Abstract
Numerous clinical studies have revealed the utility of circulating adrenomedullin (AM) or mid-regional pro-adrenomedullin (MR proAM) as an effective prognostic and diagnostic biomarker for a variety of cardiovascular-related pathophysiologies. Thus, there is strong supporting evidence encouraging the exploration of the AM- calcitonin receptor like receptor (CLR) signaling pathway as a therapeutic target. This is further bolstered because several drugs targeting the shared calcitonin gene-related peptide (CGRP) - CLR pathway are already FDA approved and on the market for the treatment of migraine. In this review, we summarize the AM – CLR signaling pathway as well as its modulatory mechanisms and provide an overview of the current understanding of the physiological and pathological roles of AM – CLR signaling and the yet untapped potentials of AM as a biomarker or therapeutic target in cardiac and vascular diseases and provide an outlook on the recently emerged strategies that may provide further boost to the possible clinical applications of AM signaling.
Keywords: Adrenomedullin, Cardiovascular Disease, Biomarker, Therapeutic Target, Clinical Trials
CELLULAR PHYSIOLOGY AND CLINICAL POTENTIAL OF ADRENOMEDULLIN
Biosynthesis
Adrenomedullin (Gene = Adm; Protein = AM) is a 52 amino acid peptide hormone that is a member of the calcitonin family of peptides alongside calcitonin gene-related peptide (CGRP), amylin, and adrenomedullin 2/intermedin. AM was first isolated from the adrenal medulla1, but secreted in high amounts by endothelial cells (EC) and vascular smooth muscle cells2. Similarly to many other hormones, during AM biosynthesis, first preproadrenomedullin, a large precursor protein is synthetized, which is then cleaved into 4 peptides (namely, proAM N-terminal 20 peptide (PAMP)-Gly, mid-regional pro-adrenomedullin 45-92 (MR pro-AM), AM-Gly, and C-terminal pro-AM3. Active AM is produced by the cleavage of C-terminal Gly from AM-Gly. Cytokines4, lipopolysaccharide (LPS)5, pressure overload6, vascular shear stress7, and hypoxia8 have been shown to stimulate AM synthesis and release.
AM has a half-life of 20-25 minutes in the blood circulation9, limiting its application as a biomarker. In contrast, the biologically inactive MR-proADM, cleaved from the precursor protein of AM, has been found to be a stable and reliable molecule acting as a surrogate of AM peptide, independent of its degradation10. In cases where real time assessment of instantaneous AM levels is needed, biologically active AM concentrations might be preferable, but given time constraints in most clinical scenarios, in almost all pathologies, MR proAM has more potential as a biomarker.
AM’s short half-life also impairs its therapeutic potential. In order to reach adequate plasma concentrations of AM, continuous intravenous administration is required. When infusion is halted plasma concentrations drop precipitously, again reflecting the short half-life of AM9. Adrecizumab is a humanized non-neutralizing monoclonal antibody targeting AM, leading to a significant increase in plasma AM levels without changing MR proAM concentrations11, effectively increasing the amount of bioactive AM in the plasma. Adrecizumab presumably functions by preventing the extravascularization or slowing the degradation of AM in the plasma (Table 1), thereby increasing effective plasma levels of AM and elongating the timeframe of its action.
Table 1:
Clinical trials of therapeutic approaches targeting AM-signaling in cardiovascular diseases.
| Disease | Clinical trial title | Type, status |
Disease condition | Agent | Arms | Outcomes |
|---|---|---|---|---|---|---|
| Healthy | Adrecizumab Phase 1 Trial (NCT02991508) | Phase I completed | 24 healthy male participants | Adrecizumab |
|
|
| Healthy | Adrecizumab-LPS Study (NCT03083171) | Phase I completed | 24 healthy male participants i.v. 0 1 ng/kg E. Coli type O113 lipopolysaccharide followed by 1 ng/kg/hour for 3 hours. | Adrecizumab |
|
|
| Sepsis | AdrenOSS-2 (NCT03085758) | Phase IIa completed | 301 patients with adrenomedullin above 70 pg/mL, at early stage of septic shock | Adrecizumab |
|
|
| Cardiogenic shock | Adrecizumabin Cardiogenic Shock (ACCOST-HH) (NCT03989531) | Phase II, completed | 150 patients with cardiogenic shock | Adrecizumab |
|
|
| Heart Failure | Adrecizumab Dose Escalation Safety and Tolerability Evaluation (NCT04252937) | Phase II, recruiting | est. 40 patients with acute heart failure (NYHA class II-IV), within 48h from hospital admission | Adrecizumab |
|
|
| Inflammatory Bowel Disease | Assessment of the Safety, Tolerability, and Pharmacokinetic of HM201 (NCT05088369) | Phase I, recruiting | est. 68 healthy male subjects | HM201 (Pegylated Human Adreniomedullin) | SAD cohorts:
|
|
| Ulcerative colitis (UC) | Adrenomedullin for steroid-resistant ulcerative colitis (Japic CTI-205255 [200410115290]) | Phase IIa completed | 28 patients with steroid-resistant UC | Adrenomedullin |
|
|
| Crohn's disease (CD) | Adrenomedullin for biologic-resistant Crohn's disease (JAPIC CTI-183947 [200612912663]) | Phase IIa completed | 24 patients with biologic-resistant CD | Adrenomedullin |
|
|
Receptors and downstream signaling mechanisms
AM predominantly acts by binding to its class B G protein coupled receptor, calcitonin-receptor like receptor (Gene = Calcrl; Protein = CLR) (Figure 1). G protein-coupled receptors (GPCRs) are 7 transmembrane receptors that play critical roles in many physiological processes through signal initiation and propagation, thereby making them ideal drug targets12. To establish biologically active receptors for the members of the calcitonin family of peptides, CLR must form a heterodimer with one of three receptor activity-modifying proteins (RAMPs1-3); the CGRP receptor (CLR/ RAMP1), the AM1 receptor (CLR/ RAMP2), and the AM2 receptor (CLR/ RAMP3). RAMPs, which were first discovered and characterized due to their relationship with CLR, are single pass transmembrane proteins that interact and allosterically regulate GPCRs. While originally thought to only interact with a select group of GPCRs, there is an ever-expanding list of known RAMP-interacting GPCRs among all GPCR classes13. RAMPs regulate GPCRs in a RAMP-GPCR dependent manner; therefore, the specific CLR-RAMP pairings have broad pharmacological and physiological consequences. Not only does CLR require interaction with RAMPs to be chaperoned to the plasma membrane, the three CLR-RAMP endogenous ligands have differential affinity for the receptor-complex dependent on which RAMP is present. That is, CGRP preferentially binds the CLR/ RAMP1 complex, while AM preferentially binds the CLR/ RAMP2 dimer, followed by the CLR/ RAMP3 coupling. The CLR/RAMP3 heterodimer is also the preferred receptor complex of AM2/intermedin14,15.
Figure 1: Schematic summarization of the intracellular signaling pathways activated by the binding of AM, CGRP and AM2.
Adrenomedullin acts through CLR/RAMP2 and CLR/RAMP3 receptor complexes and activates G-protein mediated intracellular pathways, which lead to cell-type specific cellular effects. ACKR3 degrades AM by recruiting G protein-coupled receptor kinases and β-arrestin.
Human pEC50 values for cAMP production are shown, where applicable (data from Hay et al. 201714). Arrow thickness represent the corresponding pEC50 value. Known cellular effects in blood endothelial cells (BEC), lymphatic endothelial cells (LEC), vascular smooth muscle cells (SMC) and myocardial cells (myoC) are indicated.
There has been exciting advancement in our understanding of CLR-RAMP signal initiation, propagation, and desensitization over the last several years. It is well established that ligand (CGRP, AM, AM2) binding to CLR-RAMP complexes results in intracellular cAMP accumulation and calcium mobilization. That being said, using modified yeast strains and HEK-293 cells, Weston et al. show that CLR-RAMP complexes can couple with multiple types of Gα subunits and demonstrate that coupling to these alternative Gα subunits alters the ligand binding potencies of the CLR-RAMP receptor complexes16. After agonist-stimulation, besides activation of Gα mediated signaling pathways, β-arrestins-1 and −2 are also recruited to ligand-bound CLR receptor complexes, and are required for the rapid internalization, endosomal sorting, and desensitization of these receptor complexes. However, while full agonism does not require β-arrestins-1 and −2 recruitment, their recruitment may initiate signaling cascades independent of Gαs17. Excitingly, CLR-RAMP complexes are among a growing list of GPCRs shown to continue to signal from endosomes and not solely just at the plasma membrane. For example, Yarwood et al. demonstrate that CLR-RAMP1 signaling endosomes promote migraine pain transmission18. Downstream intracellular consequences of cyclic adenosine monophosphate (cAMP) production, increase in [Ca2+]IC levels, β-arrestins-1 and −2-mediated recruitment and internalization, and endosomal signaling result in the activation of downstream effectors such as mitogen-activated protein kinase (MAPK) cascade as well as the phosphorylation of AKT and cAMP response element-binding protein (CREB) to initiate cellular responses and nitrogen monoxide (NO) production15. Recent articles found that AM-CLR signaling preferably signals through Gαs-mediated signaling mechanisms over Gαq in Cos7 and HEK293s cell lines transfected with CLR-RAMP receptor constructs 19,20. On the other hand, Clark et al. demonstrated that AM and AM2-dependent [Ca2+]IC mobilization can be impeded by co-treatment with an anti- Gαq/11/14 inhibitor in human umbilical vein endothelial cells (HUVECs) 15. Moreover, HUVECs and human umbilical arterial endothelial cells (HUAECs) show different downstream signaling bias15, highlighting the importance of the cell type for CLR signaling. It has been also demonstrated that AM signaling can lead to a ligand-independent transactivation of the vascular endothelial growth factor receptor 2 (VEGFR2) in HUVECs and immortalized endothelial cell lines21, and VEGFR3 in lymphatic endothelial cells (LECs) 22, respectively. Therefore, besides Gαs signaling, other alternative intracellular mechanisms might also contribute to AM-dependent downstream cAMP production in endothelial cells. Further studies elucidating the downstream signaling preference of AM on different cell types and unveiling the underlying molecular mechanisms may help us understand the cell type-specific properties of AM signaling.
As discussed more in detail below, both increased and decreased AM may lead to pathology. Therefore, a tight regulation of the biological levels of AM peptide by ACKR3 is essential for proper AM function. Atypical chemokine receptor 3 (ACKR3, other common aliases: CXCR7, RDC-1, GPR159) is a 7 transmembrane receptor, with the highest expression in fibroblasts and ECs23, proposing its importance during tissue repair and in vascular physiology. Currently, known ligands of ACKR3 include chemokine x ligand 11 (CXCL11, also known as I-TAC), CXCL12 (also known as SDF-1)24, macrophage migration-inhibitory factor 25, virus-encoded chemokine vCCL2/vMIP-II26, endogenous opioid peptides27,28, PAMPs 29, and AM30. Upon ligand binding, ACKR3 recruits G protein-coupled receptor kinases that phosphorylate the C-terminal tail of ACKR3 recruiting arrestin which in turn promotes ligand internalization and degradation31-34. Through this mechanism, ACKR3 eliminates the bound AM molecules and thereby regulates its function35.
Current therapeutic applications
Because of their crucial physiological and pathological roles and pharmacological tractability, GPCR signaling pathways are intensively studied drug targets. Approximately 1/3 of the FDA-approved drugs act on GPCRs12. Currently a highly successful clinical approach targeting the AM receptor CLR is being applied for the inhibition of the AM-related peptide CGRP in migraine patients. CGRP is a neuropeptide that is produced by Aδ and C nerve fibers and functions in pain sensation and vasodilation36-38. Given these functions, it is a compelling biomarker for various human pathophysiologies, the foremost being migraine39. In 1988 CGRPs role in activation of the trigeminovascular system was described by Dr. Lars Edvinsson40 and in 2003 elevated CGRP was identified in the cerebrospinal fluid in migraine patients41. A later study demonstrated CGRP elevation in jugular venous blood of patients with migraines42, suggesting it as a potential biomarker for migraine. Additionally, a recent meta-analysis of genome wide association studies examining risk loci in migraine has identified polymorphisms in the gene encoding CGRP (Calca/Calcb) as a novel risk locus in migraine43. Migraine is extremely responsive to anti-CGRP targeting medications such as monoclonal antibodies targeting a shared epitope between CGRP/RAMP1 (erenumab)44 or soluble CGRP (fremanezumab, galcanezumab, and eptinezumab) or small molecule receptor antagonists (‘gepant class medications)45. Since 2018, four CGRP-inhibitor monoclonal antibodies and three small molecule inhibitors have been approved to date.
The successful therapeutic application of agents targeting CLR/RAMP1 signaling demonstrates that CLR signaling is a clinically targetable pathway with a high clinical efficacy. Thus, it stands to reason that CLR/RAMP2 and CLR/RAMP3 heterodimers may also be exploited as therapeutic targets for manipulating AM signaling. Therefore, understanding the physiological functions of AM and its roles in disease helps us to evaluate the clinical potential of AM and to determine future directions for its use as a biomarker and therapeutical target.
VASCULAR AND CARDIAC FUNCTIONS OF ADRENOMEDULLIN
Vasoactive functions
AM peptide is best known for its vasoactive functions. As a part of its intracellular signaling cascades, AM increases intracellular cAMP and Ca2+ levels in endothelial cells, leading to endothelial nitric oxide synthase (eNOS) activation and NO formation7,15,46. Endothelial NO then diffuses to the surrounding vascular smooth muscle (VSM) cells, promoting smooth muscle relaxation and vasodilation. Other preclinical results demonstrated that AM also acts on VSM cells directly, and promotes VSM relaxation through increasing cAMP levels and inhibiting endothelin production47. A clinical study demonstrated that intravenous administration of 50 ng·kg−1·min−1 AM is efficient to decrease mean arterial blood pressure of healthy subjects48, and the authors suggested that this AM-mediated vasodilation mechanism contributes to basal vascular tone regulation. A recent article also demonstrated this role of AM signaling in basal blood pressure control 7. Regardless, because AM has many known functions, it is unlikely to be useful as a specific therapeutic agent in the management of hypertension.
Vascular development and remodeling
Besides regulating basal vascular tone, AM also contributes to numerous other cardiovascular mechanisms. Adm null and Calcrl null mouse embryos do not display abnormal vascular expansion, but studies reported reduction in arterial wall thickness and VSM coverage and abnormal basal membrane structure in the larger vessels. These findings suggest a role for AM in vascular remodeling and maintaining vascular barrier integrity, but not in embryonic angiogenesis49-51.
