1
|
Hüttl M, Miklovič M, Gawryś O, Molnár M, Škaroupková P, Vaňourková Z, Kikerlová S, Malínská H, Kala P, Honetschlägerová Z, Sadowski J, Hošková L, Sandner P, Melenovský V, Táborský M, Šnorek M, Červenka L. The treatment with soluble guanylate cyclase stimulator BAY41-8543 prevents malignant hypertension and associated organ damage. J Hypertens 2025; 43:1030-1041. [PMID: 40197357 PMCID: PMC12052048 DOI: 10.1097/hjh.0000000000004009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 11/07/2024] [Accepted: 02/27/2025] [Indexed: 04/10/2025]
Abstract
OBJECTIVE Despite availability of an array of antihypertensive drugs, malignant hypertension remains a life-threatening condition, and new therapeutic strategies for the treatment of malignant hypertension and malignant hypertension-associated organ damage are needed. The aim of the present study was to assess the effects of nitric oxide (NO)-independent soluble guanylyl cyclase (sGC) stimulator on the course of malignant hypertension. The second aim was to investigate if the treatment with sodium-glucose cotransporter type 2 (SGLT2) inhibitor would augment the expected beneficial actions of the sGC stimulation on the course of malignant hypertension. METHODS As a model of malignant hypertension, Ren-2 transgenic rats (TGR) treated with nonspecific NO synthase inhibitor (Nω-nitro- l -arginine methyl ester, l -NAME) was used. Blood pressure (BP) was monitored by radiotelemetry, and the treatment was started 3 days before administration of l -NAME. RESULTS The treatment with sGC stimulator BAY 41-8543, alone or combined with SGLT2 inhibitor empagliflozin, abolished malignant hypertension-related mortality in TGR receiving l -NAME. These two treatment regimens also prevented BP increases after l -NAME administration in TGR, and even decreased BP below values observed in control TGR, and prevented cardiac dysfunction and malignant hypertension-related morbidity. The treatment with the SGLT2 inhibitor empagliflozin did not further augment the beneficial actions of sGC stimulator on the course of malignant hypertension-related mortality. CONCLUSION The treatment with NO-independent sGC stimulator displayed marked protective actions on the course of malignant hypertension-related mortality and malignant hypertension-related cardiac damage. This suggests that application of sGC stimulator could be a promising therapeutic means for the treatment of malignant hypertension.
Collapse
Affiliation(s)
- Martina Hüttl
- Center for Experimental Medicine, Institute for Clinical and Experimental Medicine
| | - Matúš Miklovič
- Center for Experimental Medicine, Institute for Clinical and Experimental Medicine
- Department of Pathophysiology, 2nd Faculty of Medicine, Charles University
| | - Olga Gawryś
- Center for Experimental Medicine, Institute for Clinical and Experimental Medicine
| | - Matěj Molnár
- Center for Experimental Medicine, Institute for Clinical and Experimental Medicine
- Department of Pathophysiology, 2nd Faculty of Medicine, Charles University
| | - Petra Škaroupková
- Center for Experimental Medicine, Institute for Clinical and Experimental Medicine
| | - Zdeňka Vaňourková
- Center for Experimental Medicine, Institute for Clinical and Experimental Medicine
| | - Soňa Kikerlová
- Center for Experimental Medicine, Institute for Clinical and Experimental Medicine
| | - Hana Malínská
- Center for Experimental Medicine, Institute for Clinical and Experimental Medicine
| | - Petr Kala
- Center for Experimental Medicine, Institute for Clinical and Experimental Medicine
- Department of Cardiology, University Hospital Motol and 2nd Faculty of Medicine, Charles University
| | | | - Janusz Sadowski
- Center for Experimental Medicine, Institute for Clinical and Experimental Medicine
| | - Lenka Hošková
- Department of Cardiology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Peter Sandner
- Bayer AG, Pharmaceuticals R&D, Pharma Research Center, Wuppertal, Germany
- Hannover Medical School, Hannover, Germany
| | - Vojtěch Melenovský
- Department of Cardiology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Miloš Táborský
- Department of Internal Medicine I, Cardiology, University Hospital Olomouc and Palacký University, Olomouc
| | - Michal Šnorek
- Department of Cardiology, Ceske Budejovice Hospital, Ceske Budejovice, Czech Republic
| | - Luděk Červenka
- Center for Experimental Medicine, Institute for Clinical and Experimental Medicine
- Department of Internal Medicine I, Cardiology, University Hospital Olomouc and Palacký University, Olomouc
| |
Collapse
|
2
|
Bagheri L, Javanbakht M, Malekian S, Ghahderijani BH, Taghipour S, Tanha FD, Ranjkesh M, Cegolon L, Zhao S. Antifibrotic therapeutic strategies in systemic sclerosis: Critical role of the Wnt/β-catenin and TGF-β signal transduction pathways as potential targets. Eur J Pharmacol 2025:177607. [PMID: 40209848 DOI: 10.1016/j.ejphar.2025.177607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 03/25/2025] [Accepted: 04/07/2025] [Indexed: 04/12/2025]
Abstract
Systemic sclerosis (SSc) is a prototypic fibrosing disorder characterized by widespread fibrosis and immune dysregulation. Current evidence highlights the intricate cross-talk between the canonical Wnt/β-catenin signaling pathway and transforming growth factor-beta (TGF-β) signaling, both of which play fundamental roles in the pathogenesis of fibrosis. This review aims to elucidate the central role of the Wnt/β-catenin-TGF-β pathway and TGF-β signal transduction pathway in fibrotic diseases, focusing on SSc. We summarized evidence from cellular biology studies, animal model investigations, and clinical observations to provide a comprehensive view of the mechanisms by which these pathways cause pathological fibrosis. In addition, we explore the possibilities of antifibrotic therapeutic strategies against Wnt/β-catenin-TGF-β signaling to counteract fibrosis. We aim to delineate approaches towards effectively treating fibrosis in SSc by targeting these interconnected signaling pathways.
