Dawley
Healthy
NR
Yes
NR
Electron/phase-contrast/fluorescence microscopy
Co-culture
Yes*
 Kessler et al.
1984
Postnatal
8
Sprague–Dawley
Healthy
NR
NR
Phase-contrast microscopy, radioimmunoassay, HPLC
Substance P
Co-culture
No
 Lahtinen et al.
1986
Mixed
NR
Sprague–Dawley
Healthy
Either sex, number NR
NR
NR
Phase-contrast microscopy
NGF
Co-culture
Yes
 Furshpan et al. (1)
1986
Postnatal
NR
Albino
Healthy
NR
NR
Electrophysiological recordings, electron microscopy
Acetylcholine, adenosine, alprenolol, atenolol, atropine, hexamethonium, methylxanthines, norepinephrine, phentolamine, propanolol, reserpine, sotalol
Co-culture
Yes*
 Furshpan et al. (2)
1986
Postnatal
NR
Albino
Healthy
NR
NR
Electrophysiological recordings
Acetylcholine, adenosine, atenolol, atropine, methylxanthines, norepinephrine, phentolamine,
Co-culture
Yes*
 Potter et al.
1986
Postnatal
NR
Albino
Healthy
NR
NR
Electrophysiological recordings, electron microscopy
Acetylcholine, adenosine, alprenolol, atenolol, atropine, hexamethonium, methylxanthines, norepinephrine, phentolamine, propanolol, reserpine, sotalol, HCM, HCM factor
Co-culture
Yes*
 Matsumoto et al.
1987
Postnatal
NR
NR
Healthy
NR
NR
Electrophysiological recordings
Acetylcholine, adenosine, atenolol, atropine, hexamethonium, serotonin–creatinine sulfate, gramine, methylxanthines, methysergide, phentolamine, Peptide YY, reserpine, somatostatin, substance P, neurotensin, VIP
Co-culture
Yes
 Conforti et al.
1991
Postnatal
Unclear
Sprague–Dawley
Healthy
NR
NR
Electrophysiological recordings
Nifedipine
Co-culture
Yes
 Kannan et al.
1994
Postnatal
NR
Wistar
Healthy
Either sex, number NR
NR
NR
Phase-contrast microscopy
NGF
Co-culture
Yes
 Lockhart et al.
1997
Postnatal
221
Simonson White
Healthy
NR
NR
Electrophysiological recordings, phase-contrast/fluorescence microscopy, RT-PCR
NGF, norepinephrine, TrkA
Co-culture
Yes
 Ulupinar et al.
1998
Mixed
10
Sprague–Dawley
Healthy
NR
NR
Phase-contrast microscopy, DiI-labeling
-
Co-culture
Yes
 Hasan
2006
Postnatal
Unclear
Sprague–Dawley
LAD occlusion-induced MI
Female
NR
Phase-contrast microscopy
NGF
Co-Culture
Yes
 Li et al.
2010
Postnatal
18
Sprague–Dawley
Isoproterenol-induced MI
Either sex, number NR
NR
NR
Electrophysiological recordings
ATP, A-317491
Damage to the myocardium, isolated SCG neurons studied
Yes
 Miwa et al.
2010
Postnatal
Unclear
Wistar
Healthy
NR
NR
Electron/fluorescence microscopy
NGF, BDNF, GDNF, CNTF, Sema3A
Co-culture
Yes
 Takeuchi et al.
2011
Postnatal
Unclear
Wistar
Healthy
NR
NR
Electrophysiological recordings
Propanolol
Co-culture
Yes*
 Kong et al.
2013
Postnatal
Unclear
Sprague–Dawley
LAD occlusion-induced MI
Male
NR
Electrophysiological recordings
P2X7, BzATP, BBG, GF109203X
Damage to the myocardium, isolated SCG neurons studied
Yes
 Miwa et al.
2013
Postnatal
91
Wistar
Healthy
NR
NR
Electrophysiological recordings, electron/fluorescence microscopy
NGF, GDNF, synapsin-1
Co-culture
Yes
 Liu et al.
2014
Postnatal
NR
Sprague–Dawley
Isoproterenol-induced MI
Either sex, number NR
NR
NR
Electrophysiological recordings
Puerarin, A-317491
Damage to the myocardium, isolated SCG neurons studied
Yes
Mouse
 Coughlin et al.
1981
Prenatal
Unclear
CD-1
Healthy
NR
NR
Phase-contrast microscopy, biochemistry
HCM factor, TH, CHAT, NGF
Culture of whole SCG with HCM factor
Yes*
 Coughlin and Kessler
1982
Prenatal
NR
CD-1
Healthy
NR
NR
Chromatography, electrophoresis
NGF, HCM factor
Culture of whole SCG with HCM factor
Yes
 Rawdon and Dockray
1983
Postnatal
Unclear
Piebald-lethal
Healthy
NR
NR
Phase-contrast microscopy
Co-culture
Yes
 Uchida and Tomonaga
1985
Mixed
NR
C57BL/6
Healthy
Male
NR
NR
Phase-contrast microscopy
NGF, HCM
Culture of SCG neurons with HCM
Yes
 Rawdon
1991
Mixed
24
Swiss Webster
Healthy
NR
NR
Phase-contrast microscopy
NGF
Co-culture
Yes
 Shcherbakova et al.
2007
Postnatal
NR
NR
Healthy
NR
NR
Fluorescence microscopy, optical recordings
Cadherins, catenins, SAP97, AKAP79, b1Ars, caveolin-3, b2Ars, norepinephrine, epinephrine
Co-culture
Yes
 Ge et al.
2020
Mixed
81
C57BL/6 J
Healthy
Either sex, number NR
NR
NR
NR
Fluorescence microscopy, PCR, western blot
β3-Tubulin, NGF
Co-culture
Yes
Con A concanavalin A, WGA wheat germ agglutinin, SBA soybean agglutinin, HA hemagglutinin, HLPC high-performance liquid chromatography, 8-PT 8-phenyltheophylline, VIP vasoactive intestinal polypeptide, BBG Brilliant Blue G: P2X7 receptor antagonist, GF109203X: protein kinase C inhibitor, A-317491: selective P2X2/3 receptor antagonist, HCM heart-conditioned medium
*No control group was included to verify the results

Rabbit (n = 2)

After studying the functional characteristics of ion channels in SCG of postnatal MI rabbits in vivo, the electrical activity of SCG neurons was also studied in vitro after MI [51, 52]. The mean amplitude of action potentials of the neurons increased and action potential duration (APD90) was shorter after MI (Supplementary Materials, Appendix F). As mentioned above, the rabbits could be from either sex (not further specified) and SCG side was not reported.

Rat (n = 23)

Regarding experiments performed in rats, co-cultures of myocardium and neurons were most common, followed by measurements in isolated SCG neurons after damage to the myocardium or culture of SCG neurons using heart conditioned medium (Table 4). A great diversity in co-culture methods were reported, including:
1.
Single SCG neurons or multiple dissociated SCG neurons on top of cardiomyocytes
 