Cardiac development
The role of the AM/CLR/RAMP pathway in cardiac development has been extensively studied. AM deficient mouse embryos die at mid-gestation due to hydrops fetalis and cardiovascular defects, including overdeveloped ventricular trabeculae, underdeveloped arterial walls with reduced myocardial proliferation and increased apoptosis49. Deletion of AM receptors Calcrl, or Ramp2 also leads to embryonic lethality characterized by hydrops fetalis, thin vascular smooth muscle walls, and small disorganized hearts51. Additionally, Ramp2 null embryos display systemic and cardiac edema, small hearts, and vascular defects including vascular wall structure disruption, diminished junctional protein expression, increased vascular permeability, and diminished neovascularization52,53. Thus, the embryological phenotypes of these deficient mouse lines is conserved, supporting a requisite role for this signaling pathway in cardiac and vascular development.
In contrast, Admhi/hi mice overexpressing AM have enlarged hearts due to hyperplasia, which can be reversed with reduction of AM levels54. Similarly, mouse embryos lacking the AM decoy receptor ACKR3 develop enlarged hearts due to cardiomyocyte hyperplasia and die by postnatal day 1 due to cardiac valve defects55,56. Similar to Admhi/hi mice, genetic reduction of AM in the ACKR3 null mice is sufficient to reverse the embryonic cardiac hyperplasia35.
Lymphatic development
The lymphatic vasculature is an open-ended vessel system that functions primarily to drain excess interstitial fluid, traffic leukocytes to immune organs, and facilitate dietary lipid absorption, among many other tissue-specific roles57. In mice, development of the lymphatic system starts around embryonic day 10.5, when lymphatic vessel endothelial hyaluronan receptor 1 (LYVE1) / prospero homeobox protein 1 (PROX1) double positive endothelial cells first appear in the cardinal vein. These cells bud from the cardinal vein and form the jugular lymph sac, the first lymphatic structure58. Eventually, a mature lymphatic vessel network derived from both venous and non-venous LEC precursors develops to enmesh most vascularized organs59.
PROX1 is a key transcription factor for the determination and maintenance of lymphatic identity and function and is continually expressed by lymphatic endothelial cells. Overexpression of PROX1 in LECs has been shown to induce Calcrl expression levels. Because Prox1 is a key transcription factor for maintaining lymphatic cell fate, it is not surprising that expression levels of Adm, Calcrl and Ramp2 are significantly higher in cultured LECs than blood endothelial cells (BECs)52. Not surprisingly, Adm−/−, Calcrl−/− and Ramp2−/− mouse embryos all display lymph sac hypoplasia at E13.552, and hydrops fetalis49,51,60, revealing the importance of AM signaling in lymphatic development. The relevance of the AM/CLR/RAMP2 signaling pathway for human lymphatic development and survival was also uncovered from whole exome sequencing of a 4-generation family pedigree, revealing a recessive in frame mutation in the CALCRL gene that was homozygous in two infants with nonimmune hydrops fetalis. Biochemical characterization of the receptor mutation showed reduced association with RAMP2 protein, thereby precluding receptor presentation to the cell surface60.
Other studies have shown that AM treatment stabilizes LEC intercellular junctions in vitro, and AM administration improves lymphatic barrier function and decreases lymphatic permeability in vivo61. This suggests that disruption of the AM/CLR/RAMP2 signaling axis leads to systemic edema, at least partially due to impaired lymphangiogenesis and altered lymphatic vessel permeability. Of note, ACKR3 deficient mouse embryos however, had blood-filled, enlarged lymph sacs interstitial edema, and dilated and aberrant lymphatic vasculature, emphasizing that precise regulation of AM levels and signaling is crucial for lymphatic vessel function35.
In adult mice, inducible postnatal global deletion of CLR results in disorganized subcellular organization of the junctional proteins vascular endothelial cadherin (VE-Cadherin) and zona occludens 1 (ZO-1), leading to disrupted lymphatic vessel permeability and impaired dietary lipid absorption62-64, highlighting the central role of AM signaling in maintaining proper lymphatic function in adulthood. Because VE-Cadherin and ZO-1 are also important junctional proteins in the blood vasculature, it is likely that AM promotes blood vascular barrier integrity through a similar molecular mechanism. Furthermore, VE-Cadherin has been shown to be essential for maintaining pro-lymphangiogenic VEGFR3 signaling nodes at the plasma membrane in LECs22, providing additional mechanisms through which AM signaling exerts its pro-lymphangiogenic effects.
In summary, AM plays important roles in cardiac development, lymphatic growth, and vascular permeability. It is likely that upregulation of AM promotes cardiac tissue regeneration, regulates immune responses, helps resolve cardiac edema after injury, and reduces fibrosis following injury by the mechanisms discussed above in detail. In Figure 2, we summarize these diverse biological functions of AM, and mark which of these functions contribute to cardiovascular pathologies or to diseases with significant cardiovascular involvement as discussed below.
Figure 2: Involvement of AM-related biological functions in cardiovascular diseases.
Adrenomedullin acts on a diverse range of tissues generally favoring reduced cellular barrier permeability, angiogenesis, and has significant impact on cardiovascular parameters. Background colors distinguish the different types of AM’s function (modulating barrier function, promoting angiogenesis, vasoactive and cardiac functions and non-vascular functions). The involvement of these AM-mediated mechanisms in cardiovascular related diseases is summarized above and grouped by physiologic role of AM. Checkmarks mark which AM functions contribute to each disease. Color of the checkmarks label whether that given function of AM is associated with blood endothelial, lymphatic endothelial cells or other cell types.
PATHOPHYSIOLOGICAL ROLES AND POTENTIAL THERAPEUTIC APPLICATIONS OF AM IN CARDIOVASCULAR DISEASE
AM has potent cardiovascular activity. As a potent vasodilator, it reduces arterial blood pressure, decreases peripheral vascular resistance, thereby increasing heart rate and cardiac output65. Moreover, it is also essential for maintaining vascular integrity66 and promotes angiogenesis under hypoxic conditions67. Given these functions of AM in regulating cardiovascular physiology and development, it is not surprising that AM also plays a role in numerous cardiovascular diseases. Here, we provide a brief summary of those conditions where AM has emerged as a biomarker or therapeutic target due to its cardiovascular functions.
Heart failure
Currently, natriuretic peptides, such as atrial natriuretic peptide (ANP) and B-type natriuretic peptide (BNP) and their prohormones proANP and proBNP are commonly used prognostic biomarkers in patients with heart failure. Natriuretic peptides are secreted from the atria and ventricles and provide an accurate prediction on heart failure (HF) mortality and hospitalization68. Clinically however, ANP/BNP utility is limited by high variability and these biomarkers of heart failure are not sufficient to guide management of HF alone. Therefore, additional predictive molecular markers could help to improve both the specificity and sensitivity of HF related clinical testing.
Shortly after the discovery of AM, studies reported a significant increase in plasma AM levels in heart failure patients69,70, proposing AM as a potential prognostic biomarker of HF. Clinical studies have reported high levels of MR-proAM and bioactive AM as independent prognostic biomarkers of hemodynamic impairment, tissue congestion, organ dysfunction and to predict mortality risk of HF patients (Table 2)69,71-74. While some researchers proposed MR-proAM 45-92 as a more accurate prognostic marker of HF than the natriuretic peptides71,75-77, other studies questioned the additional prognostic value of MR-proADM78-80. As discussed in detail in this manuscript, AM is involved in numerous physiological or pathophysiological processes. Therefore, multiple underlying mechanisms might be responsible for the high levels of plasma AM or MR proAM. Combination of MR proAM with additional biomarkers will improve the specificity of AM as a biomarker.
Table 2:
Clinical trials investigating the diagnostic use of AM-signaling in cardiovascular diseases.
| Disease | Clinical trial title | Type, status |
Disease condition | Investigated biomarkers |
Outcomes |
|---|---|---|---|---|---|
| Acute heart failure | Lithuanian Echocardiography Study of Dyspnea in Acute Settings (LEDA) (NCT03048032) | Completed | 643 patients with acute heart failure, dyspnea, and 545 control subjects | Bio-AM NT-proBNP Troponin T CRP |
|
| Acute Heart Failure | Biomarkers in Acute Heart Failure (BACH) (NCT00537628) | Completed | 568 patients with acute heart failure, Dyspnea | MR proAM Copeptin BNP NT-proBNP MR proANP PCT Troponin T Troponin I |
|
| Sepsis | Pro-adrenomedullin as a Prognostic Marker in Neonatal Sepsis (NCT01362504) | Completed | 73 newborn infants with neonatal sepsis and 194 control subjects | MR-proAM CT-proET-1 MR-proANP (PCT). |
|
| Sepsis | Adrenomedullin and Outcome in Severe Sepsis and Septic Shock (AdrenOSS) (NCT02393781) | Completed | 583 patiens with severe sepsis, septic shock | Bio- AM BNP NT-proBNP PCT Troponin T Troponin I |
|
| Pulmonary hypertension | PulmoBind (NCT02216279) | Phase II, completed | 30 patients with Pulmonary arterial Hypertension (PAH, n=23), or chronic thromboembolic Pulmonary Hypertension (PH, n=7), 15 healthy controls after injection of 15 mCi 99mTc-PulmoBind in supine position | AM |
|
| Pancreatic cancer | Tumour Regulatory Molecules in Early Pancreatic Cancer Detection (TEM-PAC) (NCT03536793) | Recriuiting | est. 50 patients with pancreatic cysts, 50 patients with pancreas cancer, and 80 healthy controls | Bio-AM TF |
The clinical potential of AM in heart failure is not limited to its application as a biomarker. An early ex vivo study revealed the positive inotropic effect of AM81, by which AM can increase stroke volume and therefore cardiac output and systolic blood pressure82. On the other hand, however, vasodilatory effects of AM combined with its positive inotropic properties may promote the development of hyperdynamic circulation (Figure 2). An ongoing phase II clinical study is aiming to evaluate safety and tolerability of an Adrecizumab in patients with acute HF. This suggests an alternative approach for targeting AM in HF patients.
Myocardial Infarction
Heart disease is the leading cause of death and over 800,000 people have myocardial infarction (MI) annually in the United States only 83. Though MI is generally treated by coronary artery catheterization or with thrombolytic agents, novel diagnostic and therapeutic approaches are desirable in order to improve long term outcomes and provide additional information about end organ damage. Early studies reported an immediate elevation in plasma AM levels following myocardial infarction84. Recent clinical data have also indicated that MR-proAM is a reliable predictive marker to predict long-term mortality, volume overload, and congestion during recovery from MI, but similar to HF, the additional value to the currently used biomarkers is questioned (Table 2) 85,86.
In addition to ECs and smooth muscle cells, mesenchymal stem cells have also been reported to contribute to AM expression after myocardial infarction 87. An early study reported elevated ANP secretion in AM-treated cultured rat myocytes, and reduced collagen production in AM-treated cultured fibroblasts88. Preclinical studies found that AM administration after myocardial infarction reduced pathologic tissue remodeling and fibrosis formation post injury, suggesting AM as a possible therapeutic target after MI89,90. Similarly, cardioprotective effects of AM administration were shown in preclinical HF models91,92. Pilot clinical studies have suggested intravenous infusion of AM improves cardiac outcomes in patients recovering from acute MI93 and that co-treatment with ANP as a potential therapeutic approach for patients with acute decompensated HF94.
AM overexpressing Admhi/hi mice have a more robust cardiac lymphangiogenic response following MI which was correlated with improved cardiac function and reduced cardiac edema and tissue remodeling when compared to wild type controls95. Moreover, a recent study suggested that lymphatic vessels promote embryonic heart development as well as MI recovery mediated by reelin secretion96. These findings suggest that the cardioprotective effects of AM might be partly related to its pro-lymphangiogenic function. The currently available experimental results suggest a reactivation of embryonic epicardial gene expression program upon cardiac injury, including the epicardial expression of AM97.
Furthermore, the current body of literature suggests that AM plays an important role in cardiac function and recovery following insult, indicating a potential therapeutic role for AM in numerous cardiac diseases. AM’s short half-life significantly decreases its therapeutic potential. Therefore, pharmaceutical strategies to increase the half-life of AM might improve its usefulness as a therapeutic agent.
Pulmonary hypertension and congestion
Plasma AM and MR pro-AM levels have been suggested as reliable prognostic markers in patients with pulmonary hypertension98,99. Moreover, inhalation of aerosolized AM was shown to improve survival as well as reduce pulmonary arterial pressure and pulmonary vascular resistance in rats with monocrotaline-induced pulmonary hypertension (Figure 2)100. Although clinical pilot studies have proposed AM as a promising therapeutic target to treat pulmonary hypertension101,102, its short half-life is once again a significant obstacle in AM’s therapeutic potential. Though pulmonary congestion is associated with higher mortality in HF patients103,104, assessment and staging of pulmonary congestion remains challenging and lacks standardization105-107. A recent study found AM to be the strongest clinical predictor of pulmonary congestion74, suggesting that despite the debate over the added value of AM testing to predict mortality risk in HF patients, AM levels may be a valuable tool in determining therapeutic approach.
Importantly, pulmonary hypertension and congestion are not the only pulmonary vascular conditions where clinical application of AM have emerged. A promising diagnostic approach takes advantage of the fact that CLR expression is abundant in the alveolar endothelium108. PulmoBind, a radiolabeled AM derivative that can be used to detect pulmonary microcirculatory occlusions and abnormalities with single-photon emission computerized tomography (SPECT) has passed phase I and II clinical trials109,110. This represents a promising tool for the early detection of pulmonary embolism and of other pathophysiologies that alter circulation through the pulmonary vasculature.
Stroke
Under pathophysiological circumstances, such as ischemia, preclinical studies demonstrated that AM promotes blood and lymphatic vessel expansion111-113, suggesting AM as a potential biomarker and therapeutic target for ischemic diseases. Moreover, a preclinical study found anti-apoptotic effects of AM infusion on neuronal cells and transplanted mesenchymal stem cells 114, showing that direct vascular and non-vascular effects of AM might improve stroke outcomes. An earlier human study found elevated AM levels in the cerebrospinal fluid (CSF) but not in the plasma of patients with cerebral vasospasm after hemorrhage115. Other researchers reported elevated plasma AM and mid-regional pro-AM levels in both ischemic and acute hemorrhagic stroke patients116-118. These findings suggest AM as a potential diagnostic biomarker of stroke. Moreover, its neuroprotective properties, such as reducing neuron apoptosis and promoting oligodendrocyte differentiation119-121, make AM an even more promising therapeutic target in stroke (Figure 2). A current phase II study aims to use AM as a treatment option for ischemic stroke122.