Collapse
Affiliation(s)
- Leyla Bagheri
- Department of Internal Medicine, Shahid Modarres Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Mohammad Javanbakht
- Nephrology and Urology Research Center, Clinical Science Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Sheida Malekian
- Department of Internal Medicine, Shahid Modarres Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Sadra Taghipour
- Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Fatemeh Davari Tanha
- Department of Infertility, Yas Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Luca Cegolon
- Department of Medical, Surgical & Health Sciences, University of Trieste, 34128 Trieste, Italy; Public Health Department, University Health Agency Giuliano-Isontina (ASUGI), 34148 Trieste, Italy
| | - Shi Zhao
- School of Public Health, Tianjin Medical University, Tianjin, 300070, China
| |
Collapse
|
3
|
Martišková A, Sýkora M, Andelová N, Ferko M, Gawrys O, Andelová K, Kala P, Červenka L, Szeiffová Bačová B. Soluble Guanylate Cyclase Stimulator, BAY41-8543: A Promising Approach for the Treatment of Chronic Heart Failure Caused by Pressure and Volume Overload. Pharmacol Res Perspect 2025; 13:e70087. [PMID: 40159447 PMCID: PMC11955242 DOI: 10.1002/prp2.70087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 02/28/2025] [Accepted: 03/09/2025] [Indexed: 04/02/2025] Open
Abstract
Heart failure (HF) is a leading cause of morbidity and mortality, often driven by prolonged exposure to pathological stimuli such as pressure and volume overload. These factors contribute to excessive oxidative stress, adverse cardiac remodeling, and dysregulation of the nitric oxide-soluble guanylate cyclase-cyclic guanosine monophosphate (NO-sGC-cGMP) signaling pathway. Given the urgent need for effective treatments, this study investigated the potential of sGC stimulators to mitigate HF progression. We utilized male hypertensive Ren-2 transgenic (TGR) rats and a volume-overload HF model induced by an aortocaval fistula (ACF). Rats received the sGC stimulator BAY 41-8543 (3 mg/kg/day) for 30 weeks, while normotensive Hannover Sprague-Dawley rats served as controls. At the study endpoint (40 weeks of age), left ventricular tissue was analyzed using mass spectrometry, Western blotting, and histological assessment. TGR rats treated with sGC stimulators exhibited a significant increase in key antioxidant proteins (SOD1, CH10, ACSF2, NDUS1, DHE3, GSTM2, and PCCA), suggesting enhanced resistance to oxidative stress. However, sGC stimulator treatment also upregulated extracellular matrix remodeling markers (MMP-2, TGF-β, and SMAD2/3), which are typically associated with fibrosis. Despite this, Masson's trichrome staining revealed reduced collagen deposition in both TGR and TGR-ACF rats receiving sGC stimulators. Notably, all untreated TGR-ACF rats succumbed before the study endpoint, preventing direct assessment of sGC stimulator effects in advanced HF. These findings highlight the therapeutic potential of sGC stimulators in HF, particularly through their antioxidant effects. However, their concurrent influence on fibrosis warrants further investigation to optimize treatment strategies.
Collapse
Affiliation(s)
- Adriana Martišková
- Centre of Experimental MedicineSlovak Academy of Sciences, Institute for Heart ResearchBratislavaSlovakia
| | - Matúš Sýkora
- Centre of Experimental MedicineSlovak Academy of Sciences, Institute for Heart ResearchBratislavaSlovakia
| | - Natália Andelová
- Centre of Experimental MedicineSlovak Academy of Sciences, Institute for Heart ResearchBratislavaSlovakia
| | - Miroslav Ferko
- Centre of Experimental MedicineSlovak Academy of Sciences, Institute for Heart ResearchBratislavaSlovakia
| | - Olga Gawrys
- Centre for Experimental MedicineInstitute for Clinical and Experimental MedicinePragueCzech Republic
| | - Katarína Andelová
- Centre of Experimental MedicineSlovak Academy of Sciences, Institute for Heart ResearchBratislavaSlovakia
| | - Petr Kala
- Centre for Experimental MedicineInstitute for Clinical and Experimental MedicinePragueCzech Republic
| | - Luděk Červenka
- Centre for Experimental MedicineInstitute for Clinical and Experimental MedicinePragueCzech Republic
| | - Barbara Szeiffová Bačová
- Centre of Experimental MedicineSlovak Academy of Sciences, Institute for Heart ResearchBratislavaSlovakia
| |
Collapse
|
4
|
Tawa M, Nakagawa K, Ohkita M. Soluble guanylate cyclase stimulators and activators as potential antihypertensive drugs. Hypertens Res 2025; 48:1458-1470. [PMID: 39833553 DOI: 10.1038/s41440-025-02110-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 12/09/2024] [Accepted: 12/29/2024] [Indexed: 01/22/2025]
Abstract
Poor blood pressure control in treated patients with hypertension is an important topic in the field of hypertension, and an unmet need for new therapeutic drugs remains. Soluble guanylate cyclase (sGC), a key signal transduction enzyme responsible for vasodilation, has attracted increasing interest as a therapeutic target in various cardiovascular diseases. Two different sGC agonists, sGC stimulators and activators, can increase its enzymatic activity in reduced and oxidized/apo forms, respectively. With some sGC agonists being already in clinical use, drugs in this category are expected to become new therapeutic agents for various conditions, including hypertension. In this review, we summarize the current knowledge on the antihypertensive effects of sGC agonists in various preclinical studies involving animal models of spontaneous hypertension, salt-sensitive hypertension, nitric oxide-deficient hypertension, renin-angiotensin-aldosterone system-dependent hypertension, malignant hypertension, metabolic syndrome, renoprival hypertension, renovascular hypertension, drug-induced hypertension, pregnancy hypertension, and treatment-resistant hypertension. Our compilation provides a comprehensive rationale for advancing the clinical development of sGC agonists for the treatment of hypertension.
Collapse
Affiliation(s)
- Masashi Tawa
- Department of Pathological and Molecular Pharmacology, Faculty of Pharmacy, Osaka Medical and Pharmaceutical University, Takatsuki, Osaka, Japan.
| | - Keisuke Nakagawa
- Department of Pathological and Molecular Pharmacology, Faculty of Pharmacy, Osaka Medical and Pharmaceutical University, Takatsuki, Osaka, Japan
| | - Mamoru Ohkita
- Department of Pathological and Molecular Pharmacology, Faculty of Pharmacy, Osaka Medical and Pharmaceutical University, Takatsuki, Osaka, Japan
| |
Collapse
|
5
|
Ihadadene K, Fallatah AHA, Zhu Y, Tolone A, Paquet‐Durand F. Inhibition of cGMP-Signalling Rescues Retinal Ganglion Cells From Axotomy-Induced Degeneration. J Neurochem 2025; 169:e70072. [PMID: 40270249 PMCID: PMC12019586 DOI: 10.1111/jnc.70072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 03/28/2025] [Accepted: 04/02/2025] [Indexed: 04/25/2025]
Abstract
The axons of retinal ganglion cells (RGCs) form the optic nerve, which relays visual information to the brain. RGC degeneration is the root cause of a variety of blinding diseases linked to optic nerve damage, including glaucoma, the second leading cause of blindness worldwide. The underlying cellular mechanisms of RGC degeneration are largely unclear; yet, they have been connected to excessive production of the signalling molecule nitric oxide (NO) by nitric oxide synthase (NOS). NO activates soluble guanylate cyclase (sGC), which subsequently produces the second messenger cyclic guanosine monophosphate (cGMP). This, in turn, activates protein kinase G (PKG), which can phosphorylate downstream protein targets. To study the role of NO/cGMP/PKG signalling in RGC degeneration, we used organotypic retinal explant cultures in which the optic nerve had been severed. We assessed the activity of NOS, RGC death and survival at different times after optic nerve transection. While NOS activity was high right after optic nerve transection, significant RGC loss occurred with a 24-48-h delay. We then treated retinal explants with inhibitors selectively targeting either NOS, sGC, PKG, or Kv1.3 and Kv1.6 voltage-gated potassium channels. While all four treatments reduced RGC death, the PKG inhibitor CN238 and the Kv-channel blocker Margatoxin (MrgX) showed the most pronounced rescue effects. Our results confirm an involvement of NO/cGMP/PKG signalling in RGC degeneration, highlight the potential of PKG and Kv1-channel targeting drugs for treatment development, and further suggest organotypic retinal explant cultures as a useful model for investigations into optic nerve damage.