2.
Small SCG tissue clumps or whole SCG explants attached to or cultured in close proximity to cardiomyocytes/myocardium
 
3.
Mass culture of dissociated neurons and cardiomyocytes
 
Of importance, it should be noted that co-culture experiments are limited to investigating the influence of cell types on each other. This means that only an indirect link between the SCG and the heart can be investigated through co-culture experiments, but a direct link can not be unequivocally determined.
Off all included studies, only one study did not indicate a contribution of the SCG to cardiac innervation [66]. In that study, dissociated SCG neurons co-cultured with monolayers heart cells had no effect on neuropeptide content, while co-cultures with the pineal and salivary gland resulted in a striking increase.
In contrast, all other studies on a functional in vitro level supported a role for the SCG in cardiac innervation. SCG neurons co-cultured with cardiomyocytes resulted in the survival of SCG neurons, the development of functional synaptic contacts and in addition, cardiomyocytes reacted by evoked responses and increasing beat rate upon stimulation of the SCG neurons [6774]. Individual neurons within the SCG could have differential effects on the cardiomyocytes (inhibitory, excitatory, and dual function) by secreting different neurotransmitters [7577]. The functional in vivo role of purinergic signaling in the interplay between SCG and the heart was also confirmed in vitro, where many of the SCG neurons could evoke hyperpolarizations of cardiomyocytes, which were attenuated by adenosine-receptor blockers and adenosine deaminase, an enzyme that hydrolyzes adenosine to pharmacologically inactive inosine [78]. Similar evidence for a functional relation between SCG and the heart was found in experiments using small SCG tissue clumps and whole SCG explants. Co-culturing heart tissue with SCG caused stimulation and directional orientation of neurite outgrowth of SCG, while this was less evident in co-cultures of myocardium with other types of tissue [7982]. In three studies, SCG neurons were cultured after damage to the myocardium by induction of MI [55, 57, 58]. These studies revealed that MI led to pathological changes in the electrophysiological properties of SCG neurons, indicating a relation between SCG and the heart. Interestingly, the addition of heart conditioned medium induced remodeling of the SCG neurons in the form of fundamental changes in the phenotype (adrenergic/cholinergic/purinergic) as well as in the secreted neurotransmitter [77, 83]. Controls for heart tissue or cells often comprised tissue or cells from another origin, such as the gut. Of note, close to a third of all functional in vitro studies performed in rats did not use a control group for cardiomyocytes or neurons to verify the results [67, 7478, 81, 83].
Co-cultures of SCG and hearts that were both derived from prenatal rats were used in only one study [81]. Two studies used prenatal SCG, while culturing it with postnatal cardiac tissue [79, 82]. All other studies were performed with SCG and hearts from postnatal rats (19 out of 22; Table 4). The macromorphological or functional in vivo category the postnatal stage in the functional in vitro category mainly consisted of neonates instead of adults. Sporadically, it was stated that the neurons or myocytes derived from the male sex or either sex, while the exact numbers were not specified. Potential sex differences were not reported in the results [55, 57, 58, 79, 80], The sidedness of the SCG was not reported in the included studies in rat.

Mice (n = 7)

All SCG (co-)cultures performed with murine material supported the presence of a neuron–cardiomyocyte relationship. Similar to the experiments performed in rats, directional outgrowth of neurites from whole murine SCG was observed when co-cultured with heart tissue [84, 85]. Additionally, co-culturing cardiomyocytes on a layer of dissociated SCG neurons revealed the formation of functional synapses [86]. Stimulation of neuronal activity resulted in changes in sympathetic receptors in the cardiomyocytes, supporting the role of the SCG to cardiac innervation. Adding heart conditioned medium to dissociated SCG neurons induced neurite survival, including production and elongation [87, 88]. On the other hand, antiserum to a neuronal growth factor isolated from heart conditioned medium specifically blocked neurite extension [89]. These findings support the presence of a substance in heart conditioned medium influencing the SCG.
Two studies included postnatal mice [84, 86], two papers included prenatal mice [87, 89], and three used a combination of pre- and postnatal mice [85, 88, 90]. Of these studies, one was performed in tissue from adult mice ranging from 6 to 30 months; postnatal animals in other studies were from the neonatal stage. Direct comparisons between the pre- and postnatal stage in the SCG to cardiac innervation are scarce, and owing to the heterogeneity in the exact age and culture settings, it is currently impossible to draw conclusions regarding differences in pre- and postnatal stage.
The male sex was only stated in one study [88], while all other studies did not report the sex of the mice. Additionally, SCG sidedness was never reported.

Summary

  • Functional in vitro evidence indicates that the SCG indirectly contributes to cardiac innervation by its response to NGF produced by cardiomyocytes.
  • Information on the relevance of SCG sidedness is unavailable in in vitro studies.
  • No conclusion can be drawn with regard to sex differences as the sex is generally not reported.
  • Experiments were heterogeneous in age and culture settings and were predominantly performed in postnatal tissues from the neonatal stage.

Discussion

This systematic review evaluated current evidence for morphological and functional involvement of SCG in cardiac innervation in various species. The main findings are as follows:
1.
Both morphological as well as functional evidence supports an indirect contribution of the SCG to cardiac innervation. Evidence of a direct contribution has been found morphologically but, when present, only points out a minor contribution.
 
2.
Several gaps in current knowledge were found:
(a)
SCG sidedness may be relevant according to some morphological studies, but this remains uninvestigated in functional studies.
 
(b)
It is unclear whether sex differences exist, either owing to the male predominance in the studies or a lack of reporting of the sex of the included subjects.
 
(c)
It is unclear whether differences in the pre- and postnatal/adult stages exist, owing to the lack of prenatal subjects in morphological and functional in vivo studies and the heterogeneity of experiments in functional in vitro studies.
 
 

Contribution of the SCG to cardiac innervation

In all species, morphological evidence that the SCG indirectly contributes to cardiac innervation was found, but the extent of the contribution varies per species. In humans and in primates, higher levels in the evolutionary hierarchy seem to relate to an increased prevalence of the superior cardiac nerve and increased observation of a direct connection from the superior cardiac nerve toward the heart (Fig. 4) [29]. It is unclear whether an evolutionary hierarchy exists over the entire spectrum of species, as macroscopic observation is challenging in smaller species and differential methods, such as retrograde labeling, were performed. In each case, only a few cells could retrogradely be traced back.
Functional studies revealed evidence that the SCG indirectly contributes to cardiac innervation, both in vivo and in vitro. Functional in vivo studies researching the effects of superior cervical ganglionectomy on the heart, effects of myocardial infarction on the SCG, or the effects on the heart after injecting substances into the SCG did not provide evidence for direct innervation of the heart by SCG neurons. One study investigating the direct effect by SCG stimulation failed to find a contribution [50]. These data more likely reflect indirect effects of altered sympathetic nervous system activity. Triggers, such as MI or heart failure, could lead to changes in cardiac afferent signaling, resulting in alterations in sympathetic nervous system activity through a reflex pathway rather than through a direct link [91, 92].
The majority of functional in vitro studies consisted of co-cultures of cardiac and SCG tissue or cells, cultures of SCG cells in heart-conditioned medium, or induction of MI and investigating isolated SCG neurons. Of interest, other neighboring cells of the heart and SCG, such as epicardium-derived cells or carotid body cells, may influence this interaction between SCG and cardiac innervation [65, 90]. A limitation of co-culture experiments is that a direct link between SCG and the heart cannot be unequivocally determined. These experiments rather indicate whether two cell types cultured together can influence each other. Nevertheless, these indirect results are relevant and could generate new hypotheses for future studies. Most of the included functional studies have shown that SCG neurons respond to NGF produced by cardiomyocytes. Therefore, an indirect contribution to cardiac innervation may be applicable to other sympathetic ganglia than the SCG alone, as they also respond to NGF.
In summary, the morphological data indicate a minor contribution of the SCG, whereas functional data point to a more significant contribution. This discrepancy could partly be attributed to publication bias, as negative results are rarely published and particularly functional studies are designed to test a hypothesis. This means that the relevance of the SCG in cardiac innervation may be overrepresented in the functional studies.
On the basis of the findings described above, to study the direct role of specific autonomic ganglia in cardiac sympathetic innervation, it should be considered to select sympathetic ganglia located closer to the heart, such as the stellate ganglia or cardiac ganglia. However, the SCG might still be useful to study the general role of sympathetic neurons in cardiac diseases owing to its accessibility in all species.

Choosing the best SCG side for future studies

Approximately half of the studies performed in humans did not show any effect of sidedness, while in others the left side seemed to have a larger contribution to cardiac innervation. Morphological studies using retrograde labeling showed different results depending on the location of HRP injection. This is not surprising, because the peripheral cardiac autonomic nervous system shows considerable asymmetry, interindividual variations, and regional differences in anatomical, functional, and molecular characteristics [16]. With novel techniques such as single-cell RNA sequencing, the genetic characterization of tissues at single-cell resolution is possible [93]. However, these methods can be expensive, so best SCG side choice is critical. In this regard, functional studies could potentially contribute more as many papers included in the currently evaluated manuscripts failed to mention which side was used or whether sidedness was relevant.