Sepsis
Sepsis is a life threatening extreme immune reaction of the body to disseminated infection. AM and MR pro-AM levels both are increased in patients with sepsis, and correlate with increased disease severity and mortality risk123-126. The use of AM as a morbidity-independent sepsis biomarker would help predict illness severity and inform personalized therapy decisions for septic patients. The therapeutic effect of increasing AM as a treatment strategy in sepsis has also been intensively studied. Administration of AM exerts anti-inflammatory, antimicrobial, and protective effects on endothelial barrier function during sepsis127. On the other hand, as AM has both vasodilatory and positive inotropic roles, elevated AM can lead to hyperdynamic circulation, which may contribute to sepsis severity and push the patient toward developing septic shock (Figure 2). Adrecizumab has been shown to the improve vascular barrier maintenance function of AM but does not promote AM detrimental vasoactive effects128, which might be beneficial for septic patients. Of note, it is currently it is not known if Adrecizumab also alters other functions of AM. A phase II clinical trial of 301 patients found that both 2 and 4 mg/BWkg adrecizumab is well tolerated and organ failure scores were significantly improved compared to placebo (Table 2). Additionally, 28-day mortality trended lower in adrecizumab treated septic patients than placebo controls, but was not significant (Table 1)129.
Other diagnostic potentials of AM in cardiovascular diseases
Adrenomedullin has recently been implicated in a variety of cardiovascular diseases, expanding its diagnostic and therapeutic potential. Interestingly, in contrast to the low AM expression levels in adult heart tissue under physiological conditions, increased AM immunoreactivity was found to be correlated with myocyte hypertrophy in the endomyocardium of transplanted hearts in contrast to the low levels of AM expression in adult, non-hypertrophic cardiac tissue under physiological conditions130. MR pro-AM has recently emerged as a promising prognostic biomarker of aortic stenosis and severity and to predict mortality risk after transcatheter aortic valve replacement131,132, which is probably also related to AM’s vasoactive function.
Lymphedema
The lymphatic system plays an important role in maintaining tissue fluid homeostasis. Failure or impaired function of the lymphatic vascular system results in disfiguring, disabling, and sometimes life-threatening swelling of the affected tissues, called lymphedema133. Lymphedema occurs as a result of a hereditary genetic mutations in the genes involved in lymphatic development and function (primary lymphedema), or after surgical removal of lymph nodes, radiation therapy, or parasite infections (secondary lymphedema)134. Despite its high incidence and serious effects on the patients’ quality of life, no effective treatment is available for lymphedema currently. As AM signaling has important roles in promoting lymphangiogenesis and modulating lymphatic function, it is a promising therapeutic target to treat patients with lymphedema.
In addition to the severe edema reported in AM, CLR and RAMP2 deficient mouse embryos, other studies also support the anti-edematous effects of AM signaling. In addition, mice lacking RAMP3, although do not show embryonic edematous phenotype, have impaired lymphatic drainage. Moreover, in a mouse model of lymphedema, RAMP3 deficient mice represent more severe edema and display defective lymphatic cell migration when challenged with scratch wound assay135. This suggests the importance of CLR/RAMP3 mediated signaling in lymphedema in addition to CLR/RAMP2 signaling. Accordingly, dilated dermal lymphatics were reported after induced global deletion of CLR. CLR deleted mice developed a much more severe local edema after hind paw injection of complete Freund’s adjuvant62. AM haploinsufficient mice, but not wild type controls developed a lymphedematous phenotype following hind limb skin incision, which could be restored by exogenous AM administration136. Following hind limb skin incision elevated AM expression was found in both wild type and AM haploinsufficient mice compared to baseline, indicating that AM signaling was triggered by the surgery. In addition, another research group reported that continuous AM administration promoted the angiogenic and lymphangiogenic response and resolved lymphedema severity in a mouse model of lymphedema (Figure 2)112.
These preclinical studies suggest that AM is a potent therapeutic target to treat lymphedema. For its therapeutic use in patients with lymphedema however, a reliable delivery method that provides long lasting local AM signal is required, which is currently not available.
CARDIOVASCUALR CORRELATES IN OTHER PREVALENT DISEASES
Tumors
There is a large and ever-expanding amount of evidence linking AM as well as the AM receptors (AM1, AM2, and ACKR3) to cancer pathogenesis and metastasis. Vázquez et al. 2021 have provided a comprehensive review highlighting much of this research, including preclinical studies assessing the efficacy of anti-AM therapies in neoplasia (ie. AM-neutralizing antibodies)137. Overall, the pro-tumorigenic properties of AM are not surprising given that AM has known roles in lymphangiogenesis and angiogenesis, cellular proliferation, and cell survival138. A variety of solid tumors as well as other stromal cell-types present within the tumor microenvironment, such as endothelial cells, CAFs and TAMs, express the AM and its receptors (CLR/RAMP1, CLR/RAMP2, CLR/RAMP3, ACKR3)137,139-141. AM signaling promotes tumor cell growth, proliferation, and survival139. Second, AM secreted within the tumor microenvironment facilitates tumor-associated angiogenesis and -lymphangiogenesis, thereby providing the tumor microenvironment with nutrients and oxygen to promote continued survival and growth and contributing to metastasis formation. 142,143 (Figure 2). Importantly, AM and CLR overexpression is correlated with a worse prognosis, disease severity, and relapse for a variety of cancers such as in acute myeloid leukemia (AML)144-149. However, these findings are context- and cancer-dependent whereby AM is associated with a more favorable prognosis for other cancer types such as triple-negative breast cancer 150. That being said, the contribution of AM and the AM receptors to cancer pathogenesis is undeniable, sparking a new wave of anti-AM therapies that are currently being assessed for efficacy in various clinical trials137.
Pneumonia
Besides hypoxia, pro-inflammatory cytokines and LPS also promote AM release, so that infections and inflammatory responses trigger AM secretion. Broadly, the inflammatory condition of pulmonary pneumonia infection, resulting in tissue damage and alveolar fluid accumulation, has been closely associated with pathophysiological levels of AM peptide, which, given its vascular roles, might increase local blood flow and promote a lymphangiogenic response, supporting local immune responses and edema resolution. A study found increased proAM levels in community-acquired pneumonia (CAP) patients who died during follow-up compared with survivors. The researchers found that pneumonia severity index (PSI) combined with plasma proAM levels more accurately predicted mortality than PSI alone151, indicating adrenomedullin levels are a useful metric in determining the severity of pneumonia. The prognostic accuracy of proAM to predict the outcome of CAP was also confirmed by additional studies152,153, and its usefulness was not affected by various etiologies of pneumonia infection154. A recent study reported that serum AM levels are also a promising predictive marker in the assessment of ventilator associated pneumonia (VAP) diagnosis155.
Of note, it has long been demonstrated that AM also has an antimicrobial role, participating in the prevention of local infection acting synergistically with the immune response (Figure 2)156. Interestingly, antimicrobial effects of AM seem to be independent from its classical signaling pathway. Instead, AM was demonstrated to disrupt and permeabilize cell walls of E. Coli and S. Aureus157. These antimicrobial effects of AM, in combination with its vascular functions, such as improving lymphatic function, suggest that in addition to being a valuable prognostic biomarker, AM might also be a potential antimicrobial agent for infectious diseases like pneumonia.
Glaucoma
Glaucoma is an ocular disease primarily mediated by elevated intraocular pressure, eventually leading to vision loss due to damage of the optic nerve. An early pilot clinical study found higher AM levels in the aqueous humor of patients with primary open-angle glaucoma compared to neovascular glaucoma158. Accordingly, a pre-clinical study reported that CLR overexpression in the pupillary sphincter muscle led to increased intra-ocular pressure which was reversed when crossed to AM haploinsufficient mice159. AM and its receptors have both been found to be expressed in the retina160, and genetic mutations in the human RAMP2 gene have been associated with primary open-angle glaucoma161. An interesting study demonstrated reduced pore formation in the ECs of the hybrid vessel Schlemm’s canal in patients with glaucoma162. AM signaling is known to play an important role in regulating intercellular junctions in ECs and modulate their barrier function. Increased AM signaling might enhance the barrier function, reducing the uptake capability of Schlemm’s canal leading to decreased intra-ocular fluid drainage (Figure 2). Further studies are needed to understand the role of AM signaling in glaucoma and to unveil whether it can be as a diagnostic marker or therapeutic target in glaucoma.
Inflammatory Bowel Disease
AM is also widely expressed throughout the highly vascularized system of the gastrointestinal (GI) tract. AM producing cells are spread throughout the GI endocrine system, including the mucosal epithelium, glandular duct cells, enteroendocrine cells, and smooth muscle cells, indicating its important roles in digestion and nutrient absorption163,164. In addition to its various functions in non-endothelial cells of the GI tract 165-167, AM was reported to regulate lipid transport through lacteal LECs. Induced deletion of CLR in PROX1 positive cells led to lymphatic insufficiency and lymphangiectasia63, as well as disrupted lipid trafficking and enteric nerve patterning in HFD fed mice64. Similarly to its role in ECs, AM is also critical in maintaining intestinal epithelial barrier integrity (Figure 2). Lack of endogenous AM accelerates the development and severity of colitis induced by rectal instillation of 3 mg 2,4,6-trinitrobenzenesulfonic acid (TNBS) in mice mitigates the expression levels of junctional adhesion molecule-A (JAM-A) and e-cadherin168. Moreover, reduced AM expression resulted in altered colonic microbiota with a lower proportion of beneficial bacteria in mice, such as Lactobacillus gasseri and Bifidobacterium choerinum, which was correlated with significantly worse colitis169. The broad functions of AM in the GI tract propose multiple possible applications of AM in GI disorders. Currently, AM is considered as a potential therapeutic approach for inflammatory bowel disease170-173. Two phase II studies have suggested 174,175 the clinical potential of AM for treatment of inflammatory bowel disease (IBD) patients.
BOOSTING THE CLINICAL POTENTCY OF AM-TARGETING APPROACHES
As demonstrated above in detail, AM has important roles in cardiovascular physiology and pathophysiology. Given these functions, AM has been proposed as a promising predictive biomarker and therapeutic target for multiple diseases. On the other hand, the broad, systemic roles of AM could also lead to off target effects of AM. Despite the promising clinical potential of AM, there are currently no diagnostic or therapeutic approaches that exploit AM signaling in clinical practice. However, recent advances have provided numerous tools which may help us to leverage the key roles of AM signaling in this wide spectrum of diseases. These include the diagnostic use of MR proAM and the therapeutic application of adrecizumab, which both help to overcome the limitation of the short half-life of AM. In this section, we summarize other current approaches for developing AM-targeting pharmaceuticals that may facilitate or improve the clinical usefulness of AM signaling.
Improving pharmacologic abilities of AM by peptide modifications
Besides reducing the degradation of AM by non-neutralizing antibodies, such as Adrecizumab, another approach to increase the half-life of AM is biochemical modification of the peptide by methods such as PEGylation, acylation, or additional modifications to the peptide structure. These biochemical modifications often decrease the potency or the affinity of the signal for its receptor176,177. Encouragingly, an acylated α-CGRP analogue has been demonstrated to reduce end organ damage in a model of murine hypertension178. The Pioszak laboratory has developed CLR/Ramp agonists for both AM and CGRP that have improved receptor affinities down to the picomolar range and have receptor dwell times ranging from 5 to 10 hours179. This group has also designed sustained signaling AM (ss-AM) and CGRP (ss-CGRP) that increase cAMP levels in stimulated cells after washout of CLR/Ramp agonist179. Currently, the only clinically approved therapeutics that target CLR/Ramps are CGRP antagonists for migraine, these modified proteins represent exciting therapeutic avenues for CLR/Ramp agonists in the context of heart and vascular disease.
The Hay laboratory has developed small molecule agonists of the CLR receptor that are capable of upregulating cAMP in vascular endothelial cells 180. Hsu and colleagues have also successfully developed CLR/RAMP receptor agonists and antagonists that were up to 100-fold more selective to their respective receptor complexes as the natural ligands181. In their later studies, the researchers could evoke a sustained activation of CLR/RAMP1 and CLR/RAMP2 receptor signaling pathways using palmitoylated AM and AM2/IMD analogues182. Moreover, the selected AM analogue was demonstrated to have gel-like characteristics, limiting its spatial spreading. Therefore, application of these gel-forming AM analogues would also provide a predominantly local stimulation of AM signaling pathways. It is yet to be investigated whether these analogues bear the same intracellular signaling bias as AM in the different cell types, or act through a distinct signaling preference.
Therapeutic Approaches to Increase Plasma AM levels
Sacubitril/valsartan is an FDA- and EMA-approved angiotensin receptor - neprilysin inhibitor agent to treat patients with chronic heart failure with reduced ejection fraction (HFrEF). A recent clinical study aimed at characterizing the changes in natriuretic peptide levels after neprilysin-inhibition treatment of patients with HFrEF. Surprisingly however, Pavo and colleagues found that neprilysin-inhibition evoked a modest change in plasma BNP levels, but instead led to a more robust elevation in plasma bio-AM and MR proAM183. Their results also suggest that neprilysin-inhibition probably not only mitigates AM degradation, but also promotes AM production by a yet unknown mechanism. These data propose administration of neprilysin inhibitors as a possible alternative indirect approach to increase plasma levels of biologically active AM in those patients where its use is not contraindicated.
As demonstrated above in this review, another known mechanism that is responsible for the biodegradation of AM is through its decoy receptor ACKR3. Diphenylacetamides were recently discovered to inhibit ACKR3 signaling184. Administration of these pharmacological agents may be a suitable way to evoke increased AM levels. Besides small molecule inhibition of neprilysin and ACKR3, biological targeting of these molecules is also a promising option to target AM signaling.
Potential Approaches for Targeted AM Treatments
As AM receptors are expressed in various cell types all around the body, and AM binding to CLR can evoke different intracellular signaling pathways, systemic administration of AM might lead to undesired effects, as seen in sepsis. Discovering molecular mechanisms that determine which intracellular signaling pathway is activated by AM binding is essential for developing more targeted therapeutic approaches. Development of engineered AM variants or AM agonists that evoke only a distinct downstream mechanism out of the many know AM signaling pathways may be a possible approach to mitigate off-target AM effects. Developing specific AM or CLR inhibitors that mitigate undesirable AM effects without affecting the desired intracellular mechanism might also improve the clinical potential of AM signaling. Developing an administration technique enabling tissue-specific delivery of AM, such as a cell type-specific antibody labeled coating of the bioactive peptide, would help reducing off-target effects of AM. Encapsulation of AM into an enzymatically degradable coating might also help titrate desired AM concentrations locally and control the dynamics of AM administration.