Collapse
Affiliation(s)
- Katia Ihadadene
- Graduate School INTHERAPIBurgundy UniversityDijonFrance
- Institute for Ophthalmic ResearchUniversity of TübingenTübingenGermany
| | - Azdah Hamed A Fallatah
- Institute for Ophthalmic ResearchUniversity of TübingenTübingenGermany
- Graduate School for Molecular MedicineUniversity of TübingenTübingenGermany
- Graduate School for Cellular and Molecular NeuroscienceUniversity of TübingenTübingenGermany
| | - Yu Zhu
- Institute for Ophthalmic ResearchUniversity of TübingenTübingenGermany
- Graduate School for Cellular and Molecular NeuroscienceUniversity of TübingenTübingenGermany
| | - Arianna Tolone
- Institute for Ophthalmic ResearchUniversity of TübingenTübingenGermany
| | | |
Collapse
|
6
|
Quagliariello V, Berretta M, Bisceglia I, Giacobbe I, Iovine M, Barbato M, Maurea C, Canale ML, Paccone A, Inno A, Scherillo M, Oliva S, Cadeddu Dessalvi C, Mauriello A, Fonderico C, Maratea AC, Gabrielli D, Maurea N. In the Era of Cardiovascular-Kidney-Metabolic Syndrome in Cardio-Oncology: From Pathogenesis to Prevention and Therapy. Cancers (Basel) 2025; 17:1169. [PMID: 40227756 PMCID: PMC11988012 DOI: 10.3390/cancers17071169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 03/21/2025] [Accepted: 03/27/2025] [Indexed: 04/15/2025] Open
Abstract
Cardiovascular-kidney-metabolic (CKM) syndrome represents a complex interplay between cardiovascular disease (CVD), chronic kidney disease (CKD), and metabolic disorders, significantly impacting cancer patients. The presence of CKM syndrome in cancer patients not only worsens their prognosis but also increases the risk of major adverse cardiovascular events (MACE), reduces quality of life (QoL), and affects overall survival (OS). Furthermore, several anticancer therapies, including anthracyclines, tyrosine kinase inhibitors, immune checkpoint inhibitors, and hormonal treatments, can exacerbate CKM syndrome by inducing cardiotoxicity, nephrotoxicity, and metabolic dysregulation. This review explores the pathophysiology of CKM syndrome in cancer patients and highlights emerging therapeutic strategies to mitigate its impact. We discuss the role of novel pharmacological interventions, including sodium-glucose cotransporter-2 inhibitors (SGLT2i), proprotein convertase subtilisin/kexin type 9 inhibitors (PCSK9i), and soluble guanylate cyclase (sGC) activators, as well as dietary and lifestyle interventions. Optimizing the management of CKM syndrome in cancer patients is crucial to improving OS, enhancing QoL, and reducing MACE. By integrating cardiometabolic therapies into oncologic care, we can create a more comprehensive treatment approach that reduces the burden of cardiovascular and renal complications in this vulnerable population. Further research is needed to establish personalized strategies for CKM syndrome prevention and treatment in cancer patients.
Collapse
Affiliation(s)
- Vincenzo Quagliariello
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy
| | - Massimiliano Berretta
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy
| | - Irma Bisceglia
- Servizi Cardiologici Integrati, Dipartimento Cardio-Toraco-Vascolare, Azienda Ospedaliera San Camillo Forlanini, 00148 Rome, Italy
| | - Ilaria Giacobbe
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy
| | - Martina Iovine
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy
| | - Matteo Barbato
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy
| | - Carlo Maurea
- ASL NA1, UOC Neurology and Stroke Unit, Ospedale del Mare, 23807 Naples, Italy
| | | | - Andrea Paccone
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy
| | - Alessandro Inno
- Medical Oncology, IRCCS Ospedale Sacro Cuore Don Calabria, 37024 Negrar di Valpolicella, Italy
| | - Marino Scherillo
- Cardiologia Interventistica e UTIC, A.O. San Pio, Presidio Ospedaliero Gaetano Rummo, 82100 Benevento, Italy
| | - Stefano Oliva
- Cardio-Oncology Unit, IRCCS Istituto Tumori, “Giovanni Paolo II”, 70124 Bari, Italy
| | | | - Alfredo Mauriello
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy
| | - Celeste Fonderico
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy
| | - Anna Chiara Maratea
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy
| | - Domenico Gabrielli
- U.O.C. Cardiologia, Dipartimento Cardio-Toraco-Vascolare, Azienda Ospedaliera San Camillo Forlanini, 00152 Rome, Italy
| | - Nicola Maurea
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy
| |
Collapse
|
7
|
Sharma N, Liu W, Tsai XQE, Wang Z, Outtrim C, Tang A, Pieper MP, Reinhart GA, Huang Y. A novel soluble guanylate cyclase activator, avenciguat, in combination with empagliflozin, protects against renal and hepatic injury in diabetic db/db mice. Am J Physiol Endocrinol Metab 2025; 328:E362-E376. [PMID: 39907739 DOI: 10.1152/ajpendo.00254.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 08/16/2024] [Accepted: 11/15/2024] [Indexed: 02/06/2025]
Abstract
Diabetic complications are linked to oxidative stress, which hampers the cyclic guanosine monophosphate production by inhibiting nitric oxide/soluble guanylate cyclase (sGC) signaling. This study aimed to determine whether the administration of a novel sGC activator avenciguat alone or in combination with an SGLT2 inhibitor could slow the progression of renal and liver fibrosis in the type 2 diabetic and uninephrectomized db/db mouse model. Experiment groups included normal controls, untreated db/db mice terminated at 12 and 18 wk of age, and db/db mice treated with either one of two doses of avenciguat alone, empagliflozin (Empa) alone, or a combination of both from weeks 12 to 18 of age. Untreated db/db mice exhibited obesity, hyperglycemia, elevated levels of HbA1c and triglycerides (TG), and developed progressive albuminuria, glomerulosclerosis, fatty liver, and liver fibrosis between weeks 12 and 18 of age, accompanied by increased renal and liver production of fibronectin, type-IV collagen, laminin, and increased oxidative stress markers. Avenciguat had no effect on body weight but reduced both blood HbA1c and TG levels, whereas Empa reduced HbA1c but not TG levels as compared with untreated db/db. Both avenciguat and Empa alone effectively slowed the progression of diabetes-associated glomerulosclerosis and liver fibrosis. Importantly, avenciguat, especially at high doses in combination with Empa, further lowered these progression markers compared with baseline measurements. These results suggested that either avenciguat alone or in combination with Empa is therapeutic. Avenciguat in combination with Empa shows promise in halting the progression of diabetic complications.NEW & NOTEWORTHY Whether combining an sGC activator with an SGLT2 inhibitor could better control diabetes-associated oxidative stress and NO-cGMP signal deficiency has not yet been explored. Using the type 2 diabetic db/db mouse model, this study underscores the sGC activator avenciguat as a novel therapy for diabetic nephropathy and liver injury beyond sGLT2 inhibitors. It also highlights the need for further investigation into the combined effects of these two treatments in managing diabetic complications.