Sex differences exist in health and disease, but information in the SCG is lacking

It has become clear in recent years that marked sex differences exist in cardiac autonomic innervation [15, 94, 95]. It is important to take into account potential sex differences in SCG, as it may influence experimental outcome and shed light on sex differences in (outcomes of) cardiovascular disease. In the studies included in this systematic review, either male subjects were included or the sex was not reported at all. This is in line with our previous systematic review on hyperinnervation after myocardial infarction (manuscript under review), in which only 8% of studies specifically included the female sex. This underrepresentation of female experimental models concerns all species. Similar concerns have been addressed in human clinical studies [96].

Prenatal stage remains underexposed

The vast majority of the studies included postnatal subjects and tissues in all three categories (morphology, functional in vivo, and functional in vitro). It is striking that the prenatal SCG has been studied to a lesser extent than postnatal/adult SCG, as prenatal SCG have the advantage of being more prone to sprout during co-culturing in vitro than adult ganglia, at least in control (“healthy”) settings. Many researchers seem to choose neonatal SCGs, which also show better sprouting capacity as compared with adult SCG, as an alternative. The controversial embryological origin of the cervical sympathetic chain ganglia may add to the preference for using postnatal/adult subjects and tissues in research. On the basis of the observation that cellular clusters will expand from the thoracic to the cervical region [97], it has been speculated that cervical ganglia are generated from the thoracic sympathetic chain [9, 98]. As there are only 3–4 cervical ganglia in the cervical region whereas at the thoracic level each spinal level has a corresponding ganglion, alternatively, it has been suggested that the development of sympathetic ganglia is associated initially with the intersegmental vessels [99]. The limited number of cervical ganglia could, in this perspective, be attributed to regression of most of the cervical intersegmental arteries with subsequent remodeling and fusion of the corresponding ganglia. The upper four cervical ganglia would thus eventually form the superior cervical sympathetic ganglion, anatomically related or induced by the developing external carotid artery [99]. Either way, changes occur during development and the contribution of SCG to the heart may vary in different stages, which may be another motivation for the choice to study postnatal/adult subjects and tissue. More information on fetal ganglia is required to comprehend the neuronal plasticity and possible re-expression of a fetal phenotype in disease states [100].

Specific structure and function of the SCG as compared with other sympathetic chain ganglia

The SCG is a remarkable mass of nerve cells that has a unique spatial anatomical localization (Fig. 5). It is situated between the branching point of the common carotid arteries, in close proximity to the carotid body, which it innervates [101]. The innervation pattern, however, is much broader than the cardiovascular system alone. Nerve fibers originating from the SCG provide sympathetic input toward the head, where it stimulates parts of the eye and blood vessels [102]. In this respect, it may be relevant that the SCG is situated adjacent to the above-mentioned carotid body, itself an intriguing structure involved in oxygen, carbon, and pH sensing, which has been shown to produce many neurotrophic factors [65, 103]. With regard to cardiovascular disease states, a role of the carotid body in hypertension has been indicated [104]. Although the SCG is a sympathetic chain ganglion, remarkably, a connection of the sympathetic SCG with the parasympathetic nodose ganglion has been observed in mice and rhesus monkeys [14, 32]. Therefore, a contribution or functional interaction of the SCG with cardiac parasympathetic innervation may be possible.

Clinical relevance

Neuromodulatory interventions to treat refractory ventricular arrhythmias are emerging and include blockade or surgical removal of stellate ganglia [105]. The SCG seems to have limited contributions to cardiac innervation as compared with, e.g., the stellate ganglion, and shows important interindividual anatomical heterogeneity [14]. As the direct contribution of the SCG to cardiac innervation is likely limited and the dominant effect of the SCG appears to occur cranially, the clinical relevance seems to be limited on the basis of current knowledge. The SCG is useful to study the role of sympathetic neurons in cardiac diseases in experimental settings owing to its accessibility in all species.

Limitations

The overall quality of the studies was moderate using our selected quality scores. The oldest included study was published in 1976, and the most recent in 2023. As many publication reporting guidelines and checklists have been developed in more recent years and journals generally have more detailed submission guidelines, the year of publication could influence the quality of the results. Additionally, using checklists, some papers may receive a higher quality than expected, as poor English and citation of faulty references might have been overlooked. By excluding non-English-language papers, some data may have been lost.

Conclusions

Current literature supports indirect involvement of the SCG in cardiac innervation, at both a morphological and a functional level. Evidence of direct involvement seems limited. Therefore, the SCG is an adequate structure to take into account when studying the role of sympathetic structures in cardiac function in both health and disease. Studies investigating the direct contribution of sympathetic innervation to the heart should rather be focusing on the stellate or cardiac ganglia. The relevance of SCG sidedness, sex, and developmental stage in health and disease also remains unclear and warrants further exploration.

Acknowledgements

We thank Ron Slagter for drawing Fig. 1.

Declarations

Conflict of interest

The authors declare no conflicts of interest.
Open Access This 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/​.

Unsere Produktempfehlungen

Neuer Inhalt

Print-Titel

e.Med Interdisziplinär

Kombi-Abonnement

Jetzt e.Med zum Sonderpreis bestellen!

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

Jetzt bestellen und 100 € sparen!