CONCLUDING REMARKS
Adrenomedullin, predominantly known for its vasoactive effects exerts numerous additional biological functions, especially in the cardiovascular and lymphatic vascular systems. In addition to regulating vascular flow by promoting vasodilation, it is indispensable for maintaining endothelial barrier function of blood and lymphatic vessels. AM signaling is essential for cardiac and lymphatic development and under pathological conditions, it may promote adult angiogenesis and lymphangiogenesis and cardiac regeneration after injury. This wide diversity in its physiological and pathophysiological roles elevates AM as a promising biomarker and therapeutic target in numerous cardiovascular diseases, and consequently multiple therapeutic approaches targeting AM signaling are under clinical testing or trials. Recent advances in the understanding of AM-mediated pathways and development of novel approaches, such as AM-specific antibodies increasing its high-life and modifying its intracellular signaling bias or engineered AM-analogues enabling a more specific and sustained activation of AM receptors may provide us with possible tools to tackle the two main limitations of AM in the clinic, namely its short half-life and the wide variety of biological functions. These novel advances further boost the clinical potential of AM signaling pathways. Further studies are required to fully understand the mechanisms that determine which intracellular signaling pathway is activated upon AM binding by its receptor complex. Understanding these mechanisms may further improve the application of AM as a predictive biomarker and development of tissue specific AM-targeted therapeutic approaches.
Acknowledgements:
The authors thank members of the Caron Lab for helpful discussions and critical reading of the manuscript. We gratefully and humbly acknowledge all members of the scientific community who have made impactful and significant contributions to the study of AM/CLR/RAMP signaling and regret that we were unable to discuss or cite all studies in this review. This work was supported by the following grants: NIH/NHLBI HL1290986 and NIH/NIDDK DK119145 to KMC, an American Heart Association Postdoctoral Fellowship 23POST1022945 to LB, an American Heart Association Predoctoral Fellowship 909016 to NPNM, and an NIH/NHLBI F31 Fellowship F31HL163885 to DSS.
Non-standard Abbreviations and Acronyms
- ACKR3
atypical chemokine receptor 3
- AM
adrenomedullin (protein; gene: Adm)
- ANP
atrial natriuretic peptide
- BEC
blood endothelial cell
- BNP
B-type natriuretic peptide
- cAMP
cyclic adenosine monophosphate
- CAP
community-acquired pneumonia
- CGRP
calcitonin gene-related peptide (protein; gene: Calca/Calcb)
- CLR
calcitonin-receptor like receptor (protein; gene: Calcrl)
- CREB
cAMP response element-binding protein
- CSF
cerebrospinal fluid
- CXCL
chemokine x ligand
- EC
endothelial cell
- eNOS
endothelial nitric oxide synthase
- GPCR
G protein-coupled receptor
- HF
heart failure
- HFrEF.
heart failure with reduced ejection fraction
- HUAEC
human umbilical arterial endothelial cells
- HUVEC
human umbilical vein endothelial cells
- IBD
inflammatory bowel disease
- JAM-A
junctional adhesion molecule-A
- LEC
lymphatic endothelial cells
- LPS
lipopolysaccharide
- LYVE1
lymphatic vessel endothelial hyaluronan receptor 1
- MAPK
mitogen-activated protein kinase
- MI
myocardial infarction
- MR pro-ADM
pro-adrenomedullin 45-92
- NO
nitric oxide
- PAMP
proAM N-terminal 20 peptide
- Pro-AM
pro-adrenomedullin
- PROX1
prospero homeobox protein 1
- PSI
pneumonia severity index
- RAMP
receptor activity-modifying protein
- SPECT
single-photon emission computerized tomography
- ss-AM
sustained signaling AM
- TNBS
2,4,6-trinitrobenzenesulfonic acid
- VAP
ventilator associated pneumonia
- VE-Cadherin
vascular endothelial cadherin
- VEGFR
vascularar endothelial growth factor receptor
- ZO-1
zona occludens 1
Footnotes
Disclosures: None.
References
- 1.Kitamura K, Kangawa K, Kawamoto M, Ichiki Y, Nakamura S, Matsuo H, Eto T. Adrenomedullin: a novel hypotensive peptide isolated from human pheochromocytoma. Biochem Biophys Res Commun. 1993;192:553–560. doi: 10.1006/bbrc.1993.1451 [DOI] [PubMed] [Google Scholar]
- 2.Isumi Y, Shoji H, Sugo S, Tochimoto T, Yoshioka M, Kangawa K, Matsuo H, Minamino N. Regulation of adrenomedullin production in rat endothelial cells. Endocrinology. 1998;139:838–846. doi: 10.1210/endo.139.3.5789 [DOI] [PubMed] [Google Scholar]
- 3.Kitamura K, Sakata J, Kangawa K, Kojima M, Matsuo H, Eto T. Cloning and characterization of cDNA encoding a precursor for human adrenomedullin. Biochem Biophys Res Commun. 1993;194:720–725. doi: 10.1006/bbrc.1993.1881 [DOI] [PubMed] [Google Scholar]
- 4.Sugo S, Minamino N, Shoji H, Kangawa K, Kitamura K, Eto T, Matsuo H. Production and secretion of adrenomedullin from vascular smooth muscle cells: augmented production by tumor necrosis factor-alpha. Biochem Biophys Res Commun. 1994;203:719–726. doi: 10.1006/bbrc.1994.2241 [DOI] [PubMed] [Google Scholar]
- 5.Shoji H, Minamino N, Kangawa K, Matsuo H. Endotoxin markedly elevates plasma concentration and gene transcription of adrenomedullin in rat. Biochem Biophys Res Commun. 1995;215:531–537. doi: 10.1006/bbrc.1995.2497 [DOI] [PubMed] [Google Scholar]
- 6.Morimoto A, Nishikimi T, Yoshihara F, Horio T, Nagaya N, Matsuo H, Dohi K, Kangawa K. Ventricular adrenomedullin levels correlate with the extent of cardiac hypertrophy in rats. Hypertension. 1999;33:1146–1152. doi: 10.1161/01.hyp.33.5.1146 [DOI] [PubMed] [Google Scholar]
- 7.Iring A, Jin YJ, Albarran-Juarez J, Siragusa M, Wang S, Dancs PT, Nakayama A, Tonack S, Chen M, Kunne C, et al. Shear stress-induced endothelial adrenomedullin signaling regulates vascular tone and blood pressure. J Clin Invest. 2019;129:2775–2791. doi: 10.1172/JCI123825 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Nguyen SV, Claycomb WC. Hypoxia regulates the expression of the adrenomedullin and HIF-1 genes in cultured HL-1 cardiomyocytes. Biochem Biophys Res Commun. 1999;265:382–386. doi: 10.1006/bbrc.1999.1674 [DOI] [PubMed] [Google Scholar]
- 9.Meeran K, O'Shea D, Upton PD, Small CJ, Ghatei MA, Byfield PH, Bloom SR. Circulating adrenomedullin does not regulate systemic blood pressure but increases plasma prolactin after intravenous infusion in humans: a pharmacokinetic study. J Clin Endocrinol Metab. 1997;82:95–100. doi: 10.1210/jcem.82.1.3656 [DOI] [PubMed] [Google Scholar]
- 10.Struck J, Tao C, Morgenthaler NG, Bergmann A. Identification of an Adrenomedullin precursor fragment in plasma of sepsis patients. Peptides. 2004;25:1369–1372. doi: 10.1016/j.peptides.2004.06.019 [DOI] [PubMed] [Google Scholar]
- 11.Geven C, van Lier D, Blet A, Peelen R, Ten Elzen B, Mebazaa A, Kox M, Pickkers P. Safety, tolerability and pharmacokinetics/pharmacodynamics of the adrenomedullin antibody adrecizumab in a first-in-human study and during experimental human endotoxaemia in healthy subjects. Br J Clin Pharmacol. 2018;84:2129–2141. doi: 10.1111/bcp.13655 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Hauser AS, Attwood MM, Rask-Andersen M, Schioth HB, Gloriam DE. Trends in GPCR drug discovery: new agents, targets and indications. Nat Rev Drug Discov. 2017;16:829–842. doi: 10.1038/nrd.2017.178 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Serafin DS, Harris NR, Nielsen NR, Mackie DI, Caron KM. Dawn of a New RAMPage. Trends Pharmacol Sci. 2020;41:249–265. doi: 10.1016/j.tips.2020.01.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Hay DL, Garelja ML, Poyner DR, Walker CS. Update on the pharmacology of calcitonin/CGRP family of peptides: IUPHAR Review 25. Br J Pharmacol. 2018;175:3–17. doi: 10.1111/bph.14075 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Clark AJ, Mullooly N, Safitri D, Harris M, de Vries T, MaassenVanDenBrink A, Poyner DR, Gianni D, Wigglesworth M, Ladds G. CGRP, adrenomedullin and adrenomedullin 2 display endogenous GPCR agonist bias in primary human cardiovascular cells. Commun Biol. 2021;4:776. doi: 10.1038/s42003-021-02293-w [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Weston C, Winfield I, Harris M, Hodgson R, Shah A, Dowell SJ, Mobarec JC, Woodlock DA, Reynolds CA, Poyner DR, et al. Receptor Activity-modifying Protein-directed G Protein Signaling Specificity for the Calcitonin Gene-related Peptide Family of Receptors. J Biol Chem. 2016;291:21925–21944. doi: 10.1074/jbc.M116.751362 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Pearce A, Redfern-Nichols T, Harris M, Poyner DR, Wigglesworth M, Ladds G. Determining the Effects of Differential Expression of GRKs and beta-arrestins on CLR-RAMP Agonist Bias. Front Physiol. 2022;13:840763. doi: 10.3389/fphys.2022.840763 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Yarwood RE, Imlach WL, Lieu T, Veldhuis NA, Jensen DD, Klein Herenbrink C, Aurelio L, Cai Z, Christie MJ, Poole DP, et al. Endosomal signaling of the receptor for calcitonin gene-related peptide mediates pain transmission. Proc Natl Acad Sci U S A. 2017;114:12309–12314. doi: 10.1073/pnas.1706656114 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Garelja ML, Au M, Brimble MA, Gingell JJ, Hendrikse ER, Lovell A, Prodan N, Sexton PM, Siow A, Walker CS, et al. Molecular Mechanisms of Class B GPCR Activation: Insights from Adrenomedullin Receptors. ACS Pharmacol Transl Sci. 2020;3:246–262. doi: 10.1021/acsptsci.9b00083 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Roehrkasse AM, Warner ML, Booe JM, Pioszak AA. Biochemical characterization of G protein coupling to calcitonin gene-related peptide and adrenomedullin receptors using a native PAGE assay. J Biol Chem. 2020;295:9736–9751. doi: 10.1074/jbc.RA120.013854 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Guidolin D, Albertin G, Spinazzi R, Sorato E, Mascarin A, Cavallo D, Antonello M, Ribatti D. Adrenomedullin stimulates angiogenic response in cultured human vascular endothelial cells: involvement of the vascular endothelial growth factor receptor 2. Peptides. 2008;29:2013–2023. doi: 10.1016/j.peptides.2008.07.009 [DOI] [PubMed] [Google Scholar]
- 22.Harris NR, Nielsen NR, Pawlak JB, Aghajanian A, Rangarajan K, Serafin DS, Farber G, Dy DM, Nelson-Maney NP, Xu W, et al. VE-Cadherin Is Required for Cardiac Lymphatic Maintenance and Signaling. Circ Res. 2022;130:5–23. doi: 10.1161/CIRCRESAHA.121.318852 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Karlsson M, Zhang C, Mear L, Zhong W, Digre A, Katona B, Sjostedt E, Butler L, Odeberg J, Dusart P, et al. A single-cell type transcriptomics map of human tissues. Sci Adv. 2021;7. doi: 10.1126/sciadv.abh2169 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Burns JM, Summers BC, Wang Y, Melikian A, Berahovich R, Miao Z, Penfold ME, Sunshine MJ, Littman DR, Kuo CJ, et al. A novel chemokine receptor for SDF-1 and I-TAC involved in cell survival, cell adhesion, and tumor development. J Exp Med. 2006;203:2201–2213. doi: 10.1084/jem.20052144 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Alampour-Rajabi S, El Bounkari O, Rot A, Muller-Newen G, Bachelerie F, Gawaz M, Weber C, Schober A, Bernhagen J. MIF interacts with CXCR7 to promote receptor internalization, ERK1/2 and ZAP-70 signaling, and lymphocyte chemotaxis. FASEB J. 2015;29:4497–4511. doi: 10.1096/fj.15-273904 [DOI] [PubMed] [Google Scholar]
- 26.Szpakowska M, Dupuis N, Baragli A, Counson M, Hanson J, Piette J, Chevigne A. Human herpesvirus 8-encoded chemokine vCCL2/vMIP-II is an agonist of the atypical chemokine receptor ACKR3/CXCR7. Biochem Pharmacol. 2016;114:14–21. doi: 10.1016/j.bcp.2016.05.012 [DOI] [PubMed] [Google Scholar]