Collapse
Affiliation(s)
- Nisha Sharma
- Department of Internal Medicine, Division of Nephrology & Hypertension, University of Utah Health, Salt Lake City, Utah, United States
| | - Wenjin Liu
- Department of Internal Medicine, Division of Nephrology & Hypertension, University of Utah Health, Salt Lake City, Utah, United States
| | - Xiao-Qing E Tsai
- Department of Internal Medicine, Division of Nephrology & Hypertension, University of Utah Health, Salt Lake City, Utah, United States
| | - Zhou Wang
- Department of Internal Medicine, Division of Nephrology & Hypertension, University of Utah Health, Salt Lake City, Utah, United States
| | - Connor Outtrim
- Department of Internal Medicine, Division of Nephrology & Hypertension, University of Utah Health, Salt Lake City, Utah, United States
| | - Anna Tang
- Department of Internal Medicine, Division of Nephrology & Hypertension, University of Utah Health, Salt Lake City, Utah, United States
| | - Michael P Pieper
- Global Cardio-metabolic Diseases, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Glenn A Reinhart
- Cardiometabolic Diseases Research, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut, United States
| | - Yufeng Huang
- Department of Internal Medicine, Division of Nephrology & Hypertension, University of Utah Health, Salt Lake City, Utah, United States
| |
Collapse
|
8
|
Fonseca C, Baptista R, Franco F, Moura B, Pimenta J, Moraes Sarmento P, Cardoso JS, Brito D. Worsening heart failure: progress, pitfalls, and perspectives. Heart Fail Rev 2025:10.1007/s10741-025-10497-z. [PMID: 39976853 DOI: 10.1007/s10741-025-10497-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/11/2025] [Indexed: 03/28/2025]
Abstract
For most patients with chronic heart failure (HF), the clinical course of the disease includes periods of apparent clinical stability punctuated by episodes of clinical deterioration with worsening signs and symptoms, a condition referred to as worsening heart failure (WHF). Over time, episodes of WHF may become more frequent, and patients may enter a cycle of recurrent events associated with deterioration in their quality of life and functional capacity, hospitalizations, and ultimately death. WHF is apparently an old concept but seems to have acquired new boundaries in terms of definition and clinical and prognostic value due to the fast-paced evolution of the HF treatment landscape and the emergence of new drugs in this setting. As a result, the management of WHF is being reshaped. In the present paper, a group of HF experts gathered to discuss the concept, prevention, detection, and treatment of WHF.
Collapse
Affiliation(s)
- Cândida Fonseca
- Heart Failure Clinic, Hospital S. Francisco Xavier, Unidade Local de Saúde Lisboa Ocidental, Lisbon, Portugal.
- Internal Medicine Department, Hospital de S. Francisco Xavier, Unidade Local de Saúde Lisboa Ocidental, Estrada Forte Do Alto Do Duque, 1449-005, Lisbon, Portugal.
- NOVA Medical School, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisbon, Portugal.
| | - Rui Baptista
- Cardiology Department, Unidade Local de Saúde de Entre Douro E Vouga, Santa Maria da Feira, Portugal
- Faculty of Medicine of the University of Coimbra, Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
| | - Fátima Franco
- Advanced Heart Failure Unit, Cardiology Department, Unidade Local de Saúde Coimbra, Coimbra, Portugal
| | - Brenda Moura
- Hospital das Forças Armadas, Porto Campus, Porto, Portugal
- Faculty of Medicine of the University of Porto, Porto, Portugal
| | - Joana Pimenta
- Internal Medicine Department, Hospital Eduardo Santos Silva, Unidade Local de Saúde Gaia e Espinho, Vila Nova de Gaia, Portugal
- Cardiovascular R&D Centre - UnIC@RISE, Department of Medicine, Faculty of Medicine of the University of Porto, Porto, Portugal
| | - Pedro Moraes Sarmento
- Heart Failure Clinic, Hospital da Luz, Lisbon, Portugal
- Centro de Investigação Clínica, Hospital da Luz Learning Health, Lisbon, Portugal
- Faculdade de Medicina, Católica Medical School, Universidade Católica Portuguesa, Lisbon, Portugal
| | - José Silva Cardoso
- Department of Cardiology, Unidade Local de Saúde São João, Porto, Portugal
- Center for Health Technology and Services Research (CINTESIS), Faculty of Medicine of the University of Porto, Porto, Portugal
- RISE-Health, Faculty of Medicine of the University of Porto, Porto, Portugal
| | - Dulce Brito
- Department of Cardiology, Unidade Local de Saúde Santa Maria, Lisboa, Portugal
- Lisbon Academic Medical Center (CAML), Lisbon, Portugal
- Centro Cardiovascular da Universidade de Lisboa (CCUL@RISE), Faculty of Medicine of the University of Lisbon, Lisbon, Portugal
| |
Collapse
|
9
|
Polhemus D, Almodiel D, Harb T, Ziogos E, Amat-Codina N, Ranek M, Santhanam L, Gerstenblith G, Leucker TM. Vericiguat prevents high glucose-mediated impaired vascular smooth muscle cGMP production and vasorelaxation. Sci Rep 2025; 15:4939. [PMID: 39929946 PMCID: PMC11811225 DOI: 10.1038/s41598-025-88938-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 01/31/2025] [Indexed: 02/13/2025] Open
Abstract
Normal endothelial cell dependent vascular smooth muscle cell function is mediated by nitric oxide (NO), which stimulates soluble guanylyl cyclase (sGC) production of the second messenger cyclic guanosine monophosphate (cGMP) leading to increased protein kinase G (PKG) activity and vascular smooth muscle relaxation. NO bioavailability is impaired in high glucose (HG). We tested the hypothesis that the sGC sensitizer vericiguat reverses HG-mediated decreased sGC activity in two experimental models, human aortic vascular smooth muscle cells (HVSMCs) and isolated mouse aortic rings. HVSMCs were exposed to normal glucose (NG) or to HG with or without 1 μm vericiguat for 24 h and cGMP and PKG activity were measured. Murine aortic rings were incubated in NG or HG for 24 h. Following incubation, the aortic rings were placed in an organ chamber bath containing the same NG or HG concentration used during the incubation. Dose-response curves to increasing concentrations of acetylcholine (ACh) and sodium nitroprusside were constructed for four groups: control (NG), NG + vericiguat, HG, and HG + vericiguat. As compared with the results in the NG group, cGMP production and PKG activity were significantly impaired in the HG cells incubated without, but not in those incubated with, vericiguat. In isolated aortic rings, ACh-mediated relaxation was impaired following treatment with HG, but not when a HG group was treated with vericiguat. The findings suggest clinical studies are warranted to investigate the potential of sGC sensitization as a therapeutic intervention to improve vascular NO-cGMP signaling endothelium -dependent function that is impaired in HG settings such as diabetes and the metabolic syndrome.