Anhänge

Supplementary Information

Below is the link to the electronic supplementary material.
Literatur
1.
Zurück zum Zitat Kapa S, Venkatachalam KL, Asirvatham SJ (2010) The autonomic nervous system in cardiac electrophysiology: an elegant interaction and emerging concepts. Cardiol Rev 18(6):275–284PubMedCrossRef Kapa S, Venkatachalam KL, Asirvatham SJ (2010) The autonomic nervous system in cardiac electrophysiology: an elegant interaction and emerging concepts. Cardiol Rev 18(6):275–284PubMedCrossRef
2.
Zurück zum Zitat Wink J et al (2020) Human adult cardiac autonomic innervation: Controversies in anatomical knowledge and relevance for cardiac neuromodulation. Auton Neurosci 227:102674PubMedCrossRef Wink J et al (2020) Human adult cardiac autonomic innervation: Controversies in anatomical knowledge and relevance for cardiac neuromodulation. Auton Neurosci 227:102674PubMedCrossRef
3.
5.
Zurück zum Zitat Ajijola OA et al (2015) Remodeling of stellate ganglion neurons after spatially targeted myocardial infarction: neuropeptide and morphologic changes. Heart Rhythm 12(5):1027–1035PubMedPubMedCentralCrossRef Ajijola OA et al (2015) Remodeling of stellate ganglion neurons after spatially targeted myocardial infarction: neuropeptide and morphologic changes. Heart Rhythm 12(5):1027–1035PubMedPubMedCentralCrossRef
6.
Zurück zum Zitat Chen PS et al (2001) Sympathetic nerve sprouting, electrical remodeling and the mechanisms of sudden cardiac death. Cardiovasc Res 50(2):409–416ADSPubMedCrossRef Chen PS et al (2001) Sympathetic nerve sprouting, electrical remodeling and the mechanisms of sudden cardiac death. Cardiovasc Res 50(2):409–416ADSPubMedCrossRef
7.
Zurück zum Zitat Li CY, Li YG (2015) Cardiac sympathetic nerve sprouting and susceptibility to ventricular arrhythmias after myocardial infarction. Cardiol Res Pract 2015:698368PubMedPubMedCentralCrossRef Li CY, Li YG (2015) Cardiac sympathetic nerve sprouting and susceptibility to ventricular arrhythmias after myocardial infarction. Cardiol Res Pract 2015:698368PubMedPubMedCentralCrossRef
9.
Zurück zum Zitat Kawashima T (2005) The autonomic nervous system of the human heart with special reference to its origin, course, and peripheral distribution. Anat Embryol (Berl) 209(6):425–438PubMedCrossRef Kawashima T (2005) The autonomic nervous system of the human heart with special reference to its origin, course, and peripheral distribution. Anat Embryol (Berl) 209(6):425–438PubMedCrossRef
10.
Zurück zum Zitat De Stefano ME et al (2005) Lack of dystrophin leads to the selective loss of superior cervical ganglion neurons projecting to muscular targets in genetically dystrophic mdx mice. Neurobiol Dis 20(3):929–942PubMedCrossRef De Stefano ME et al (2005) Lack of dystrophin leads to the selective loss of superior cervical ganglion neurons projecting to muscular targets in genetically dystrophic mdx mice. Neurobiol Dis 20(3):929–942PubMedCrossRef
11.
12.
Zurück zum Zitat Mul Fedele ML et al (2017) Alterations in metabolism and diurnal rhythms following bilateral surgical removal of the superior cervical Ganglia in rats. Front Endocrinol (Lausanne) 8:370PubMedCrossRef Mul Fedele ML et al (2017) Alterations in metabolism and diurnal rhythms following bilateral surgical removal of the superior cervical Ganglia in rats. Front Endocrinol (Lausanne) 8:370PubMedCrossRef
13.
Zurück zum Zitat Brognara F et al (2021) Autonomic innervation of the carotid body as a determinant of its sensitivity: implications for cardiovascular physiology and pathology. Cardiovasc Res 117(4):1015–1032PubMedCrossRef Brognara F et al (2021) Autonomic innervation of the carotid body as a determinant of its sensitivity: implications for cardiovascular physiology and pathology. Cardiovasc Res 117(4):1015–1032PubMedCrossRef
14.
Zurück zum Zitat Bookout AL, Gautron L (2021) Characterization of a cell bridge variant connecting the nodose and superior cervical ganglia in the mouse: prevalence, anatomical features, and practical implications. J Comp Neurol 529(1):111–128PubMedCrossRef Bookout AL, Gautron L (2021) Characterization of a cell bridge variant connecting the nodose and superior cervical ganglia in the mouse: prevalence, anatomical features, and practical implications. J Comp Neurol 529(1):111–128PubMedCrossRef
15.
Zurück zum Zitat Smetana P, Malik M (2013) Sex differences in cardiac autonomic regulation and in repolarisation electrocardiography. Pflugers Arch 465(5):699–717PubMedCrossRef Smetana P, Malik M (2013) Sex differences in cardiac autonomic regulation and in repolarisation electrocardiography. Pflugers Arch 465(5):699–717PubMedCrossRef
16.
17.
Zurück zum Zitat Nakamura K et al (2016) Pathological effects of chronic myocardial infarction on peripheral neurons mediating cardiac neurotransmission. Auton Neurosci 197:34–40PubMedPubMedCentralCrossRef Nakamura K et al (2016) Pathological effects of chronic myocardial infarction on peripheral neurons mediating cardiac neurotransmission. Auton Neurosci 197:34–40PubMedPubMedCentralCrossRef
18.
Zurück zum Zitat Moher D et al (2009) Preferred reporting items for systematic reviews and meta-analyses: the PRISMA statement. J Clin Epidemiol 62(10):1006–1012PubMedCrossRef Moher D et al (2009) Preferred reporting items for systematic reviews and meta-analyses: the PRISMA statement. J Clin Epidemiol 62(10):1006–1012PubMedCrossRef
19.
Zurück zum Zitat Koppel CJ et al (2020) Coronary anomalies in tetralogy of Fallot—a meta-analysis. Int J Cardiol 306:78–85PubMedCrossRef Koppel CJ et al (2020) Coronary anomalies in tetralogy of Fallot—a meta-analysis. Int J Cardiol 306:78–85PubMedCrossRef
20.
Zurück zum Zitat Engele LJ et al (2021) The coronary arteries in adults after the arterial switch operation: a systematic review. J Cardiovasc Dev Dis 8(9):102PubMedPubMedCentral Engele LJ et al (2021) The coronary arteries in adults after the arterial switch operation: a systematic review. J Cardiovasc Dev Dis 8(9):102PubMedPubMedCentral
21.
22.
23.
Zurück zum Zitat Du Sert NP et al (2020) The ARRIVE guidelines 2.0: updated guidelines for reporting animal research. J Physiol 598(18):3793–3801CrossRef Du Sert NP et al (2020) The ARRIVE guidelines 2.0: updated guidelines for reporting animal research. J Physiol 598(18):3793–3801CrossRef
25.
Zurück zum Zitat Kawashima T, Sasaki H (2005) Topological changes of the human autonomic cardiac nervous system in individuals with a retroesophageal right subclavian artery: two case reports and a brief review. Anat Embryol (Berl) 210(4):327–334PubMedCrossRef Kawashima T, Sasaki H (2005) Topological changes of the human autonomic cardiac nervous system in individuals with a retroesophageal right subclavian artery: two case reports and a brief review. Anat Embryol (Berl) 210(4):327–334PubMedCrossRef
26.
Zurück zum Zitat Pather N et al (2003) The sympathetic contributions to the cardiac plexus. Surg Radiol Anat 25(3–4):210–215PubMedCrossRef Pather N et al (2003) The sympathetic contributions to the cardiac plexus. Surg Radiol Anat 25(3–4):210–215PubMedCrossRef
27.
Zurück zum Zitat Kawashima T, Sasaki H (2007) Morphological comparison of the cardiac autonomic nervous system between normal and abnormal great arterial branching pattern with a brief review of the literature. Auton Neurosci 132(1–2):37–43PubMedCrossRef Kawashima T, Sasaki H (2007) Morphological comparison of the cardiac autonomic nervous system between normal and abnormal great arterial branching pattern with a brief review of the literature. Auton Neurosci 132(1–2):37–43PubMedCrossRef