- 27.Meyrath M, Szpakowska M, Zeiner J, Massotte L, Merz MP, Benkel T, Simon K, Ohnmacht J, Turner JD, Kruger R, et al. The atypical chemokine receptor ACKR3/CXCR7 is a broad-spectrum scavenger for opioid peptides. Nat Commun. 2020;11:3033. doi: 10.1038/s41467-020-16664-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Ikeda Y, Kumagai H, Skach A, Sato M, Yanagisawa M. Modulation of circadian glucocorticoid oscillation via adrenal opioid-CXCR7 signaling alters emotional behavior. Cell. 2013;155:1323–1336. doi: 10.1016/j.cell.2013.10.052 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Meyrath M, Palmer CB, Reynders N, Vanderplasschen A, Ollert M, Bouvier M, Szpakowska M, Chevigne A. Proadrenomedullin N-Terminal 20 Peptides (PAMPs) Are Agonists of the Chemokine Scavenger Receptor ACKR3/CXCR7. ACS Pharmacol Transl Sci. 2021;4:813–823. doi: 10.1021/acsptsci.1c00006 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Kapas S, Clark AJ. Identification of an orphan receptor gene as a type 1 calcitonin gene-related peptide receptor. Biochem Biophys Res Commun. 1995;217:832–838. doi: 10.1006/bbrc.1995.2847 [DOI] [PubMed] [Google Scholar]
- 31.Boldajipour B, Mahabaleshwar H, Kardash E, Reichman-Fried M, Blaser H, Minina S, Wilson D, Xu Q, Raz E. Control of chemokine-guided cell migration by ligand sequestration. Cell. 2008;132:463–473. doi: 10.1016/j.cell.2007.12.034 [DOI] [PubMed] [Google Scholar]
- 32.Rajagopal S, Kim J, Ahn S, Craig S, Lam CM, Gerard NP, Gerard C, Lefkowitz RJ. Beta-arrestin- but not G protein-mediated signaling by the "decoy" receptor CXCR7. Proc Natl Acad Sci U S A. 2010;107:628–632. doi: 10.1073/pnas.0912852107 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Naumann U, Cameroni E, Pruenster M, Mahabaleshwar H, Raz E, Zerwes HG, Rot A, Thelen M. CXCR7 functions as a scavenger for CXCL12 and CXCL11. PLoS One. 2010;5:e9175. doi: 10.1371/journal.pone.0009175 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Zarca A, Perez C, van den Bor J, Bebelman JP, Heuninck J, de Jonker RJF, Durroux T, Vischer HF, Siderius M, Smit MJ. Differential Involvement of ACKR3 C-Tail in beta-Arrestin Recruitment, Trafficking and Internalization. Cells. 2021;10. doi: 10.3390/cells10030618 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Klein KR, Karpinich NO, Espenschied ST, Willcockson HH, Dunworth WP, Hoopes SL, Kushner EJ, Bautch VL, Caron KM. Decoy receptor CXCR7 modulates adrenomedullin-mediated cardiac and lymphatic vascular development. Dev Cell. 2014;30:528–540. doi: 10.1016/j.devcel.2014.07.012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Rosenfeld MG, Mermod JJ, Amara SG, Swanson LW, Sawchenko PE, Rivier J, Vale WW, Evans RM. Production of a novel neuropeptide encoded by the calcitonin gene via tissue-specific RNA processing. Nature. 1983;304:129–135. doi: 10.1038/304129a0 [DOI] [PubMed] [Google Scholar]
- 37.Maggi CA. Tachykinins and calcitonin gene-related peptide (CGRP) as co-transmitters released from peripheral endings of sensory nerves. Prog Neurobiol. 1995;45:1–98. doi: 10.1016/0301-0082(94)e0017-b [DOI] [PubMed] [Google Scholar]
- 38.Iyengar S, Ossipov MH, Johnson KW. The role of calcitonin gene-related peptide in peripheral and central pain mechanisms including migraine. Pain. 2017;158:543–559. doi: 10.1097/j.pain.0000000000000831 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Ashina M, Terwindt GM, Al-Karagholi MA, de Boer I, Lee MJ, Hay DL, Schulte LH, Hadjikhani N, Sinclair AJ, Ashina H, et al. Migraine: disease characterisation, biomarkers, and precision medicine. Lancet. 2021;397:1496–1504. doi: 10.1016/S0140-6736(20)32162-0 [DOI] [PubMed] [Google Scholar]
- 40.Goadsby PJ, Edvinsson L, Ekman R. Release of vasoactive peptides in the extracerebral circulation of humans and the cat during activation of the trigeminovascular system. Ann Neurol. 1988;23:193–196. doi: 10.1002/ana.410230214 [DOI] [PubMed] [Google Scholar]
- 41.Gallai V, Alberti A, Gallai B, Coppola F, Floridi A, Sarchielli P. Glutamate and nitric oxide pathway in chronic daily headache: evidence from cerebrospinal fluid. Cephalalgia. 2003;23:166–174. doi: 10.1046/j.1468-2982.2003.00552.x [DOI] [PubMed] [Google Scholar]
- 42.Fan PC, Kuo PH, Lee MT, Chang SH, Chiou LC. Plasma Calcitonin Gene-Related Peptide: A Potential Biomarker for Diagnosis and Therapeutic Responses in Pediatric Migraine. Front Neurol. 2019;10:10. doi: 10.3389/fneur.2019.00010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Hautakangas H, Winsvold BS, Ruotsalainen SE, Bjornsdottir G, Harder AVE, Kogelman LJA, Thomas LF, Noordam R, Benner C, Gormley P, et al. Genome-wide analysis of 102,084 migraine cases identifies 123 risk loci and subtype-specific risk alleles. Nat Genet. 2022;54:152–160. doi: 10.1038/s41588-021-00990-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Sherman ML, Shafman TD, Colman MS, Kufe DW. Tiazofurin induction of mouse erythroleukemia cell hemoglobin production in the absence of commitment or changes in protooncogene expression. Blood. 1989;73:431–434. [PubMed] [Google Scholar]
- 45.Robbins MS. Diagnosis and Management of Headache: A Review. JAMA. 2021;325:1874–1885. doi: 10.1001/jama.2021.1640 [DOI] [PubMed] [Google Scholar]
- 46.Shimekake Y, Nagata K, Ohta S, Kambayashi Y, Teraoka H, Kitamura K, Eto T, Kangawa K, Matsuo H. Adrenomedullin stimulates two signal transduction pathways, cAMP accumulation and Ca2+ mobilization, in bovine aortic endothelial cells. J Biol Chem. 1995;270:4412–4417. doi: 10.1074/jbc.270.9.4412 [DOI] [PubMed] [Google Scholar]
- 47.Kohno M, Kano H, Horio T, Yokokawa K, Yasunari K, Takeda T. Inhibition of endothelin production by adrenomedullin in vascular smooth muscle cells. Hypertension. 1995;25:1185–1190. doi: 10.1161/01.hyp.25.6.1185 [DOI] [PubMed] [Google Scholar]
- 48.Nagaya N, Satoh T, Nishikimi T, Uematsu M, Furuichi S, Sakamaki F, Oya H, Kyotani S, Nakanishi N, Goto Y, et al. Hemodynamic, renal, and hormonal effects of adrenomedullin infusion in patients with congestive heart failure. Circulation. 2000;101:498–503. doi: 10.1161/01.cir.101.5.498 [DOI] [PubMed] [Google Scholar]
- 49.Caron KM, Smithies O. Extreme hydrops fetalis and cardiovascular abnormalities in mice lacking a functional Adrenomedullin gene. Proc Natl Acad Sci U S A. 2001;98:615–619. doi: 10.1073/pnas.98.2.615 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Shindo T, Kurihara Y, Nishimatsu H, Moriyama N, Kakoki M, Wang Y, Imai Y, Ebihara A, Kuwaki T, Ju KH, et al. Vascular abnormalities and elevated blood pressure in mice lacking adrenomedullin gene. Circulation. 2001;104:1964–1971. doi: 10.1161/hc4101.097111 [DOI] [PubMed] [Google Scholar]
- 51.Dackor RT, Fritz-Six K, Dunworth WP, Gibbons CL, Smithies O, Caron KM. Hydrops fetalis, cardiovascular defects, and embryonic lethality in mice lacking the calcitonin receptor-like receptor gene. Mol Cell Biol. 2006;26:2511–2518. doi: 10.1128/MCB.26.7.2511-2518.2006 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Fritz-Six KL, Dunworth WP, Li M, Caron KM. Adrenomedullin signaling is necessary for murine lymphatic vascular development. J Clin Invest. 2008;118:40–50. doi: 10.1172/JCI33302 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Ichikawa-Shindo Y, Sakurai T, Kamiyoshi A, Kawate H, Iinuma N, Yoshizawa T, Koyama T, Fukuchi J, Iimuro S, Moriyama N, et al. The GPCR modulator protein RAMP2 is essential for angiogenesis and vascular integrity. J Clin Invest. 2008;118:29–39. doi: 10.1172/JCI33022 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Wetzel-Strong SE, Li M, Klein KR, Nishikimi T, Caron KM. Epicardial-derived adrenomedullin drives cardiac hyperplasia during embryogenesis. Dev Dyn. 2014;243:243–256. doi: 10.1002/dvdy.24065 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Yu S, Crawford D, Tsuchihashi T, Behrens TW, Srivastava D. The chemokine receptor CXCR7 functions to regulate cardiac valve remodeling. Dev Dyn. 2011;240:384–393. doi: 10.1002/dvdy.22549 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Gerrits H, van Ingen Schenau DS, Bakker NE, van Disseldorp AJ, Strik A, Hermens LS, Koenen TB, Krajnc-Franken MA, Gossen JA. Early postnatal lethality and cardiovascular defects in CXCR7-deficient mice. Genesis. 2008;46:235–245. doi: 10.1002/dvg.20387 [DOI] [PubMed] [Google Scholar]
- 57.Oliver G, Kipnis J, Randolph GJ, Harvey NL. The Lymphatic Vasculature in the 21(st) Century: Novel Functional Roles in Homeostasis and Disease. Cell. 2020;182:270–296. doi: 10.1016/j.cell.2020.06.039 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Wigle JT, Harvey N, Detmar M, Lagutina I, Grosveld G, Gunn MD, Jackson DG, Oliver G. An essential role for Prox1 in the induction of the lymphatic endothelial cell phenotype. EMBO J. 2002;21:1505–1513. doi: 10.1093/emboj/21.7.1505 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Gonzalez-Loyola A, Petrova TV. Development and aging of the lymphatic vascular system. Adv Drug Deliv Rev. 2021;169:63–78. doi: 10.1016/j.addr.2020.12.005 [DOI] [PubMed] [Google Scholar]
- 60.Mackie DI, Al Mutairi F, Davis RB, Kechele DO, Nielsen NR, Snyder JC, Caron MG, Kliman HJ, Berg JS, Simms J, et al. hCALCRL mutation causes autosomal recessive nonimmune hydrops fetalis with lymphatic dysplasia. J Exp Med. 2018;215:2339–2353. doi: 10.1084/jem.20180528 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Dunworth WP, Fritz-Six KL, Caron KM. Adrenomedullin stabilizes the lymphatic endothelial barrier in vitro and in vivo. Peptides. 2008;29:2243–2249. doi: 10.1016/j.peptides.2008.09.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Hoopes SL, Willcockson HH, Caron KM. Characteristics of multi-organ lymphangiectasia resulting from temporal deletion of calcitonin receptor-like receptor in adult mice. PLoS One. 2012;7:e45261. doi: 10.1371/journal.pone.0045261 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Davis RB, Kechele DO, Blakeney ES, Pawlak JB, Caron KM. Lymphatic deletion of calcitonin receptor-like receptor exacerbates intestinal inflammation. JCI Insight. 2017;2:e92465. doi: 10.1172/jci.insight.92465 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Davis RB, Ding S, Nielsen NR, Pawlak JB, Blakeney ES, Caron KM. Calcitonin-Receptor-Like Receptor Signaling Governs Intestinal Lymphatic Innervation and Lipid Uptake. ACS Pharmacol Transl Sci. 2019;2:114–121. doi: 10.1021/acsptsci.8b00061 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Nicholls MG. Hemodynamic and hormonal actions of adrenomedullin. Braz J Med Biol Res. 2004;37:1247–1253. doi: 10.1590/s0100-879x2004000800016 [DOI] [PubMed] [Google Scholar]
- 66.Koyama T, Ochoa-Callejero L, Sakurai T, Kamiyoshi A, Ichikawa-Shindo Y, Iinuma N, Arai T, Yoshizawa T, Iesato Y, Lei Y, et al. Vascular endothelial adrenomedullin-RAMP2 system is essential for vascular integrity and organ homeostasis. Circulation. 2013;127:842–853. doi: 10.1161/CIRCULATIONAHA.112.000756 [DOI] [PubMed] [Google Scholar]
- 67.Martinez A. A new family of angiogenic factors. Cancer Lett. 2006;236:157–163. doi: 10.1016/j.canlet.2005.04.008 [DOI] [PubMed] [Google Scholar]
- 68.Mueller C, McDonald K, de Boer RA, Maisel A, Cleland JGF, Kozhuharov N, Coats AJS, Metra M, Mebazaa A, Ruschitzka F, et al. Heart Failure Association of the European Society of Cardiology practical guidance on the use of natriuretic peptide concentrations. Eur J Heart Fail. 2019;21:715–731. doi: 10.1002/ejhf.1494 [DOI] [PubMed] [Google Scholar]
- 69.Jougasaki M, Wei CM, McKinley LJ, Burnett JC Jr. Elevation of circulating and ventricular adrenomedullin in human congestive heart failure. Circulation. 1995;92:286–289. doi: 10.1161/01.cir.92.3.286 [DOI] [PubMed] [Google Scholar]
- 70.Nishikimi T, Saito Y, Kitamura K, Ishimitsu T, Eto T, Kangawa K, Matsuo H, Omae T, Matsuoka H. Increased plasma levels of adrenomedullin in patients with heart failure. J Am Coll Cardiol. 1995;26:1424–1431. doi: 10.1016/0735-1097(95)00338-X [DOI] [PubMed] [Google Scholar]
- 71.Maisel A, Mueller C, Nowak R, Peacock WF, Landsberg JW, Ponikowski P, Mockel M, Hogan C, Wu AH, Richards M, et al. Mid-region pro-hormone markers for diagnosis and prognosis in acute dyspnea: results from the BACH (Biomarkers in Acute Heart Failure) trial. J Am Coll Cardiol. 2010;55:2062–2076. doi: 10.1016/j.jacc.2010.02.025 [DOI] [PubMed] [Google Scholar]