Collapse
Affiliation(s)
- David Polhemus
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Diego Almodiel
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Tarek Harb
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Efthymios Ziogos
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Nuria Amat-Codina
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Mark Ranek
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Lakshmi Santhanam
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Gary Gerstenblith
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Thorsten M Leucker
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
10
|
Abdelrahman AM, Al Maskari R, Ali H, Manoj P, Al Suleimani Y. Effect of Riociguat on Adenine-Induced Chronic Kidney Disease in Rats. BIOLOGY 2025; 14:161. [PMID: 40001929 PMCID: PMC11851967 DOI: 10.3390/biology14020161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Revised: 01/22/2025] [Accepted: 01/26/2025] [Indexed: 02/27/2025]
Abstract
Riociguat is a soluble guanylate cyclase (sGC) activator that increases the levels of cyclic guanosine monophosphate (cGMP). cGMP is known to play a key role in regulating kidney function. This research sought to investigate the possible protective effects of riociguat on the kidneys in the context of chronic kidney disease (CKD). CKD was induced in male Wistar rats through adenine administration. A total of 24 rats were allocated into four groups and administered treatments over a period of 35 days. Group 1 received a normal diet and a vehicle (carboxymethylcellulose (0.5%)), serving as the control. Group 2 received adenine (0.25% w/w) in the feed and a vehicle. Groups 3 and 4 received adenine in the feed (0.25% w/w) plus riociguat (3 mg/kg/day) and riociguat (10 mg/kg/day), respectively. Adenine administration significantly elevated systolic blood pressure, plasma creatinine, urea, and neutrophil gelatinase-associated lipocalin (NGAL). Furthermore, adenine reduced creatinine clearance and increased the urinary albumin-to-creatinine ratio and urinary N-Acetyl-β-D-Glucosaminidase (NAG). Histopathologically, adenine caused renal tubular necrosis and fibrosis. Furthermore, adenine elevated the plasma concentration of interleukins (IL-1β and IL-6) and tumor necrosis factor-alpha (TNF-α). Adenine significantly increased renal malondialdehyde (MDA) and reduced glutathione reductase (GR), superoxide dismutase (SOD), catalase (CAT), and total antioxidant capacity (TAC). Treatment with riociguat attenuated adenine-induced hypertension, improved kidney function, and ameliorated histopathological changes. Riociguat also reduced kidney injury markers, inflammation, and renal oxidative stress. The renoprotective effect of riociguat is probably due to anti-inflammatory and antioxidant actions. This indicates that riociguat may have the potential to slow the progression of kidney damage in chronic kidney disease (CKD).
Collapse
Affiliation(s)
- Aly M. Abdelrahman
- Department of Pharmacology and Clinical Pharmacy, College of Medicine and Health Sciences, Sultan Qaboos University, P.O. Box 35, Muscat 123, Oman; (A.M.A.); (R.A.M.); (P.M.)
| | - Raya Al Maskari
- Department of Pharmacology and Clinical Pharmacy, College of Medicine and Health Sciences, Sultan Qaboos University, P.O. Box 35, Muscat 123, Oman; (A.M.A.); (R.A.M.); (P.M.)
| | - Haytham Ali
- Department of Animal and Veterinary Sciences, College of Agricultural and Marine Sciences, Sultan Qaboos University, P.O. Box 35, Muscat 123, Oman;
| | - Priyadarsini Manoj
- Department of Pharmacology and Clinical Pharmacy, College of Medicine and Health Sciences, Sultan Qaboos University, P.O. Box 35, Muscat 123, Oman; (A.M.A.); (R.A.M.); (P.M.)
| | - Yousuf Al Suleimani
- Department of Pharmacology and Clinical Pharmacy, College of Medicine and Health Sciences, Sultan Qaboos University, P.O. Box 35, Muscat 123, Oman; (A.M.A.); (R.A.M.); (P.M.)
| |
Collapse
|
11
|
Zhang X, Chen Z, Xiong Y, Zhou Q, Zhu LQ, Liu D. The emerging role of nitric oxide in the synaptic dysfunction of vascular dementia. Neural Regen Res 2025; 20:402-415. [PMID: 38819044 PMCID: PMC11317957 DOI: 10.4103/nrr.nrr-d-23-01353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 10/23/2023] [Accepted: 11/30/2023] [Indexed: 06/01/2024] Open
Abstract
With an increase in global aging, the number of people affected by cerebrovascular diseases is also increasing, and the incidence of vascular dementia-closely related to cerebrovascular risk-is increasing at an epidemic rate. However, few therapeutic options exist that can markedly improve the cognitive impairment and prognosis of vascular dementia patients. Similarly in Alzheimer's disease and other neurological disorders, synaptic dysfunction is recognized as the main reason for cognitive decline. Nitric oxide is one of the ubiquitous gaseous cellular messengers involved in multiple physiological and pathological processes of the central nervous system. Recently, nitric oxide has been implicated in regulating synaptic plasticity and plays an important role in the pathogenesis of vascular dementia. This review introduces in detail the emerging role of nitric oxide in physiological and pathological states of vascular dementia and summarizes the diverse effects of nitric oxide on different aspects of synaptic dysfunction, neuroinflammation, oxidative stress, and blood-brain barrier dysfunction that underlie the progress of vascular dementia. Additionally, we propose that targeting the nitric oxide-sGC-cGMP pathway using certain specific approaches may provide a novel therapeutic strategy for vascular dementia.
Collapse
Affiliation(s)
- Xiaorong Zhang
- Department of Pathology, Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi Province, China
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi Province, China
- Center for Cognitive Science and Transdisciplinary Studies, Jiujiang University, Jiangxi Province, China
| | - Zhiying Chen
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi Province, China
- Department of Neurology, Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi Province, China
| | - Yinyi Xiong
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi Province, China
- Department of Rehabilitation, Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi Province, China
| | - Qin Zhou
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi Province, China
| | - Ling-Qiang Zhu
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Dan Liu
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| |
Collapse
|
12
|
Kreutz L, Gaab A, Dona M, Pinto AR, Tallquist MD, Groneberg D, Friebe A. Analysis of cellular NO-GC expression in the murine heart and lineage determination in angiotensin II-induced fibrosis. iScience 2025; 28:111615. [PMID: 39829679 PMCID: PMC11742323 DOI: 10.1016/j.isci.2024.111615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 11/22/2024] [Accepted: 12/12/2024] [Indexed: 01/22/2025] Open
Abstract
NO-sensitive guanylyl cyclase (NO-GC) is involved in the (patho)physiology of the mammalian heart. However, little is known about the individual cardiac cell types that express NO-GC and the role of the enzyme in cardiac fibrosis. Here, we describe the cellular expression of NO-GC in healthy and fibrotic murine myocardium; these data were compared with scRNA-seq data. In healthy myocardium, NO-GC is strongly expressed in pericytes and smooth muscle cells but not in endothelial cells or cardiomyocytes. Angiotensin II induced cardiac hypertrophy and fibrosis; fibrotic lesions contained cells positive for NO-GC identified as activated fibroblasts. Lineage tracing indicates that NO-GC-expressing activated fibroblasts originate from PDGFRβ- and Tcf21-positive fibroblast precursors. Our data indicate NO-GC expression in cardiac pericytes and SMC in naive myocardium and in activated fibroblast in fibrotic heart tissue. NO-mediated signaling may modulate fibrotic responses underlying the action of NO-GC stimulators used in the therapy of heart failure.