28.
Zurück zum Zitat Janes RD et al (1986) Anatomy of human extrinsic cardiac nerves and ganglia. Am J Cardiol 57(4):299–309PubMedCrossRef Janes RD et al (1986) Anatomy of human extrinsic cardiac nerves and ganglia. Am J Cardiol 57(4):299–309PubMedCrossRef
29.
Zurück zum Zitat Kawashima T, Thorington RW Jr (2011) Comparative morphological configuration of the cardiac nervous system in lorises and galagos (infraorder Lorisiformes, Strepsirrhini, primates) with evolutionary perspective. Anat Rec (Hoboken) 294(3):412–426PubMedCrossRef Kawashima T, Thorington RW Jr (2011) Comparative morphological configuration of the cardiac nervous system in lorises and galagos (infraorder Lorisiformes, Strepsirrhini, primates) with evolutionary perspective. Anat Rec (Hoboken) 294(3):412–426PubMedCrossRef
30.
Zurück zum Zitat Kawashima T et al (2013) Evolutionary anatomy and phyletic implication of the extrinsic cardiac nervous system in the Philippine tarsier (Tarsius syrichta, Primates) in comparisons with Strepsirrhines and New World monkeys. Anat Rec (Hoboken) 296(5):798–806PubMedCrossRef Kawashima T et al (2013) Evolutionary anatomy and phyletic implication of the extrinsic cardiac nervous system in the Philippine tarsier (Tarsius syrichta, Primates) in comparisons with Strepsirrhines and New World monkeys. Anat Rec (Hoboken) 296(5):798–806PubMedCrossRef
31.
Zurück zum Zitat Kawashima T, Thorington RW Jr, Whatton JF (2009) Comparative anatomy and evolution of the cardiac innervation in New World monkeys (Platyrrhini, e. Geoffroy, 1812). Anat Rec (Hoboken) 292(5):670–691PubMedCrossRef Kawashima T, Thorington RW Jr, Whatton JF (2009) Comparative anatomy and evolution of the cardiac innervation in New World monkeys (Platyrrhini, e. Geoffroy, 1812). Anat Rec (Hoboken) 292(5):670–691PubMedCrossRef
32.
Zurück zum Zitat Billman GE, Dickey DT, Stone HL (1982) A description of the upper thoracic autonomic nervous system in the rhesus monkey (Macaca mulatta). Am J Primatol 2(2):159–166PubMedCrossRef Billman GE, Dickey DT, Stone HL (1982) A description of the upper thoracic autonomic nervous system in the rhesus monkey (Macaca mulatta). Am J Primatol 2(2):159–166PubMedCrossRef
33.
Zurück zum Zitat Kawashima T et al (2005) Comparative anatomical study of the autonomic cardiac nervous system in macaque monkeys. J Morphol 266(1):112–124PubMedCrossRef Kawashima T et al (2005) Comparative anatomical study of the autonomic cardiac nervous system in macaque monkeys. J Morphol 266(1):112–124PubMedCrossRef
34.
Zurück zum Zitat Chuang KS et al (2004) Horseradish peroxidase localization of sympathetic postganglionic and parasympathetic preganglionic neurons innervating the monkey heart. Chin J Physiol 47(2):95–99PubMed Chuang KS et al (2004) Horseradish peroxidase localization of sympathetic postganglionic and parasympathetic preganglionic neurons innervating the monkey heart. Chin J Physiol 47(2):95–99PubMed
35.
Zurück zum Zitat Kawashima T et al (2008) Systematic morphology and evolutionary anatomy of the autonomic cardiac nervous system in the lesser apes, gibbons (hylobatidae). Anat Rec (Hoboken) 291(8):939–959PubMedCrossRef Kawashima T et al (2008) Systematic morphology and evolutionary anatomy of the autonomic cardiac nervous system in the lesser apes, gibbons (hylobatidae). Anat Rec (Hoboken) 291(8):939–959PubMedCrossRef
36.
Zurück zum Zitat Chuang KS et al (1992) Localization of the sympathetic postganglionic neurons innervating cardiac coronary artery with horseradish peroxidase in monkeys. Chin J Physiol 35(3):219–226PubMed Chuang KS et al (1992) Localization of the sympathetic postganglionic neurons innervating cardiac coronary artery with horseradish peroxidase in monkeys. Chin J Physiol 35(3):219–226PubMed
37.
Zurück zum Zitat Hopkins DA, Armour JA (1984) Localization of sympathetic postganglionic and parasympathetic preganglionic neurons which innervate different regions of the dog heart. J Comp Neurol 229(2):186–198PubMedCrossRef Hopkins DA, Armour JA (1984) Localization of sympathetic postganglionic and parasympathetic preganglionic neurons which innervate different regions of the dog heart. J Comp Neurol 229(2):186–198PubMedCrossRef
38.
Zurück zum Zitat Hirakawa N, Morimoto M, Totoki T (1993) Sympathetic innervation of the young canine heart using antero- and retrograde axonal tracer methods. Brain Res Bull 31(6):673–680PubMedCrossRef Hirakawa N, Morimoto M, Totoki T (1993) Sympathetic innervation of the young canine heart using antero- and retrograde axonal tracer methods. Brain Res Bull 31(6):673–680PubMedCrossRef
39.
Zurück zum Zitat Armour JA, Hopkins DA (1981) Localization of sympathetic postganglionic neurons of physiologically identified cardiac nerves in the dog. J Comp Neurol 202(2):169–184PubMedCrossRef Armour JA, Hopkins DA (1981) Localization of sympathetic postganglionic neurons of physiologically identified cardiac nerves in the dog. J Comp Neurol 202(2):169–184PubMedCrossRef
40.
Zurück zum Zitat Shih CJ et al (1985) Horseradish peroxidase localization of the sympathetic postganglionic neurons innervating the cat heart. J Auton Nerv Syst 13(3):179–189PubMedCrossRef Shih CJ et al (1985) Horseradish peroxidase localization of the sympathetic postganglionic neurons innervating the cat heart. J Auton Nerv Syst 13(3):179–189PubMedCrossRef
41.
Zurück zum Zitat Wu JJ et al (1988) Sympathetic postganglionic innervation of the cardiac coronary artery in cats. J Auton Nerv Syst 24(3):215–220PubMedCrossRef Wu JJ et al (1988) Sympathetic postganglionic innervation of the cardiac coronary artery in cats. J Auton Nerv Syst 24(3):215–220PubMedCrossRef
42.
Zurück zum Zitat Li M et al (2006) Evidence for histamine as a neurotransmitter in the cardiac sympathetic nervous system. Am J Physiol Heart Circ Physiol 291(1):H45-51ADSPubMedCrossRef Li M et al (2006) Evidence for histamine as a neurotransmitter in the cardiac sympathetic nervous system. Am J Physiol Heart Circ Physiol 291(1):H45-51ADSPubMedCrossRef
43.
Zurück zum Zitat Liu J et al (2013) Sensory-sympathetic coupling in superior cervical ganglia after myocardial ischemic injury facilitates sympathoexcitatory action via P2X7 receptor. Purinergic Signal 9(3):463–479PubMedPubMedCentralCrossRef Liu J et al (2013) Sensory-sympathetic coupling in superior cervical ganglia after myocardial ischemic injury facilitates sympathoexcitatory action via P2X7 receptor. Purinergic Signal 9(3):463–479PubMedPubMedCentralCrossRef
44.
Zurück zum Zitat Pardini BJ, Lund DD, Schmid PG (1989) Organization of the sympathetic postganglionic innervation of the rat heart. J Auton Nerv Syst 28(3):193–201PubMedCrossRef Pardini BJ, Lund DD, Schmid PG (1989) Organization of the sympathetic postganglionic innervation of the rat heart. J Auton Nerv Syst 28(3):193–201PubMedCrossRef
45.
Zurück zum Zitat Yu R et al (2018) Expression of oxytocin receptor in the rat superior cervical ganglion after myocardial infarction. Int J Clin Exp Pathol 11(2):739–747PubMedPubMedCentral Yu R et al (2018) Expression of oxytocin receptor in the rat superior cervical ganglion after myocardial infarction. Int J Clin Exp Pathol 11(2):739–747PubMedPubMedCentral
46.