- 72.Self WH, Storrow AB, Hartmann O, Barrett TW, Fermann GJ, Maisel AS, Struck J, Bergmann A, Collins SP. Plasma bioactive adrenomedullin as a prognostic biomarker in acute heart failure. Am J Emerg Med. 2016;34:257–262. doi: 10.1016/j.ajem.2015.10.033 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Tolppanen H, Rivas-Lasarte M, Lassus J, Sans-Rosello J, Hartmann O, Lindholm M, Arrigo M, Tarvasmaki T, Kober L, Thiele H, et al. Adrenomedullin: a marker of impaired hemodynamics, organ dysfunction, and poor prognosis in cardiogenic shock. Ann Intensive Care. 2017;7:6. doi: 10.1186/s13613-016-0229-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Ter Maaten JM, Kremer D, Demissei BG, Struck J, Bergmann A, Anker SD, Ng LL, Dickstein K, Metra M, Samani NJ, et al. Bio-adrenomedullin as a marker of congestion in patients with new-onset and worsening heart failure. Eur J Heart Fail. 2019;21:732–743. doi: 10.1002/ejhf.1437 [DOI] [PubMed] [Google Scholar]
- 75.Morbach C, Marx A, Kaspar M, Guder G, Brenner S, Feldmann C, Stork S, Vollert JO, Ertl G, Angermann CE, et al. Prognostic potential of midregional pro-adrenomedullin following decompensation for systolic heart failure: comparison with cardiac natriuretic peptides. Eur J Heart Fail. 2017;19:1166–1175. doi: 10.1002/ejhf.859 [DOI] [PubMed] [Google Scholar]
- 76.Kuan WS, Ibrahim I, Chan SP, Li Z, Liew OW, Frampton C, Troughton R, Pemberton CJ, Chong JPC, Tan LL, et al. Mid-regional pro-adrenomedullin outperforms N-terminal pro-B-type natriuretic peptide for the diagnosis of acute heart failure in the presence of atrial fibrillation. Eur J Heart Fail. 2020;22:692–700. doi: 10.1002/ejhf.1660 [DOI] [PubMed] [Google Scholar]
- 77.Feng Z, Akinrimisi OP, Gornbein JA, Truong QA, Das S, Singh JP, Ajijola O. Combination Biomarkers for Risk Stratification in Patients With Chronic Heart Failure Biomarkers Prognostication in HF. J Card Fail. 2021;27:1321–1327. doi: 10.1016/j.cardfail.2021.05.028 [DOI] [PubMed] [Google Scholar]
- 78.Dungen HD, Tscholl V, Obradovic D, Radenovic S, Matic D, Musial Bright L, Tahirovic E, Marx A, Inkrot S, Hashemi D, et al. Prognostic performance of serial in-hospital measurements of copeptin and multiple novel biomarkers among patients with worsening heart failure: results from the MOLITOR study. ESC Heart Fail. 2018;5:288–296. doi: 10.1002/ehf2.12231 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Fraty M, Velho G, Gand E, Fumeron F, Ragot S, Sosner P, Mohammedi K, Gellen B, Saulnier PJ, Halimi JM, et al. Prognostic value of plasma MR-proADM vs NT-proBNP for heart failure in people with type 2 diabetes: the SURDIAGENE prospective study. Diabetologia. 2018;61:2643–2653. doi: 10.1007/s00125-018-4727-7 [DOI] [PubMed] [Google Scholar]
- 80.Welsh P, Kou L, Yu C, Anand I, van Veldhuisen DJ, Maggioni AP, Desai AS, Solomon SD, Pfeffer MA, Cheng S, et al. Prognostic importance of emerging cardiac, inflammatory, and renal biomarkers in chronic heart failure patients with reduced ejection fraction and anaemia: RED-HF study. Eur J Heart Fail. 2018;20:268–277. doi: 10.1002/ejhf.988 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Szokodi I, Kinnunen P, Tavi P, Weckstrom M, Toth M, Ruskoaho H. Evidence for cAMP-independent mechanisms mediating the effects of adrenomedullin, a new inotropic peptide. Circulation. 1998;97:1062–1070. doi: 10.1161/01.cir.97.11.1062 [DOI] [PubMed] [Google Scholar]
- 82.Wang P, Ba ZF, Cioffi WG, Bland KI, Chaudry IH. The pivotal role of adrenomedullin in producing hyperdynamic circulation during the early stage of sepsis. Arch Surg. 1998;133:1298–1304. doi: 10.1001/archsurg.133.12.1298 [DOI] [PubMed] [Google Scholar]
- 83.Tsao CW, Aday AW, Almarzooq ZI, Alonso A, Beaton AZ, Bittencourt MS, Boehme AK, Buxton AE, Carson AP, Commodore-Mensah Y, et al. Heart Disease and Stroke Statistics-2022 Update: A Report From the American Heart Association. Circulation. 2022;145:e153–e639. doi: 10.1161/CIR.0000000000001052 [DOI] [PubMed] [Google Scholar]
- 84.Kobayashi K, Kitamura K, Hirayama N, Date H, Kashiwagi T, Ikushima I, Hanada Y, Nagatomo Y, Takenaga M, Ishikawa T, et al. Increased plasma adrenomedullin in acute myocardial infarction. Am Heart J. 1996;131:676–680. doi: 10.1016/s0002-8703(96)90270-7 [DOI] [PubMed] [Google Scholar]
- 85.Falkentoft AC, Rorth R, Iversen K, Hofsten DE, Kelbaek H, Holmvang L, Frydland M, Schoos MM, Helqvist S, Axelsson A, et al. MR-proADM as a Prognostic Marker in Patients With ST-Segment-Elevation Myocardial Infarction-DANAMI-3 (a Danish Study of Optimal Acute Treatment of Patients With STEMI) Substudy. J Am Heart Assoc. 2018;7. doi: 10.1161/JAHA.117.008123 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Horiuchi Y, Wettersten N, Patel MP, Mueller C, Neath SX, Christenson RH, Morgenthaler NG, McCord J, Nowak RM, Vilke GM, et al. Biomarkers Enhance Discrimination and Prognosis of Type 2 Myocardial Infarction. Circulation. 2020;142:1532–1544. doi: 10.1161/CIRCULATIONAHA.120.046682 [DOI] [PubMed] [Google Scholar]
- 87.Li L, Zhang S, Zhang Y, Yu B, Xu Y, Guan Z. Paracrine action mediate the antifibrotic effect of transplanted mesenchymal stem cells in a rat model of global heart failure. Mol Biol Rep. 2009;36:725–731. doi: 10.1007/s11033-008-9235-2 [DOI] [PubMed] [Google Scholar]
- 88.Horio T, Nishikimi T, Yoshihara F, Matsuo H, Takishita S, Kangawa K. Effects of adrenomedullin on cultured rat cardiac myocytes and fibroblasts. Eur J Pharmacol. 1999;382:1–9. doi: 10.1016/s0014-2999(99)00559-2 [DOI] [PubMed] [Google Scholar]
- 89.Nakamura R, Kato J, Kitamura K, Onitsuka H, Imamura T, Marutsuka K, Asada Y, Kangawa K, Eto T. Beneficial effects of adrenomedullin on left ventricular remodeling after myocardial infarction in rats. Cardiovasc Res. 2002;56:373–380. doi: 10.1016/s0008-6363(02)00594-1 [DOI] [PubMed] [Google Scholar]
- 90.Okumura H, Nagaya N, Kangawa K. Adrenomedullin infusion during ischemia/reperfusion attenuates left ventricular remodeling and myocardial fibrosis in rats. Hypertens Res. 2003;26 Suppl:S99–104. doi: 10.1291/hypres.26.s99 [DOI] [PubMed] [Google Scholar]
- 91.Nishikimi T, Yoshihara F, Horinaka S, Kobayashi N, Mori Y, Tadokoro K, Akimoto K, Minamino N, Kangawa K, Matsuoka H. Chronic administration of adrenomedullin attenuates transition from left ventricular hypertrophy to heart failure in rats. Hypertension. 2003;42:1034–1041. doi: 10.1161/01.HYP.0000097604.64716.D2 [DOI] [PubMed] [Google Scholar]
- 92.Yoshizawa T, Takizawa S, Shimada S, Tokudome T, Shindo T, Matsumoto K. Effects of Adrenomedullin on Doxorubicin-Induced Cardiac Damage in Mice. Biol Pharm Bull. 2016;39:737–746. doi: 10.1248/bpb.b15-00832 [DOI] [PubMed] [Google Scholar]
- 93.Kataoka Y, Miyazaki S, Yasuda S, Nagaya N, Noguchi T, Yamada N, Morii I, Kawamura A, Doi K, Miyatake K, et al. The first clinical pilot study of intravenous adrenomedullin administration in patients with acute myocardial infarction. J Cardiovasc Pharmacol. 2010;56:413–419. doi: 10.1097/FJC.0b013e3181f15b45 [DOI] [PubMed] [Google Scholar]
- 94.Nishikimi T, Karasawa T, Inaba C, Ishimura K, Tadokoro K, Koshikawa S, Yoshihara F, Nagaya N, Sakio H, Kangawa K, et al. Effects of long-term intravenous administration of adrenomedullin (AM) plus hANP therapy in acute decompensated heart failure: a pilot study. Circ J. 2009;73:892–898. doi: 10.1253/circj.cj-08-0487 [DOI] [PubMed] [Google Scholar]
- 95.Trincot CE, Xu W, Zhang H, Kulikauskas MR, Caranasos TG, Jensen BC, Sabine A, Petrova TV, Caron KM. Adrenomedullin Induces Cardiac Lymphangiogenesis After Myocardial Infarction and Regulates Cardiac Edema Via Connexin 43. Circ Res. 2019;124:101–113. doi: 10.1161/CIRCRESAHA.118.313835 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Liu X, De la Cruz E, Gu X, Balint L, Oxendine-Burns M, Terrones T, Ma W, Kuo HH, Lantz C, Bansal T, et al. Lymphoangiocrine signals promote cardiac growth and repair. Nature. 2020;588:705–711. doi: 10.1038/s41586-020-2998-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Harris NR, Balint L, Dy DM, Nielsen NR, Mendez HG, Aghajanian A, Caron KM. The ebb and flow of cardiac lymphatics: a tidal wave of new discoveries. Physiol Rev. 2023;103:391–432. doi: 10.1152/physrev.00052.2021 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Kolditz M, Seyfarth HJ, Wilkens H, Ewert R, Bollmann T, Dinter C, Hertel S, Klose H, Opitz C, Grunig E, et al. MR-proADM Predicts Exercise Capacity and Survival Superior to Other Biomarkers in PH. Lung. 2015;193:901–910. doi: 10.1007/s00408-015-9802-y [DOI] [PubMed] [Google Scholar]
- 99.Bouzina H, Radegran G. Plasma adrenomedullin peptides and precursor levels in pulmonary arterial hypertension disease severity and risk stratification. Pulm Circ. 2020;10:2045894020931317. doi: 10.1177/2045894020931317 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Nagaya N, Okumura H, Uematsu M, Shimizu W, Ono F, Shirai M, Mori H, Miyatake K, Kangawa K. Repeated inhalation of adrenomedullin ameliorates pulmonary hypertension and survival in monocrotaline rats. Am J Physiol Heart Circ Physiol. 2003;285:H2125–2131. doi: 10.1152/ajpheart.00548.2002 [DOI] [PubMed] [Google Scholar]
- 101.Nagaya N, Miyatake K, Kyotani S, Nishikimi T, Nakanishi N, Kangawa K. Pulmonary vasodilator response to adrenomedullin in patients with pulmonary hypertension. Hypertens Res. 2003;26 Suppl:S141–146. doi: 10.1291/hypres.26.s141 [DOI] [PubMed] [Google Scholar]
- 102.Nagaya N, Kyotani S, Uematsu M, Ueno K, Oya H, Nakanishi N, Shirai M, Mori H, Miyatake K, Kangawa K. Effects of adrenomedullin inhalation on hemodynamics and exercise capacity in patients with idiopathic pulmonary arterial hypertension. Circulation. 2004;109:351–356. doi: 10.1161/01.CIR.0000109493.05849.14 [DOI] [PubMed] [Google Scholar]
- 103.Melenovsky V, Andersen MJ, Andress K, Reddy YN, Borlaug BA. Lung congestion in chronic heart failure: haemodynamic, clinical, and prognostic implications. Eur J Heart Fail. 2015;17:1161–1171. doi: 10.1002/ejhf.417 [DOI] [PubMed] [Google Scholar]
- 104.Ambrosy AP, Pang PS, Khan S, Konstam MA, Fonarow GC, Traver B, Maggioni AP, Cook T, Swedberg K, Burnett JC Jr., et al. Clinical course and predictive value of congestion during hospitalization in patients admitted for worsening signs and symptoms of heart failure with reduced ejection fraction: findings from the EVEREST trial. Eur Heart J. 2013;34:835–843. doi: 10.1093/eurheartj/ehs444 [DOI] [PubMed] [Google Scholar]
- 105.Van Aelst LNL, Arrigo M, Placido R, Akiyama E, Girerd N, Zannad F, Manivet P, Rossignol P, Badoz M, Sadoune M, et al. Acutely decompensated heart failure with preserved and reduced ejection fraction present with comparable haemodynamic congestion. Eur J Heart Fail. 2018;20:738–747. doi: 10.1002/ejhf.1050 [DOI] [PubMed] [Google Scholar]
- 106.Gheorghiade M, Follath F, Ponikowski P, Barsuk JH, Blair JE, Cleland JG, Dickstein K, Drazner MH, Fonarow GC, Jaarsma T, et al. Assessing and grading congestion in acute heart failure: a scientific statement from the acute heart failure committee of the heart failure association of the European Society of Cardiology and endorsed by the European Society of Intensive Care Medicine. Eur J Heart Fail. 2010;12:423–433. doi: 10.1093/eurjhf/hfq045 [DOI] [PubMed] [Google Scholar]
- 107.Voors AA, Ter Maaten JM. Tackling Early Heart Failure Deaths and Readmissions by Estimating Congestion. JACC Heart Fail. 2015;3:894–895. doi: 10.1016/j.jchf.2015.06.013 [DOI] [PubMed] [Google Scholar]
- 108.Hagner S, Stahl U, Knoblauch B, McGregor GP, Lang RE. Calcitonin receptor-like receptor: identification and distribution in human peripheral tissues. Cell Tissue Res. 2002;310:41–50. doi: 10.1007/s00441-002-0616-x [DOI] [PubMed] [Google Scholar]
- 109.Harel F, Levac X, Nguyen QT, Letourneau M, Marcil S, Finnerty V, Cossette M, Fournier A, Dupuis J. Molecular Imaging of the Human Pulmonary Vascular Endothelium Using an Adrenomedullin Receptor Ligand. Mol Imaging. 2015;14:7290201500003. doi: 10.2310/7290.2015.00003 [DOI] [PubMed] [Google Scholar]