Collapse
Affiliation(s)
- Lennart Kreutz
- Physiologisches Institut, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - Annika Gaab
- Physiologisches Institut, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - Malathi Dona
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | | | - Michelle D. Tallquist
- Center for Cardiovascular Research, University of Hawaii at Manoa, Honolulu, HI, USA
| | - Dieter Groneberg
- Translational Center for Regenerative Therapies (TLC-RT), Fraunhofer Institute for Silicate Research (ISC), 97082 Würzburg, Germany
| | - Andreas Friebe
- Physiologisches Institut, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| |
Collapse
|
13
|
Gawrys O, Kala P, Sadowski J, Melenovský V, Sandner P, Červenka L. Soluble guanylyl cyclase stimulators and activators: Promising drugs for the treatment of hypertension? Eur J Pharmacol 2025; 987:177175. [PMID: 39645219 DOI: 10.1016/j.ejphar.2024.177175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 10/21/2024] [Accepted: 12/02/2024] [Indexed: 12/09/2024]
Abstract
Nitric oxide (NO)-stimulated cyclic guanosine monophosphate (cGMP) is a key regulator of cardiovascular health, as NO-cGMP signalling is impaired in diseases like pulmonary hypertension, heart failure and chronic kidney disease. The development of NO-independent sGC stimulators and activators provide a novel therapeutic option to restore altered NO signalling. sGC stimulators have been already approved for the treatment of pulmonary arterial hypertension (PAH), chronic thromboembolic pulmonary hypertension (CTEPH), and chronic heart failure (HFrEF), while sGC activators are currently in phase-2 clinical trials for CKD. The best characterized effect of increased cGMP via the NO-sGC-cGMP pathway is vasodilation. However, to date, none of the sGC agonists are in development for hypertension (HTN). According to WHO, the global prevalence of uncontrolled HTN continues to rise, contributing significantly to cardiovascular mortality. While there are effective antihypertensive treatments, many patients require multiple drugs, and some remain resistant to all therapies. Thus, in addition to improved diagnosis and lifestyle changes, new pharmacological strategies remain in high demand. In this review we explore the potential of sGC stimulators and activators as novel antihypertensive agents, starting with the overview of NO-sGC-cGMP signalling, followed by potential mechanisms by which the increase in cGMP may regulate vascular tone and BP. These effects may encompass not only acute vasodilation, but also mid-term and chronic effects, such as the regulation of salt and water balance, as well as mitigation of vascular ageing and remodelling. The main section summarizes the preclinical and clinical evidence supporting the BP-lowering efficacy of sGC agonists.
Collapse
Affiliation(s)
- Olga Gawrys
- Center for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic.
| | - Petr Kala
- Center for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic; Department of Cardiology, Motol University Hospital and Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Janusz Sadowski
- Center for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Vojtěch Melenovský
- Department of Cardiology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Peter Sandner
- Bayer AG, Pharmaceuticals, Drug Discovery, Pharma Research Centre, 42113, Wuppertal, Germany; Hannover Medical School, Institute of Pharmacology, 30625, Hannover, Germany
| | - Luděk Červenka
- Center for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic; First Department of Internal Medicine, Cardiology, Olomouc University Hospital and Palacký University, Olomouc, Czech Republic
| |
Collapse
|
14
|
DeVaughn H, Rich HE, Shadid A, Vaidya PK, Doursout MF, Shivshankar P. Complement Immune System in Pulmonary Hypertension-Cooperating Roles of Circadian Rhythmicity in Complement-Mediated Vascular Pathology. Int J Mol Sci 2024; 25:12823. [PMID: 39684535 PMCID: PMC11641342 DOI: 10.3390/ijms252312823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 11/21/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
Originally discovered in the 1890s, the complement system has traditionally been viewed as a "compliment" to the body's innate and adaptive immune response. However, emerging data have shown that the complement system is a much more complex mechanism within the body involved in regulating inflammation, gene transcription, attraction of macrophages, and many more processes. Sustained complement activation contributes to autoimmunity and chronic inflammation. Pulmonary hypertension is a disease with a poor prognosis and an average life expectancy of 2-3 years that leads to vascular remodeling of the pulmonary arteries; the pulmonary arteries are essential to host homeostasis, as they divert deoxygenated blood from the right ventricle of the heart to the lungs for gas exchange. This review focuses on direct links between the complement system's involvement in pulmonary hypertension, along with autoimmune conditions, and the reliance on the complement system for vascular remodeling processes of the pulmonary artery. Furthermore, circadian rhythmicity is highlighted as the disrupted homeostatic mechanism in the inflammatory consequences in the vascular remodeling within the pulmonary arteries, which could potentially open new therapeutic cues. The current treatment options for pulmonary hypertension are discussed with clinical trials using complement inhibitors and potential therapeutic targets that impact immune cell functions and complement activation, which could alleviate symptoms and block the progression of the disease. Further research on complement's involvement in interstitial lung diseases and pulmonary hypertension could prove beneficial for our understanding of these various diseases and potential treatment options to prevent vascular remodeling of the pulmonary arteries.
Collapse
Affiliation(s)
- Hunter DeVaughn
- Center for Metabolic and Degenerative Diseases, The Brown Foundation Institute of Molecular Medicine for Prevention of Human Diseases, UTHealth-McGovern Medical School, Houston, TX 77030, USA; (H.D.); (H.E.R.); (A.S.); (P.K.V.)
- Center for Immunology and Autoimmune Diseases, The Brown Foundation Institute of Molecular Medicine for Prevention of Human Diseases, UTHealth-McGovern Medical School, Houston, TX 77030, USA
| | - Haydn E. Rich
- Center for Metabolic and Degenerative Diseases, The Brown Foundation Institute of Molecular Medicine for Prevention of Human Diseases, UTHealth-McGovern Medical School, Houston, TX 77030, USA; (H.D.); (H.E.R.); (A.S.); (P.K.V.)
| | - Anthony Shadid
- Center for Metabolic and Degenerative Diseases, The Brown Foundation Institute of Molecular Medicine for Prevention of Human Diseases, UTHealth-McGovern Medical School, Houston, TX 77030, USA; (H.D.); (H.E.R.); (A.S.); (P.K.V.)
| | - Priyanka K. Vaidya
- Center for Metabolic and Degenerative Diseases, The Brown Foundation Institute of Molecular Medicine for Prevention of Human Diseases, UTHealth-McGovern Medical School, Houston, TX 77030, USA; (H.D.); (H.E.R.); (A.S.); (P.K.V.)
| | - Marie-Francoise Doursout
- Department of Anesthesiology, Critical Care and Pain Medicine, UTHealth-McGovern Medical School, Houston, TX 77030, USA;
| | - Pooja Shivshankar
- Center for Metabolic and Degenerative Diseases, The Brown Foundation Institute of Molecular Medicine for Prevention of Human Diseases, UTHealth-McGovern Medical School, Houston, TX 77030, USA; (H.D.); (H.E.R.); (A.S.); (P.K.V.)