Zurück zum Zitat Manousiouthakis E et al (2014) Venous endothelin guides sympathetic innervation of the developing mouse heart. Nat Commun 5:3918ADSPubMedCrossRef Manousiouthakis E et al (2014) Venous endothelin guides sympathetic innervation of the developing mouse heart. Nat Commun 5:3918ADSPubMedCrossRef
47.
Zurück zum Zitat Tanaka A et al (2007) Gross anatomical study of the sympathetic cardiac nerves in the house musk shrew (Suncus murinus). Anat Rec (Hoboken) 290(5):468–476PubMedCrossRef Tanaka A et al (2007) Gross anatomical study of the sympathetic cardiac nerves in the house musk shrew (Suncus murinus). Anat Rec (Hoboken) 290(5):468–476PubMedCrossRef
48.
Zurück zum Zitat Kirby ML, McKenzie JW, Weidman TA (1980) Developing innervation of the chick heart: a histofluorescence and light microscopic study of sympthetic innervation. Anat Rec 196(3):333–340PubMedCrossRef Kirby ML, McKenzie JW, Weidman TA (1980) Developing innervation of the chick heart: a histofluorescence and light microscopic study of sympthetic innervation. Anat Rec 196(3):333–340PubMedCrossRef
49.
Zurück zum Zitat Verberne ME et al (1999) Contribution of the cervical sympathetic ganglia to the innervation of the pharyngeal arch arteries and the heart in the chick embryo. Anat Rec 255(4):407–419PubMedCrossRef Verberne ME et al (1999) Contribution of the cervical sympathetic ganglia to the innervation of the pharyngeal arch arteries and the heart in the chick embryo. Anat Rec 255(4):407–419PubMedCrossRef
50.
Zurück zum Zitat Brandys JC, Hopkins DA, Armour JA (1984) Cardiac responses to stimulation of discrete loci within canine sympathetic ganglia following hexamethonium. J Auton Nerv Syst 11(3):243–255PubMedCrossRef Brandys JC, Hopkins DA, Armour JA (1984) Cardiac responses to stimulation of discrete loci within canine sympathetic ganglia following hexamethonium. J Auton Nerv Syst 11(3):243–255PubMedCrossRef
51.
Zurück zum Zitat Cheng L et al (2018) Modulation of Ion channels in the superior cervical ganglion neurons by myocardial ischemia and fluvastatin treatment. Front Physiol 9:1157PubMedPubMedCentralCrossRef Cheng L et al (2018) Modulation of Ion channels in the superior cervical ganglion neurons by myocardial ischemia and fluvastatin treatment. Front Physiol 9:1157PubMedPubMedCentralCrossRef
52.
Zurück zum Zitat Cheng L et al (2023) Ticagrelor can regulate the ion channel characteristics of superior cervical ganglion neurons after myocardial infarction. J Cardiovasc Dev Dis 10(2):71PubMedPubMedCentral Cheng L et al (2023) Ticagrelor can regulate the ion channel characteristics of superior cervical ganglion neurons after myocardial infarction. J Cardiovasc Dev Dis 10(2):71PubMedPubMedCentral
53.
Zurück zum Zitat Zhang Z et al (2023) Schisandrin B alleviates diabetic cardiac autonomic neuropathy induced by P2X7 receptor in superior cervical Ganglion via NLRP3. Dis Markers 2023:9956950PubMedPubMedCentralCrossRef Zhang Z et al (2023) Schisandrin B alleviates diabetic cardiac autonomic neuropathy induced by P2X7 receptor in superior cervical Ganglion via NLRP3. Dis Markers 2023:9956950PubMedPubMedCentralCrossRef
54.
Zurück zum Zitat Zhang CP et al (2007) The involvement of P2X3 receptors of rat sympathetic ganglia in cardiac nociceptive transmission. J Physiol Biochem 63(3):249–257PubMedCrossRef Zhang CP et al (2007) The involvement of P2X3 receptors of rat sympathetic ganglia in cardiac nociceptive transmission. J Physiol Biochem 63(3):249–257PubMedCrossRef
55.
Zurück zum Zitat Li G et al (2010) Increased sympathoexcitatory reflex induced by myocardial ischemic nociceptive signaling via P2X2/3 receptor in rat superior cervical ganglia. Neurochem Int 56(8):984–990PubMedCrossRef Li G et al (2010) Increased sympathoexcitatory reflex induced by myocardial ischemic nociceptive signaling via P2X2/3 receptor in rat superior cervical ganglia. Neurochem Int 56(8):984–990PubMedCrossRef
56.
Zurück zum Zitat Li G et al (2011) Effects of oxymatrine on sympathoexcitatory reflex induced by myocardial ischemic signaling mediated by P2X(3) receptors in rat SCG and DRG. Brain Res Bull 84(6):419–424ADSPubMedCrossRef Li G et al (2011) Effects of oxymatrine on sympathoexcitatory reflex induced by myocardial ischemic signaling mediated by P2X(3) receptors in rat SCG and DRG. Brain Res Bull 84(6):419–424ADSPubMedCrossRef
57.
Zurück zum Zitat Kong F et al (2013) Electrophysiological studies of upregulated P2X7 receptors in rat superior cervical ganglia after myocardial ischemic injury. Neurochem Int 63(3):230–237ADSPubMedCrossRef Kong F et al (2013) Electrophysiological studies of upregulated P2X7 receptors in rat superior cervical ganglia after myocardial ischemic injury. Neurochem Int 63(3):230–237ADSPubMedCrossRef
58.
Zurück zum Zitat Liu S et al (2014) Puerarin alleviates aggravated sympathoexcitatory response induced by myocardial ischemia via regulating P2X3 receptor in rat superior cervical ganglia. Neurochem Int 70:39–49ADSPubMedCrossRef Liu S et al (2014) Puerarin alleviates aggravated sympathoexcitatory response induced by myocardial ischemia via regulating P2X3 receptor in rat superior cervical ganglia. Neurochem Int 70:39–49ADSPubMedCrossRef
59.
Zurück zum Zitat Zhang J et al (2015) Study of baicalin on sympathoexcitation induced by myocardial ischemia via P2X3 receptor in superior cervical ganglia. Auton Neurosci 189:8–15PubMedCrossRef Zhang J et al (2015) Study of baicalin on sympathoexcitation induced by myocardial ischemia via P2X3 receptor in superior cervical ganglia. Auton Neurosci 189:8–15PubMedCrossRef
60.
Zurück zum Zitat Tu G et al (2016) Long noncoding NONRATT021972 siRNA normalized abnormal sympathetic activity mediated by the upregulation of P2X7 receptor in superior cervical ganglia after myocardial ischemia. Purinergic Signal 12(3):521–535MathSciNetPubMedPubMedCentralCrossRef Tu G et al (2016) Long noncoding NONRATT021972 siRNA normalized abnormal sympathetic activity mediated by the upregulation of P2X7 receptor in superior cervical ganglia after myocardial ischemia. Purinergic Signal 12(3):521–535MathSciNetPubMedPubMedCentralCrossRef
61.
Zurück zum Zitat Wu B et al (2016) LncRNA uc48+ siRNA improved diabetic sympathetic neuropathy in type 2 diabetic rats mediated by P2X7 receptor in SCG. Auton Neurosci 197:14–18PubMedCrossRef Wu B et al (2016) LncRNA uc48+ siRNA improved diabetic sympathetic neuropathy in type 2 diabetic rats mediated by P2X7 receptor in SCG. Auton Neurosci 197:14–18PubMedCrossRef
62.
Zurück zum Zitat Zou L et al (2018) Downregulation of P2Y12 in the superior cervical ganglia alleviates abnormal sympathetic activity after myocardial ischemia. J Cell Physiol 233(4):3375–3383PubMedCrossRef Zou L et al (2018) Downregulation of P2Y12 in the superior cervical ganglia alleviates abnormal sympathetic activity after myocardial ischemia. J Cell Physiol 233(4):3375–3383PubMedCrossRef
63.
Zurück zum Zitat Zou L et al (2022) Implication of P2Y(12) receptor in uc.48+-mediated abnormal sympathoexcitatory reflex via superior cervical ganglia in myocardial ischemic rats. Eur J Pharmacol 927:175049PubMedCrossRef Zou L et al (2022) Implication of P2Y(12) receptor in uc.48+-mediated abnormal sympathoexcitatory reflex via superior cervical ganglia in myocardial ischemic rats. Eur J Pharmacol 927:175049PubMedCrossRef
64.