- 110.Harel F, Langleben D, Provencher S, Fournier A, Finnerty V, Nguyen QT, Letourneau M, Levac X, Abikhzer G, Guimond J, et al. Molecular imaging of the human pulmonary vascular endothelium in pulmonary hypertension: a phase II safety and proof of principle trial. Eur J Nucl Med Mol Imaging. 2017;44:1136–1144. doi: 10.1007/s00259-017-3655-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Iimuro S, Shindo T, Moriyama N, Amaki T, Niu P, Takeda N, Iwata H, Zhang Y, Ebihara A, Nagai R. Angiogenic effects of adrenomedullin in ischemia and tumor growth. Circ Res. 2004;95:415–423. doi: 10.1161/01.RES.0000138018.61065.d1 [DOI] [PubMed] [Google Scholar]
- 112.Jin D, Harada K, Ohnishi S, Yamahara K, Kangawa K, Nagaya N. Adrenomedullin induces lymphangiogenesis and ameliorates secondary lymphoedema. Cardiovasc Res. 2008;80:339–345. doi: 10.1093/cvr/cvn228 [DOI] [PubMed] [Google Scholar]
- 113.Maki T, Ihara M, Fujita Y, Nambu T, Miyashita K, Yamada M, Washida K, Nishio K, Ito H, Harada H, et al. Angiogenic and vasoprotective effects of adrenomedullin on prevention of cognitive decline after chronic cerebral hypoperfusion in mice. Stroke. 2011;42:1122–1128. doi: 10.1161/STROKEAHA.110.603399 [DOI] [PubMed] [Google Scholar]
- 114.Hanabusa K, Nagaya N, Iwase T, Itoh T, Murakami S, Shimizu Y, Taki W, Miyatake K, Kangawa K. Adrenomedullin enhances therapeutic potency of mesenchymal stem cells after experimental stroke in rats. Stroke. 2005;36:853–858. doi: 10.1161/01.STR.0000157661.69482.76 [DOI] [PubMed] [Google Scholar]
- 115.Fujioka M, Nishio K, Sakaki T, Minamino N, Kitamura K. Adrenomedullin in patients with cerebral vasospasm after aneurysmal subarachnoid hemorrhage. Stroke. 2000;31:3079–3083. doi: 10.1161/01.str.31.12.3079-d [DOI] [PubMed] [Google Scholar]
- 116.Seifert-Held T, Pekar T, Gattringer T, Simmet NE, Scharnagl H, Bocksrucker C, Lampl C, Storch MK, Stojakovic T, Fazekas F. Plasma midregional pro-adrenomedullin improves prediction of functional outcome in ischemic stroke. PLoS One. 2013;8:e68768. doi: 10.1371/journal.pone.0068768 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Zhang H, Tang B, Yin CG, Chen Y, Meng QL, Jiang L, Wang WP, Niu GZ. Plasma adrenomedullin levels are associated with long-term outcomes of acute ischemic stroke. Peptides. 2014;52:44–48. doi: 10.1016/j.peptides.2013.11.025 [DOI] [PubMed] [Google Scholar]
- 118.Julian-Villaverde FJ, Ochoa-Callejero L, Siles E, Martinez-Lara E, Martinez A. Adrenomedullin Is a Diagnostic and Prognostic Biomarker for Acute Intracerebral Hemorrhage. Curr Issues Mol Biol. 2021;43:324–334. doi: 10.3390/cimb43010027 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Xia CF, Yin H, Borlongan CV, Chao J, Chao L. Postischemic infusion of adrenomedullin protects against ischemic stroke by inhibiting apoptosis and promoting angiogenesis. Exp Neurol. 2006;197:521–530. doi: 10.1016/j.expneurol.2005.10.027 [DOI] [PubMed] [Google Scholar]
- 120.Chaung WW, Wu R, Ji Y, Wang Z, Dong W, Cheyuo C, Qi L, Qiang X, Wang H, Wang P. Peripheral administration of human adrenomedullin and its binding protein attenuates stroke-induced apoptosis and brain injury in rats. Mol Med. 2011;17:1075–1083. doi: 10.2119/molmed.2010.00104 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Maki T, Takahashi Y, Miyamoto N, Liang AC, Ihara M, Lo EH, Arai K. Adrenomedullin promotes differentiation of oligodendrocyte precursor cells into myelin-basic-protein expressing oligodendrocytes under pathological conditions in vitro. Stem Cell Res. 2015;15:68–74. doi: 10.1016/j.scr.2015.05.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Yoshimoto T, Saito S, Omae K, Hattori Y, Fukuma K, Kitamura K, Kakuta R, Kita T, Maruyama H, Yamamoto H, et al. Study Protocol for a Randomized, Double-Blind, Placebo-Controlled, Phase-II Trial: AdrenoMedullin for Ischemic Stroke Study. J Stroke Cerebrovasc Dis. 2021;30:105761. doi: 10.1016/j.jstrokecerebrovasdis.2021.105761 [DOI] [PubMed] [Google Scholar]
- 123.Ueda S, Nishio K, Minamino N, Kubo A, Akai Y, Kangawa K, Matsuo H, Fujimura Y, Yoshioka A, Masui K, et al. Increased plasma levels of adrenomedullin in patients with systemic inflammatory response syndrome. Am J Respir Crit Care Med. 1999;160:132–136. doi: 10.1164/ajrccm.160.1.9810006 [DOI] [PubMed] [Google Scholar]
- 124.Guignant C, Voirin N, Venet F, Poitevin F, Malcus C, Bohe J, Lepape A, Monneret G. Assessment of pro-vasopressin and pro-adrenomedullin as predictors of 28-day mortality in septic shock patients. Intensive Care Med. 2009;35:1859–1867. doi: 10.1007/s00134-009-1610-5 [DOI] [PubMed] [Google Scholar]
- 125.Marino R, Struck J, Maisel AS, Magrini L, Bergmann A, Di Somma S. Plasma adrenomedullin is associated with short-term mortality and vasopressor requirement in patients admitted with sepsis. Crit Care. 2014;18:R34. doi: 10.1186/cc13731 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Lundberg OHM, Lengquist M, Spangfors M, Annborn M, Bergmann D, Schulte J, Levin H, Melander O, Frigyesi A, Friberg H. Circulating bioactive adrenomedullin as a marker of sepsis, septic shock and critical illness. Crit Care. 2020;24:636. doi: 10.1186/s13054-020-03351-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Temmesfeld-Wollbruck B, Brell B, David I, Dorenberg M, Adolphs J, Schmeck B, Suttorp N, Hippenstiel S. Adrenomedullin reduces vascular hyperpermeability and improves survival in rat septic shock. Intensive Care Med. 2007;33:703–710. doi: 10.1007/s00134-007-0561-y [DOI] [PubMed] [Google Scholar]
- 128.Geven C, Bergmann A, Kox M, Pickkers P. Vascular Effects of Adrenomedullin and the Anti-Adrenomedullin Antibody Adrecizumab in Sepsis. Shock. 2018;50:132–140. doi: 10.1097/SHK.0000000000001103 [DOI] [PubMed] [Google Scholar]
- 129.Laterre PF, Pickkers P, Marx G, Wittebole X, Meziani F, Dugernier T, Huberlant V, Schuerholz T, Francois B, Lascarrou JB, et al. Safety and tolerability of non-neutralizing adrenomedullin antibody adrecizumab (HAM8101) in septic shock patients: the AdrenOSS-2 phase 2a biomarker-guided trial. Intensive Care Med. 2021;47:1284–1294. doi: 10.1007/s00134-021-06537-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Tsuruda T, Jougasaki M, Boerrigter G, Costello-Boerrigter LC, Cataliotti A, Lee SC, Salz-Gilman L, Nordstrom LJ, McGregor CG, Burnett JC. Ventricular adrenomedullin is associated with myocyte hypertrophy in human transplanted heart. Regul Pept. 2003;112:161–166. doi: 10.1016/s0167-0115(03)00035-1 [DOI] [PubMed] [Google Scholar]
- 131.Csordas A, Nietlispach F, Schuetz P, Huber A, Muller B, Maisano F, Taramasso M, Moarof I, Obeid S, Stahli BE, et al. Midregional Proadrenomedullin Improves Risk Stratification beyond Surgical Risk Scores in Patients Undergoing Transcatheter Aortic Valve Replacement. PLoS One. 2015;10:e0143761. doi: 10.1371/journal.pone.0143761 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Tan ESJ, Oon YY, Chan SP, Liew OW, Chong JPC, Tay E, Soo WM, Yip JWL, Gong L, Lunaria JB, et al. Novel predictive role for mid-regional proadrenomedullin in moderate to severe aortic stenosis. Heart. 2022;108:1319–1327. doi: 10.1136/heartjnl-2021-320707 [DOI] [PubMed] [Google Scholar]
- 133.Witte MH, Bernas MJ, Martin CP, Witte CL. Lymphangiogenesis and lymphangiodysplasia: from molecular to clinical lymphology. Microsc Res Tech. 2001;55:122–145. doi: 10.1002/jemt.1163 [DOI] [PubMed] [Google Scholar]
- 134.Karkkainen MJ, Alitalo K. Lymphatic endothelial regulation, lymphoedema, and lymph node metastasis. Semin Cell Dev Biol. 2002;13:9–18. doi: 10.1006/scdb.2001.0286 [DOI] [PubMed] [Google Scholar]
- 135.Yamauchi A, Sakurai T, Kamiyoshi A, Ichikawa-Shindo Y, Kawate H, Igarashi K, Toriyama Y, Tanaka M, Liu T, Xian X, et al. Functional differentiation of RAMP2 and RAMP3 in their regulation of the vascular system. J Mol Cell Cardiol. 2014;77:73–85. doi: 10.1016/j.yjmcc.2014.09.017 [DOI] [PubMed] [Google Scholar]
- 136.Nikitenko LL, Shimosawa T, Henderson S, Makinen T, Shimosawa H, Qureshi U, Pedley RB, Rees MC, Fujita T, Boshoff C. Adrenomedullin haploinsufficiency predisposes to secondary lymphedema. J Invest Dermatol. 2013;133:1768–1776. doi: 10.1038/jid.2013.47 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Vazquez R, Riveiro ME, Berenguer-Daize C, O'Kane A, Gormley J, Touzelet O, Rezai K, Bekradda M, Ouafik L. Targeting Adrenomedullin in Oncology: A Feasible Strategy With Potential as Much More Than an Alternative Anti-Angiogenic Therapy. Front Oncol. 2020;10:589218. doi: 10.3389/fonc.2020.589218 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Wu XY, Hao CP, Ling M, Guo CH, Ma W. Hypoxia-induced apoptosis is blocked by adrenomedullin via upregulation of Bcl-2 in human osteosarcoma cells. Oncol Rep. 2015;34:787–794. doi: 10.3892/or.2015.4011 [DOI] [PubMed] [Google Scholar]
- 139.Benyahia Z, Dussault N, Cayol M, Sigaud R, Berenguer-Daize C, Delfino C, Tounsi A, Garcia S, Martin PM, Mabrouk K, et al. Stromal fibroblasts present in breast carcinomas promote tumor growth and angiogenesis through adrenomedullin secretion. Oncotarget. 2017;8:15744–15762. doi: 10.18632/oncotarget.14999 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Chen P, Huang Y, Bong R, Ding Y, Song N, Wang X, Song X, Luo Y. Tumor-associated macrophages promote angiogenesis and melanoma growth via adrenomedullin in a paracrine and autocrine manner. Clin Cancer Res. 2011;17:7230–7239. doi: 10.1158/1078-0432.CCR-11-1354 [DOI] [PubMed] [Google Scholar]
- 141.Zhao Q, Zhang P, Qin G, Ren F, Zheng Y, Qiao Y, Sun T, Zhang Y. Role of CXCR7 as a Common Predictor for Prognosis in Solid Tumors: a Meta-Analysis. J Cancer. 2018;9:3138–3148. doi: 10.7150/jca.25377 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Karpinich NO, Kechele DO, Espenschied ST, Willcockson HH, Fedoriw Y, Caron KM. Adrenomedullin gene dosage correlates with tumor and lymph node lymphangiogenesis. FASEB J. 2013;27:590–600. doi: 10.1096/fj.12-214080 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Karpinich NO, Caron KM. Gap junction coupling is required for tumor cell migration through lymphatic endothelium. Arterioscler Thromb Vasc Biol. 2015;35:1147–1155. doi: 10.1161/ATVBAHA.114.304752 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Dai X, Ma W, He XJ, Jha RK. Elevated expression of adrenomedullin is correlated with prognosis and disease severity in osteosarcoma. Med Oncol. 2013;30:347. doi: 10.1007/s12032-012-0347-0 [DOI] [PubMed] [Google Scholar]
- 145.Angenendt L, Bormann E, Pabst C, Alla V, Gorlich D, Braun L, Dohlich K, Schwoppe C, Bohlander SK, Arteaga MF, et al. The neuropeptide receptor calcitonin receptor-like (CALCRL) is a potential therapeutic target in acute myeloid leukemia. Leukemia. 2019;33:2830–2841. doi: 10.1038/s41375-019-0505-x [DOI] [PubMed] [Google Scholar]
- 146.Larrue C, Guiraud N, Mouchel PL, Dubois M, Farge T, Gotanegre M, Bosc C, Saland E, Nicolau-Travers ML, Sabatier M, et al. Adrenomedullin-CALCRL axis controls relapse-initiating drug tolerant acute myeloid leukemia cells. Nat Commun. 2021;12:422. doi: 10.1038/s41467-020-20717-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Angenendt L, Woste M, Mikesch JH, Arteaga MF, Angenendt A, Sandmann S, Berdel WE, Lenz G, Dugas M, Meshinchi S, et al. Calcitonin receptor-like (CALCRL) is a marker of stemness and an independent predictor of outcome in pediatric AML. Blood Adv. 2021;5:4413–4421. doi: 10.1182/bloodadvances.2021005236 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Huang Z, Zhang H, Xing C, Zhang L, Zhu H, Deng Z, Yin L, Dong E, Wang C, Peng H. Identification and validation of CALCRL-associated prognostic genes in acute myeloid leukemia. Gene. 2022;809:146009. doi: 10.1016/j.gene.2021.146009 [DOI] [PubMed] [Google Scholar]
- 149.Wang R, Li M, Bai Y, Jiao Y, Qi X. CALCRL Gene is a Suitable Prognostic Factor in AML/ETO(+) AML Patients. J Oncol. 2022;2022:3024360. doi: 10.1155/2022/3024360 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Liu LL, Chen SL, Huang YH, Yang X, Wang CH, He JH, Yun JP, Luo RZ. Adrenomedullin inhibits tumor metastasis and is associated with good prognosis in triple-negative breast cancer patients. Am J Transl Res. 2020;12:773–786. [PMC free article] [PubMed] [Google Scholar]