- Center for Immunology and Autoimmune Diseases, The Brown Foundation Institute of Molecular Medicine for Prevention of Human Diseases, UTHealth-McGovern Medical School, Houston, TX 77030, USA
| |
Collapse
|
15
|
Sandner P, Follmann M, Becker-Pelster E, Hahn MG, Meier C, Freitas C, Roessig L, Stasch JP. Soluble GC stimulators and activators: Past, present and future. Br J Pharmacol 2024; 181:4130-4151. [PMID: 34600441 DOI: 10.1111/bph.15698] [Citation(s) in RCA: 48] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 08/30/2021] [Indexed: 12/20/2022] Open
Abstract
The discovery of soluble GC (sGC) stimulators and sGC activators provided valuable tools to elucidate NO-sGC signalling and opened novel pharmacological opportunities for cardiovascular indications and beyond. The first-in-class sGC stimulator riociguat was approved for pulmonary hypertension in 2013 and vericiguat very recently for heart failure. sGC stimulators enhance sGC activity independent of NO and also act synergistically with endogenous NO. The sGC activators specifically bind to, and activate, the oxidised haem-free form of sGC. Substantial research efforts improved on the first-generation sGC activators such as cinaciguat, culminating in the discovery of runcaciguat, currently in clinical Phase II trials for chronic kidney disease and diabetic retinopathy. Here, we highlight the discovery and development of sGC stimulators and sGC activators, their unique modes of action, their preclinical characteristics and the clinical studies. In the future, we expect to see more sGC agonists in new indications, reflecting their unique therapeutic potential.
Collapse
Affiliation(s)
- Peter Sandner
- Pharmaceuticals Research & Development, Bayer AG, Wuppertal, Germany
- Institute of Pharmacology, Hannover Medical School, Hanover, Germany
| | - Markus Follmann
- Pharmaceuticals Research & Development, Bayer AG, Wuppertal, Germany
| | | | - Michael G Hahn
- Pharmaceuticals Research & Development, Bayer AG, Wuppertal, Germany
| | - Christian Meier
- Pharmaceuticals Medical Affairs and Pharmacovigilance, Bayer AG, Berlin, Germany
| | - Cecilia Freitas
- Pharmaceuticals Research & Development, Bayer AG, Wuppertal, Germany
| | - Lothar Roessig
- Pharmaceuticals Research & Development, Bayer AG, Wuppertal, Germany
| | - Johannes-Peter Stasch
- Pharmaceuticals Research & Development, Bayer AG, Wuppertal, Germany
- Institute of Pharmacy, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| |
Collapse
|
16
|
Moore SM, Gawron J, Stevens M, Marziali LN, Buys ES, Milne GT, Feltri ML, VerPlank JJS. Pharmacologically increasing cGMP improves proteostasis and reduces neuropathy in mouse models of CMT1. Cell Mol Life Sci 2024; 81:434. [PMID: 39400753 PMCID: PMC11473742 DOI: 10.1007/s00018-024-05463-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 08/27/2024] [Accepted: 09/23/2024] [Indexed: 10/15/2024]
Abstract
Increasing cyclic GMP activates 26S proteasomes via phosphorylation by Protein Kinase G and stimulates the intracellular degradation of misfolded proteins. Therefore, agents that raise cGMP may be useful therapeutics against neurodegenerative diseases and other diseases in which protein degradation is reduced and misfolded proteins accumulate, including Charcot Marie Tooth 1A and 1B peripheral neuropathies, for which there are no treatments. Here we increased cGMP in the S63del mouse model of CMT1B by treating for three weeks with either the phosphodiesterase 5 inhibitor tadalafil, or the brain-penetrant soluble guanylyl cyclase stimulator CYR119. Both molecules activated proteasomes in the affected peripheral nerves, reduced polyubiquitinated proteins, and improved myelin thickness and nerve conduction. CYR119 increased cGMP more than tadalafil in the peripheral nerves of S63del mice and elicited greater biochemical and functional improvements. To determine whether raising cGMP could be beneficial in other neuropathies, we first showed that polyubiquitinated proteins and the disease-causing protein accumulate in the sciatic nerves of the C3 mouse model of CMT1A. Treatment of these mice with CYR119 reduced the levels of polyubiquitinated proteins and the disease-causing protein, presumably by increasing their degradation, and improved myelination, nerve conduction, and motor coordination. Thus, pharmacological agents that increase cGMP are promising treatments for CMT1 neuropathies and may be useful against other proteotoxic and neurodegenerative diseases.
Collapse
Affiliation(s)
- Seth M Moore
- Department of Biochemistry, Institute for Myelin and Glia Exploration, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, 14203, USA
| | - Joseph Gawron
- Department of Biochemistry, Institute for Myelin and Glia Exploration, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, 14203, USA
| | - Mckayla Stevens
- Department of Anatomy, Physiology, and Genetics, F. Edward Hebert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA
| | - Leandro N Marziali
- Department of Biochemistry, Institute for Myelin and Glia Exploration, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, 14203, USA
| | - Emmanuel S Buys
- Cyclerion Therapeutics, 245 First Street Riverview II, 18th floor, Cambridge, MA, 02142, USA
| | - G Todd Milne
- Cyclerion Therapeutics, 245 First Street Riverview II, 18th floor, Cambridge, MA, 02142, USA
| | - Maria Laura Feltri
- Department of Biochemistry, Institute for Myelin and Glia Exploration, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, 14203, USA
- IRCCS Neurological institute 'Carlo Besta', Milano, Italy
- Department of Medical Biotechnology and Translational Medicine, Universita' degli Studi di Milano, Milano, Italy
| | - Jordan J S VerPlank
- Department of Biochemistry, Institute for Myelin and Glia Exploration, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, 14203, USA.