Zurück zum Zitat Ziegler KA et al (2018) Local sympathetic denervation attenuates myocardial inflammation and improves cardiac function after myocardial infarction in mice. Cardiovasc Res 114(2):291–299PubMedCrossRef Ziegler KA et al (2018) Local sympathetic denervation attenuates myocardial inflammation and improves cardiac function after myocardial infarction in mice. Cardiovasc Res 114(2):291–299PubMedCrossRef
65.
Zurück zum Zitat Ge Y et al (2022) Acute myocardial infarction induces remodeling of the murine superior cervical ganglia and the carotid body. Front Cardiovasc Med 9:758265PubMedPubMedCentralCrossRef Ge Y et al (2022) Acute myocardial infarction induces remodeling of the murine superior cervical ganglia and the carotid body. Front Cardiovasc Med 9:758265PubMedPubMedCentralCrossRef
66.
Zurück zum Zitat Kessler JA et al (1984) Target organ regulation of substance P in sympathetic neurons in culture. Dev Biol 103(1):71–79PubMedCrossRef Kessler JA et al (1984) Target organ regulation of substance P in sympathetic neurons in culture. Dev Biol 103(1):71–79PubMedCrossRef
67.
Zurück zum Zitat Landis SC (1976) Rat sympathetic neurons and cardiac myocytes developing in microcultures: correlation of the fine structure of endings with neurotransmitter function in single neurons. Proc Natl Acad Sci USA 73(11):4220–4224ADSPubMedPubMedCentralCrossRef Landis SC (1976) Rat sympathetic neurons and cardiac myocytes developing in microcultures: correlation of the fine structure of endings with neurotransmitter function in single neurons. Proc Natl Acad Sci USA 73(11):4220–4224ADSPubMedPubMedCentralCrossRef
68.
Zurück zum Zitat Chun LL, Patterson PH (1977) Role of nerve growth factor in the development of rat sympathetic neurons in vitro. III. Effect on acetylcholine production. J Cell Biol 75(3):712–718PubMedCrossRef Chun LL, Patterson PH (1977) Role of nerve growth factor in the development of rat sympathetic neurons in vitro. III. Effect on acetylcholine production. J Cell Biol 75(3):712–718PubMedCrossRef
69.
Zurück zum Zitat Conforti L, Tohse N, Sperelakis N (1991) Influence of sympathetic innervation on the membrane electrical properties of neonatal rat cardiomyocytes in culture. J Dev Physiol 15(4):237–246PubMed Conforti L, Tohse N, Sperelakis N (1991) Influence of sympathetic innervation on the membrane electrical properties of neonatal rat cardiomyocytes in culture. J Dev Physiol 15(4):237–246PubMed
70.
Zurück zum Zitat Lockhart ST, Turrigiano GG, Birren SJ (1997) Nerve growth factor modulates synaptic transmission between sympathetic neurons and cardiac myocytes. J Neurosci 17(24):9573–9582PubMedPubMedCentralCrossRef Lockhart ST, Turrigiano GG, Birren SJ (1997) Nerve growth factor modulates synaptic transmission between sympathetic neurons and cardiac myocytes. J Neurosci 17(24):9573–9582PubMedPubMedCentralCrossRef
71.
Zurück zum Zitat Miwa K et al (2010) Glial cell line-derived neurotrophic factor (GDNF) enhances sympathetic neurite growth in rat hearts at early developmental stages. Biomed Res 31(6):353–361PubMedCrossRef Miwa K et al (2010) Glial cell line-derived neurotrophic factor (GDNF) enhances sympathetic neurite growth in rat hearts at early developmental stages. Biomed Res 31(6):353–361PubMedCrossRef
72.
73.
Zurück zum Zitat King KL et al (1978) Chronotropic effect of tyramine on rat heart cells cultured with sympathetic neurons. Eur J Pharmacol 51(4):331–335PubMedCrossRef King KL et al (1978) Chronotropic effect of tyramine on rat heart cells cultured with sympathetic neurons. Eur J Pharmacol 51(4):331–335PubMedCrossRef
74.
Zurück zum Zitat Takeuchi A et al (2011) Device for co-culture of sympathetic neurons and cardiomyocytes using microfabrication. Lab Chip 11(13):2268–2275PubMedCrossRef Takeuchi A et al (2011) Device for co-culture of sympathetic neurons and cardiomyocytes using microfabrication. Lab Chip 11(13):2268–2275PubMedCrossRef
75.
Zurück zum Zitat Furshpan EJ et al (1976) Chemical transmission between rat sympathetic neurons and cardiac myocytes developing in microcultures: evidence for cholinergic, adrenergic, and dual-function neurons. Proc Natl Acad Sci USA 73(11):4225–4229ADSPubMedPubMedCentralCrossRef Furshpan EJ et al (1976) Chemical transmission between rat sympathetic neurons and cardiac myocytes developing in microcultures: evidence for cholinergic, adrenergic, and dual-function neurons. Proc Natl Acad Sci USA 73(11):4225–4229ADSPubMedPubMedCentralCrossRef
76.
Zurück zum Zitat Furshpan EJ et al (1986) Synaptic functions in rat sympathetic neurons in microcultures. I. Secretion of norepinephrine and acetylcholine. J Neurosci 6(4):1061–1079PubMedPubMedCentralCrossRef Furshpan EJ et al (1986) Synaptic functions in rat sympathetic neurons in microcultures. I. Secretion of norepinephrine and acetylcholine. J Neurosci 6(4):1061–1079PubMedPubMedCentralCrossRef
77.
Zurück zum Zitat Potter DD et al (1986) Synaptic functions in rat sympathetic neurons in microcultures. II. Adrenergic/cholinergic dual status and plasticity. J Neurosci 6(4):1080–1098PubMedPubMedCentralCrossRef Potter DD et al (1986) Synaptic functions in rat sympathetic neurons in microcultures. II. Adrenergic/cholinergic dual status and plasticity. J Neurosci 6(4):1080–1098PubMedPubMedCentralCrossRef
78.
Zurück zum Zitat Furshpan EJ, Potter DD, Matsumoto SG (1986) Synaptic functions in rat sympathetic neurons in microcultures. III. A Purinergic effect on cardiac myocytes. J Neurosci 6(4):1099–1107PubMedPubMedCentralCrossRef Furshpan EJ, Potter DD, Matsumoto SG (1986) Synaptic functions in rat sympathetic neurons in microcultures. III. A Purinergic effect on cardiac myocytes. J Neurosci 6(4):1099–1107PubMedPubMedCentralCrossRef
79.
Zurück zum Zitat Lahtinen T, Soinila S, Eranko O (1986) Age-dependent stimulation by atrium explants or nerve growth factor of nerve fibre outgrowth from cocultured embryonic rat sympathetic ganglia. Brain Res 392(1–2):51–57PubMedCrossRef Lahtinen T, Soinila S, Eranko O (1986) Age-dependent stimulation by atrium explants or nerve growth factor of nerve fibre outgrowth from cocultured embryonic rat sympathetic ganglia. Brain Res 392(1–2):51–57PubMedCrossRef
80.
Zurück zum Zitat Kannan Y et al (1994) Lymphoid tissues induce NGF-dependent and NGF-independent neurite outgrowth from rat superior cervical ganglia explants in culture. J Neurosci Res 37(3):374–383PubMedCrossRef Kannan Y et al (1994) Lymphoid tissues induce NGF-dependent and NGF-independent neurite outgrowth from rat superior cervical ganglia explants in culture. J Neurosci Res 37(3):374–383PubMedCrossRef
81.
Zurück zum Zitat De Ridder L, De Potter RW (1983) Interaction between embryonic rat superior cervical ganglion and syngeneic heart fragments in a confronting culture. Cell Biol Int Rep 7(4):293–301PubMedCrossRef De Ridder L, De Potter RW (1983) Interaction between embryonic rat superior cervical ganglion and syngeneic heart fragments in a confronting culture. Cell Biol Int Rep 7(4):293–301PubMedCrossRef
82.
Zurück zum Zitat Ulupinar E, Erzurumlu RS (1998) Peripheral target-specific influences on embryonic neurite growth vigor and patterns. J Comp Neurol 399(4):427–439PubMedCrossRef Ulupinar E, Erzurumlu RS (1998) Peripheral target-specific influences on embryonic neurite growth vigor and patterns. J Comp Neurol 399(4):427–439PubMedCrossRef
83.
Zurück zum Zitat Schwab M, Landis S (1981) Membrane properties of cultured rat sympathetic neurons: morphological studies of adrenergic and cholinergic differentiation. Dev Biol 84(1):67–78PubMedCrossRef Schwab M, Landis S (1981) Membrane properties of cultured rat sympathetic neurons: morphological studies of adrenergic and cholinergic differentiation. Dev Biol 84(1):67–78PubMedCrossRef