- 151.Christ-Crain M, Morgenthaler NG, Stolz D, Muller C, Bingisser R, Harbarth S, Tamm M, Struck J, Bergmann A, Muller B. Pro-adrenomedullin to predict severity and outcome in community-acquired pneumonia [ISRCTN04176397]. Crit Care. 2006;10:R96. doi: 10.1186/cc4955 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Espana PP, Capelastegui A, Mar C, Bilbao A, Quintana JM, Diez R, Esteban C, Bereciartua E, Unanue U, Uranga A. Performance of pro-adrenomedullin for identifying adverse outcomes in community-acquired pneumonia. J Infect. 2015;70:457–466. doi: 10.1016/j.jinf.2014.12.003 [DOI] [PubMed] [Google Scholar]
- 153.Legramante JM, Mastropasqua M, Susi B, Porzio O, Mazza M, Miranda Agrippino G, C DA, Brandi A, Giovagnoli G, Di Lecce VN, et al. Prognostic performance of MR-pro-adrenomedullin in patients with community acquired pneumonia in the Emergency Department compared to clinical severity scores PSI and CURB. PLoS One. 2017;12:e0187702. doi: 10.1371/journal.pone.0187702 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Bello S, Lasierra AB, Minchole E, Fandos S, Ruiz MA, Vera E, de Pablo F, Ferrer M, Menendez R, Torres A. Prognostic power of proadrenomedullin in community-acquired pneumonia is independent of aetiology. Eur Respir J. 2012;39:1144–1155. doi: 10.1183/09031936.00080411 [DOI] [PubMed] [Google Scholar]
- 155.Helmy TA, Tammam HH, Leuis ME, Beshey BN. Prognostic Role of Serum Adrenomedullin in Patients with Ventilator Associated Pneumonia. Adv Respir Med. 2022;90:349–359. doi: 10.3390/arm90040044 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Allaker RP, Zihni C, Kapas S. An investigation into the antimicrobial effects of adrenomedullin on members of the skin, oral, respiratory tract and gut microflora. FEMS Immunol Med Microbiol. 1999;23:289–293. doi: 10.1111/j.1574-695X.1999.tb01250.x [DOI] [PubMed] [Google Scholar]
- 157.Allaker RP, Grosvenor PW, McAnerney DC, Sheehan BE, Srikanta BH, Pell K, Kapas S. Mechanisms of adrenomedullin antimicrobial action. Peptides. 2006;27:661–666. doi: 10.1016/j.peptides.2005.09.003 [DOI] [PubMed] [Google Scholar]
- 158.Evereklioglu C, Doganay S, Er H, Yurekli M. Aqueous humor adrenomedullin levels differ in patients with different types of glaucoma. Jpn J Ophthalmol. 2002;46:203–208. doi: 10.1016/s0021-5155(01)00501-9 [DOI] [PubMed] [Google Scholar]
- 159.Ittner LM, Schwerdtfeger K, Kunz TH, Muff R, Husmann K, Grimm C, Hafezi F, Lang KS, Kurrer MO, Gotz J, et al. Transgenic mice with ocular overexpression of an adrenomedullin receptor reflect human acute angle-closure glaucoma. Clin Sci (Lond). 2008;114:49–58. doi: 10.1042/CS20070163 [DOI] [PubMed] [Google Scholar]
- 160.Blom J, Giove TJ, Pong WW, Blute TA, Eldred WD. Evidence for a functional adrenomedullin signaling pathway in the mouse retina. Mol Vis. 2012;18:1339–1353. [PMC free article] [PubMed] [Google Scholar]
- 161.Gong B, Zhang H, Huang L, Chen Y, Shi Y, Tam PO, Zhu X, Huang Y, Lei B, Sundaresan P, et al. Mutant RAMP2 causes primary open-angle glaucoma via the CRLR-cAMP axis. Genet Med. 2019;21:2345–2354. doi: 10.1038/s41436-019-0507-0 [DOI] [PubMed] [Google Scholar]
- 162.Overby DR, Zhou EH, Vargas-Pinto R, Pedrigi RM, Fuchshofer R, Braakman ST, Gupta R, Perkumas KM, Sherwood JM, Vahabikashi A, et al. Altered mechanobiology of Schlemm's canal endothelial cells in glaucoma. Proc Natl Acad Sci U S A. 2014;111:13876–13881. doi: 10.1073/pnas.1410602111 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Sakata J, Asada Y, Shimokubo T, Kitani M, Inatsu H, Kitamura K, Kangawa K, Matsuo H, Sumiyoshi A, Eto T. Adrenomedullin in the gastrointestinal tract. Distribution and gene expression in rat and augmented gastric adrenomedullin after fasting. J Gastroenterol. 1998;33:828–834. doi: 10.1007/s005350050183 [DOI] [PubMed] [Google Scholar]
- 164.Martinez-Herrero S, Martinez A. Adrenomedullin regulates intestinal physiology and pathophysiology. Domest Anim Endocrinol. 2016;56 Suppl:S66–83. doi: 10.1016/j.domaniend.2016.02.004 [DOI] [PubMed] [Google Scholar]
- 165.Fukuda K, Tsukada H, Oya M, Onomura M, Kodama M, Nakamura H, Hosokawa M, Seino Y. Adrenomedullin promotes epithelial restitution of rat and human gastric mucosa in vitro. Peptides. 1999;20:127–132. doi: 10.1016/s0196-9781(98)00146-6 [DOI] [PubMed] [Google Scholar]
- 166.Fernandez de Arcaya I, Lostao MP, Martinez A, Berjon A, Barber A. Effect of adrenomedullin and proadrenomedullin N-terminal 20 peptide on sugar transport in the rat intestine. Regul Pept. 2005;129:147–154. doi: 10.1016/j.regpep.2005.02.001 [DOI] [PubMed] [Google Scholar]
- 167.Fukuda K, Tsukada H, Onomura M, Saito T, Kodama M, Nakamura H, Taniguchi T, Tominaga M, Hosokawa M, Seino Y. Effect of adrenomedullin on ion transport and muscle contraction in rat distal colon. Peptides. 1998;19:1043–1047. doi: 10.1016/s0196-9781(98)00043-6 [DOI] [PubMed] [Google Scholar]
- 168.Martinez-Herrero S, Larrayoz IM, Narro-Iniguez J, Rubio-Mediavilla S, Martinez A. Lack of Adrenomedullin Aggravates Acute TNBS-Induced Colitis Symptoms in Mice, Especially in Females. Front Physiol. 2017;8:1058. doi: 10.3389/fphys.2017.01058 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Martinez-Herrero S, Larrayoz IM, Narro-Iniguez J, Villanueva-Millan MJ, Recio-Fernandez E, Perez-Matute P, Oteo JA, Martinez A. Lack of Adrenomedullin Results in Microbiota Changes and Aggravates Azoxymethane and Dextran Sulfate Sodium-Induced Colitis in Mice. Front Physiol. 2016;7:595. doi: 10.3389/fphys.2016.00595 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Ashizuka S, Inagaki-Ohara K, Kuwasako K, Kato J, Inatsu H, Kitamura K. Adrenomedullin treatment reduces intestinal inflammation and maintains epithelial barrier function in mice administered dextran sulphate sodium. Microbiol Immunol. 2009;53:573–581. doi: 10.1111/j.1348-0421.2009.00159.x [DOI] [PubMed] [Google Scholar]
- 171.Ashizuka S, Ishikawa N, Kato J, Yamaga J, Inatsu H, Eto T, Kitamura K. Effect of adrenomedullin administration on acetic acid-induced colitis in rats. Peptides. 2005;26:2610–2615. doi: 10.1016/j.peptides.2005.05.007 [DOI] [PubMed] [Google Scholar]
- 172.Ashizuka S, Kita T, Inatsu H, Kitamura K. Adrenomedullin: a novel therapy for intractable ulcerative colitis. Inflamm Bowel Dis. 2013;19:E26–27. doi: 10.1002/ibd.22891 [DOI] [PubMed] [Google Scholar]
- 173.Ashizuka S, Inatsu H, Kita T, Kitamura K. Adrenomedullin Therapy in Patients with Refractory Ulcerative Colitis: A Case Series. Dig Dis Sci. 2016;61:872–880. doi: 10.1007/s10620-015-3917-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Kita T, Ashizuka S, Ohmiya N, Yamamoto T, Kanai T, Motoya S, Hirai F, Nakase H, Moriyama T, Nakamura M, et al. Adrenomedullin for steroid-resistant ulcerative colitis: a randomized, double-blind, placebo-controlled phase-2a clinical trial. J Gastroenterol. 2021;56:147–157. doi: 10.1007/s00535-020-01741-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Kita T, Ashizuka S, Takeda T, Matsumoto T, Ohmiya N, Nakase H, Motoya S, Ohi H, Mitsuyama K, Hisamatsu T, et al. Adrenomedullin for biologic-resistant Crohn’s disease: A randomized, double-blind, placebo-controlled phase 2a clinical trial. J Gastroenterol Hepatol. 2022;37:2051–2059. doi: 10.1111/jgh.15945 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Kubo K, Tokashiki M, Kuwasako K, Tamura M, Tsuda S, Kubo S, Yoshizawa-Kumagaye K, Kato J, Kitamura K. Biological properties of adrenomedullin conjugated with polyethylene glycol. Peptides. 2014;57:118–121. doi: 10.1016/j.peptides.2014.05.005 [DOI] [PubMed] [Google Scholar]
- 177.Nilsson C, Hansen TK, Rosenquist C, Hartmann B, Kodra JT, Lau JF, Clausen TR, Raun K, Sams A. Long acting analogue of the calcitonin gene-related peptide induces positive metabolic effects and secretion of the glucagon-like peptide-1. Eur J Pharmacol. 2016;773:24–31. doi: 10.1016/j.ejphar.2016.01.003 [DOI] [PubMed] [Google Scholar]
- 178.Aubdool AA, Thakore P, Argunhan F, Smillie SJ, Schnelle M, Srivastava S, Alawi KM, Wilde E, Mitchell J, Farrell-Dillon K, et al. A Novel alpha-Calcitonin Gene-Related Peptide Analogue Protects Against End-Organ Damage in Experimental Hypertension, Cardiac Hypertrophy, and Heart Failure. Circulation. 2017;136:367–383. doi: 10.1161/CIRCULATIONAHA.117.028388 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Booe JM, Warner ML, Pioszak AA. Picomolar Affinity Antagonist and Sustained Signaling Agonist Peptide Ligands for the Adrenomedullin and Calcitonin Gene-Related Peptide Receptors. ACS Pharmacol Transl Sci. 2020;3:759–772. doi: 10.1021/acsptsci.0c00031 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Hendrikse ER, Liew LP, Bower RL, Bonnet M, Jamaluddin MA, Prodan N, Richards KD, Walker CS, Pairaudeau G, Smith DM, et al. Identification of Small-Molecule Positive Modulators of Calcitonin-like Receptor-Based Receptors. ACS Pharmacol Transl Sci. 2020;3:305–320. doi: 10.1021/acsptsci.9b00108 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.Chang CL, Hsu SYT. Development of chimeric and bifunctional antagonists for CLR/RAMP receptors. PLoS One. 2019;14:e0216996. doi: 10.1371/journal.pone.0216996 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.Chang CL, Cai Z, Hsu SYT. Sustained Activation of CLR/RAMP Receptors by Gel-Forming Agonists. Int J Mol Sci. 2022;23. doi: 10.3390/ijms232113408 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Arfsten H, Goliasch G, Bartko PE, Prausmuller S, Spinka G, Cho A, Novak J, Haslacher H, Strunk G, Struck J, et al. Increased concentrations of bioactive adrenomedullin subsequently to angiotensin-receptor/neprilysin-inhibitor treatment in chronic systolic heart failure. Br J Clin Pharmacol. 2021;87:916–924. doi: 10.1111/bcp.14442 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184.Menhaji-Klotz E, Ward J, Brown JA, Loria PM, Tan C, Hesp KD, Riccardi KA, Litchfield J, Boehm M. Discovery of Diphenylacetamides as CXCR7 Inhibitors with Novel beta-Arrestin Antagonist Activity. ACS Med Chem Lett. 2020;11:1330–1334. doi: 10.1021/acsmedchemlett.0c00163 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185.Karakas M, Akin I, Burdelski C, Clemmensen P, Grahn H, Jarczak D, Kessler M, Kirchhof P, Landmesser U, Lezius S, et al. Single-dose of adrecizumab versus placebo in acute cardiogenic shock (ACCOST-HH): an investigator-initiated, randomised, double-blinded, placebo-controlled, multicentre trial. Lancet Respir Med. 2022;10:247–254. doi: 10.1016/S2213-2600(21)00439-2 [DOI] [PubMed] [Google Scholar]
- 186.Simonavicius J, Mikalauskas A, Cerlinskaite K, Gayat E, Juknevicius V, Paleviciute E, Alitoit-Marrote I, Kablucko D, Bagdonaite L, Balciunas M, et al. Biologically Active Adrenomedullin (bio-ADM) is of Potential Value in Identifying Congestion and Selecting Patients for Neurohormonal Blockade in Acute Dyspnea. Am J Med. 2022;135:e165–e181. doi: 10.1016/j.amjmed.2022.02.006 [DOI] [PubMed] [Google Scholar]
- 187.Maisel A, Mueller C, Nowak RM, Peacock WF, Ponikowski P, Mockel M, Hogan C, Wu AH, Richards M, Clopton P, et al. Midregion prohormone adrenomedullin and prognosis in patients presenting with acute dyspnea: results from the BACH (Biomarkers in Acute Heart Failure) trial. J Am Coll Cardiol. 2011;58:1057–1067. doi: 10.1016/j.jacc.2011.06.006 [DOI] [PubMed] [Google Scholar]
- 188.Miguel D, Prieto B, Costa M, Coto D, Alvarez FV. Cord blood plasma reference intervals for potential sepsis markers: pro-adrenomedullin, pro-endothelin, and pro-atrial natriuretic peptide. Clin Biochem. 2011;44:337–341. doi: 10.1016/j.clinbiochem.2010.12.012 [DOI] [PubMed] [Google Scholar]
- 189.Mebazaa A, Geven C, Hollinger A, Wittebole X, Chousterman BG, Blet A, Gayat E, Hartmann O, Scigalla P, Struck J, et al. Circulating adrenomedullin estimates survival and reversibility of organ failure in sepsis: the prospective observational multinational Adrenomedullin and Outcome in Sepsis and Septic Shock-1 (AdrenOSS-1) study. Crit Care. 2018;22:354. doi: 10.1186/s13054-018-2243-2 [DOI] [PMC free article] [PubMed] [Google Scholar]