84.
Zurück zum Zitat Rawdon BB, Dockray GJ (1983) Directional growth of sympathetic nerve fibres in vitro towards enteric smooth muscle and heart from mice with congenital aganglionic colon and their normal littermates. Brain Res 283(1):53–59PubMedCrossRef Rawdon BB, Dockray GJ (1983) Directional growth of sympathetic nerve fibres in vitro towards enteric smooth muscle and heart from mice with congenital aganglionic colon and their normal littermates. Brain Res 283(1):53–59PubMedCrossRef
85.
Zurück zum Zitat Rawdon BB (1991) Extension of sympathetic neurites in vitro towards explants of embryonic and neonatal mouse heart and stomach: ontogeny of neuronotrophic factors. Brain Res Dev Brain Res 59(1):49–58PubMedCrossRef Rawdon BB (1991) Extension of sympathetic neurites in vitro towards explants of embryonic and neonatal mouse heart and stomach: ontogeny of neuronotrophic factors. Brain Res Dev Brain Res 59(1):49–58PubMedCrossRef
86.
Zurück zum Zitat Shcherbakova OG et al (2007) Organization of beta-adrenoceptor signaling compartments by sympathetic innervation of cardiac myocytes. J Cell Biol 176(4):521–533PubMedPubMedCentralCrossRef Shcherbakova OG et al (2007) Organization of beta-adrenoceptor signaling compartments by sympathetic innervation of cardiac myocytes. J Cell Biol 176(4):521–533PubMedPubMedCentralCrossRef
87.
Zurück zum Zitat Coughlin MD, Bloom EM, Black IB (1981) Characterization of a neuronal growth factor from mouse heart-cell-conditioned medium. Dev Biol 82(1):56–68PubMedCrossRef Coughlin MD, Bloom EM, Black IB (1981) Characterization of a neuronal growth factor from mouse heart-cell-conditioned medium. Dev Biol 82(1):56–68PubMedCrossRef
88.
Zurück zum Zitat Uchida Y, Tomonaga M (1985) Effects of nerve growth factor and heart cell conditioned medium on neurite regeneration of aged sympathetic neurons in culture. Brain Res 348(1):100–106PubMedCrossRef Uchida Y, Tomonaga M (1985) Effects of nerve growth factor and heart cell conditioned medium on neurite regeneration of aged sympathetic neurons in culture. Brain Res 348(1):100–106PubMedCrossRef
89.
Zurück zum Zitat Coughlin MD, Kessler JA (1982) Antiserum to a new neuronal growth factor: effects on neurite outgrowth. J Neurosci Res 8(2–3):289–302PubMedCrossRef Coughlin MD, Kessler JA (1982) Antiserum to a new neuronal growth factor: effects on neurite outgrowth. J Neurosci Res 8(2–3):289–302PubMedCrossRef
90.
Zurück zum Zitat Ge Y et al (2020) Human epicardium-derived cells reinforce cardiac sympathetic innervation. J Mol Cell Cardiol 143:26–37PubMedCrossRef Ge Y et al (2020) Human epicardium-derived cells reinforce cardiac sympathetic innervation. J Mol Cell Cardiol 143:26–37PubMedCrossRef
91.
Zurück zum Zitat Malliani A, Montano N (2002) Emerging excitatory role of cardiovascular sympathetic afferents in pathophysiological conditions. Hypertension 39(1):63–68PubMedCrossRef Malliani A, Montano N (2002) Emerging excitatory role of cardiovascular sympathetic afferents in pathophysiological conditions. Hypertension 39(1):63–68PubMedCrossRef
92.
Zurück zum Zitat Wang HJ, Rozanski GJ, Zucker IH (2017) Cardiac sympathetic afferent reflex control of cardiac function in normal and chronic heart failure states. J Physiol 595(8):2519–2534PubMedPubMedCentralCrossRef Wang HJ, Rozanski GJ, Zucker IH (2017) Cardiac sympathetic afferent reflex control of cardiac function in normal and chronic heart failure states. J Physiol 595(8):2519–2534PubMedPubMedCentralCrossRef
94.
Zurück zum Zitat Larson TA (2018) Sex steroids, adult neurogenesis, and inflammation in cns homeostasis, degeneration, and repair. Front Endocrinol (Lausanne) 9:205PubMedPubMedCentralCrossRef Larson TA (2018) Sex steroids, adult neurogenesis, and inflammation in cns homeostasis, degeneration, and repair. Front Endocrinol (Lausanne) 9:205PubMedPubMedCentralCrossRef
95.
Zurück zum Zitat Smits AM, Dronkers E, Goumans MJ (2018) The epicardium as a source of multipotent adult cardiac progenitor cells: their origin, role and fate. Pharmacol Res 127:129–140PubMedCrossRef Smits AM, Dronkers E, Goumans MJ (2018) The epicardium as a source of multipotent adult cardiac progenitor cells: their origin, role and fate. Pharmacol Res 127:129–140PubMedCrossRef
97.
Zurück zum Zitat Sadler TW (2005) Embryology of neural tube development. Am J Med Genet C Semin Med Genet 135C(1):2–8PubMedCrossRef Sadler TW (2005) Embryology of neural tube development. Am J Med Genet C Semin Med Genet 135C(1):2–8PubMedCrossRef
99.
Zurück zum Zitat Kanagasuntheram R, Dharshini P (1994) The cervical sympathetic trunk–a new hypothesis. Ann Acad Med Singap 23(6):923–928PubMed Kanagasuntheram R, Dharshini P (1994) The cervical sympathetic trunk–a new hypothesis. Ann Acad Med Singap 23(6):923–928PubMed
101.
Zurück zum Zitat Ichikawa H (2002) Innervation of the carotid body: immunohistochemical, denervation, and retrograde tracing studies. Microsc Res Tech 59(3):188–195PubMedCrossRef Ichikawa H (2002) Innervation of the carotid body: immunohistochemical, denervation, and retrograde tracing studies. Microsc Res Tech 59(3):188–195PubMedCrossRef
103.
104.
Zurück zum Zitat Abdala AP et al (2012) Hypertension is critically dependent on the carotid body input in the spontaneously hypertensive rat. J Physiol 590(17):4269–4277PubMedPubMedCentralCrossRef Abdala AP et al (2012) Hypertension is critically dependent on the carotid body input in the spontaneously hypertensive rat. J Physiol 590(17):4269–4277PubMedPubMedCentralCrossRef
105.
Zurück zum Zitat Bradfield JS et al (2018) Mechanisms and management of refractory ventricular arrhythmias in the age of autonomic modulation. Heart Rhythm 15(8):1252–1260PubMedCrossRef Bradfield JS et al (2018) Mechanisms and management of refractory ventricular arrhythmias in the age of autonomic modulation. Heart Rhythm 15(8):1252–1260PubMedCrossRef
Metadaten
Titel
The relevance of the superior cervical ganglion for cardiac autonomic innervation in health and disease: a systematic review
verfasst von
H. Sophia Chen
Lieke van Roon
Yang Ge
Janine M. van Gils
Jan W. Schoones
Marco C. DeRuiter
Katja Zeppenfeld
Monique R. M. Jongbloed
Publikationsdatum
23.02.2024
Verlag
Springer Berlin Heidelberg
Erschienen in
Clinical Autonomic Research / Ausgabe 1/2024
Print ISSN: 0959-9851
Elektronische ISSN: 1619-1560
DOI
https://doi.org/10.1007/s10286-024-01019-2

Weitere Artikel der Ausgabe 1/2024

Clinical Autonomic Research 1/2024 Zur Ausgabe

Neu in den Fachgebieten Neurologie und Psychiatrie

Blutdrucksenkung schon im Rettungswagen bei akutem Schlaganfall?

31.05.2024 Apoplex Nachrichten

Der optimale Ansatz für die Blutdruckkontrolle bei Patientinnen und Patienten mit akutem Schlaganfall ist noch nicht gefunden. Ob sich eine frühzeitige Therapie der Hypertonie noch während des Transports in die Klinik lohnt, hat jetzt eine Studie aus China untersucht.

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.

Hörschwäche erhöht Demenzrisiko unabhängig von Beta-Amyloid

29.05.2024 Hörstörungen Nachrichten

Hört jemand im Alter schlecht, nimmt das Hirn- und Hippocampusvolumen besonders schnell ab, was auch mit einem beschleunigten kognitiven Abbau einhergeht. Und diese Prozesse scheinen sich unabhängig von der Amyloidablagerung zu ereignen.