- Department of Anatomy, Physiology, and Genetics, F. Edward Hebert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA.
| |
Collapse
|
17
|
Wong D, Seitz F, Bauer V, Giessmann T, Schulze F. Safety, tolerability, pharmacokinetics, and pharmacodynamics of BI 685509, a soluble guanylyl cyclase activator, in healthy volunteers: Results from two randomized controlled trials. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:8101-8116. [PMID: 38789635 PMCID: PMC11449976 DOI: 10.1007/s00210-024-03165-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 05/13/2024] [Indexed: 05/26/2024]
Abstract
This study evaluated the safety, tolerability, pharmacokinetics, and pharmacodynamics of BI 685509 after oral single rising doses (SRDs) or multiple rising doses (MRDs) in healthy volunteers. In the SRD trial (NCT02694354; February 29, 2016), within each of the three dose groups (DGs), six subjects received BI 685509 (1.0, 2.5, or 5.0 mg) and two received placebo (N = 24). In the MRD trial (NCT03116906; April 17, 2017), within each of the five DGs, nine subjects received BI 685509 (uptitrated to 1 mg once daily [qd; DG1], 2.5 mg twice daily [DG2], 5.0 mg qd [DG3]; 3.0 mg three times daily [tid; DG4] or 4.0 mg tid [DG5]) and three received placebo, for 14-17 days (N = 60). In the SRD trial, 7/24 subjects (29.2%) had ≥ 1 adverse event (AE), most frequently orthostatic dysregulation (n = 4). In the MRD trial, 26/45 subjects (57.8%) receiving BI 685509 had ≥ 1 AE, most frequently orthostatic dysregulation and fatigue (each n = 12). Tolerance development led to a marked decrease in orthostatic dysregulation events (DG3). BI 685509 was rapidly absorbed after oral administration, and exposure increased in a dose-proportional manner after single doses. Multiple dosing resulted in near-dose-proportional increase in exposure and limited accumulation. BI 685509 pharmacokinetics appeared linear with time; steady state occurred 3-5 days after each multiple-dosing period. Increased plasma cyclic guanosine monophosphate and decreased blood pressure followed by a compensatory increase in heart rate indicated target engagement. BI 685509 was generally well tolerated; orthostatic dysregulation may be appropriately countered by careful uptitration.
Collapse
Affiliation(s)
- Diane Wong
- Translational Medicine and Clinical Pharmacology, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, CT, 06877, USA.
| | | | - Verena Bauer
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an Der Riss, Germany
| | - Thomas Giessmann
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an Der Riss, Germany
| | | |
Collapse
|
18
|
Chakrabarty B, Winder M, Kanai AJ, Hashitani H, Drake M, Abrams P, Fry CH. Nitric oxide signaling pathways in the normal and pathological bladder: Do they provide new pharmacological pathways?-ICI-RS 2023. Neurourol Urodyn 2024; 43:1344-1352. [PMID: 37902298 DOI: 10.1002/nau.25321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 10/22/2023] [Indexed: 10/31/2023]
Abstract
AIMS The nitric oxide (NO•)/soluble guanylate cyclase/cyclic-GMP (cGMP) signaling pathway is ubiquitous and regulates several functions in physiological systems as diverse as the vascular, nervous, and renal systems. However, its roles in determining normal and abnormal lower urinary tract functions are unclear. The aim was to identify potential therapeutic targets associated with this pathway to manage lower urinary tract functional disorders. METHODS This review summarizes a workshop held under the auspices of ICI-RS with a view to address these questions. RESULTS Four areas were addressed: NO• signaling to regulate neurotransmitter release to detrusor smooth muscle; its potential dual roles in alleviating and exacerbating inflammatory pathways; its ability to act as an antifibrotic mediator; and the control by nitrergic nerves of lower urinary tract vascular dynamics and the contractile performance of muscular regions of the bladder wall. Central to much of the discussion was the role of the NO• receptor, soluble guanylate cyclase (sGC) in regulating the generation of the enzyme product, the second messenger cGMP. The redox state of sGC is crucial in determining its enzymic activity and the role of a class of novel agents, sGC activators, to optimize activity and to potentially alleviate the consequences of lower urinary tract disorders was highlighted. In addition, the consequences of a functional relationship between nitrergic and sympathetic nerves to regulate vascular dynamics was discussed. CONCLUSIONS Several potential NO•-dependent drug targets in the lower urinary tract were identified that provide the basis for future research and translation to clinical trials.
Collapse
Affiliation(s)
- Basu Chakrabarty
- School of Physiology, Pharmacology & Neuroscience, University of Bristol, Bristol, UK
| | - Michael Winder
- Department of Pharmacology, University of Gothenburg, Gothenburg, Sweden
| | - Anthony J Kanai
- Departments of Medicine and Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Hikaru Hashitani
- Department of Cell Physiology, Nagoya City University, Nagoya, Japan
| | - Marcus Drake
- Department of Surgery and Cancer, Hammersmith Hospital, London, UK
| | - Paul Abrams
- Bristol Urological Institute, North Bristol NHS Trust, Bristol, UK
| | - Christopher H Fry
- School of Physiology, Pharmacology & Neuroscience, University of Bristol, Bristol, UK
| |
Collapse
|
19
|
Pereira DA, Calmasini FB, Costa FF, Burnett AL, Silva FH. Nitric Oxide Resistance in Priapism Associated with Sickle Cell Disease: Mechanisms, Therapeutic Challenges, and Future Directions. J Pharmacol Exp Ther 2024; 390:203-212. [PMID: 38262744 DOI: 10.1124/jpet.123.001962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 12/07/2023] [Accepted: 01/05/2024] [Indexed: 01/25/2024] Open
Abstract
Patients with sickle cell disease (SCD) display priapism, a prolonged penile erection in the absence of sexual arousal. The current pharmacological treatments for SCD-associated priapism are limited and focused on acute interventions rather than prevention. Thus, there is an urgent need for new drug targets and preventive pharmacological therapies for this condition. This review focuses on the molecular mechanisms linked to the dysfunction of the NO-cyclic guanosine monophosphate (cGMP)-phosphodiesterase type 5 (PDE5) pathway implicated in SCD-associated priapism. In murine models of SCD, reduced nitric oxide (NO)-cGMP bioavailability in the corpus cavernosum is associated with elevated plasma hemoglobin levels, increased reactive oxygen species levels that inactive NO, and testosterone deficiency that leads to endothelial nitric oxide synthase downregulation. We discuss the consequences of the reduced cGMP-dependent PDE5 activity in response to these molecular changes, highlighting it as the primary pathophysiological mechanism leading to excessive corpus cavernosum relaxation, culminating in priapism. We also further discuss the impact of intravascular hemolysis on therapeutic approaches, present current pharmacological strategies targeting the NO-cGMP-PDE5 pathway in the penis, and identify potential pharmacological targets for future priapism therapies. In men with SCD and priapism, PDE5 inhibitor therapy and testosterone replacement have shown promising results. Recent preclinical research reported the beneficial effect of treatment with haptoglobin and NO donors. SIGNIFICANCE STATEMENT: This review discusses the molecular changes that reduce NO-cGMP bioavailability in the penis in SCD and highlights pharmacological targets and therapeutic strategies for the treatment of priapism, including PDE5 inhibitors, hormonal modulators, NO donors, hydroxyurea, soluble guanylate cyclase stimulators, haptoglobin, hemopexin, and antioxidants.
Collapse
Affiliation(s)
- Dalila Andrade Pereira
- Laboratory of Pharmacology, São Francisco University Medical School, Bragança Paulista, SP, Brazil (D.A.P., F.H.S.); Universidade Federal de São Paulo, Escola Paulista de Medicina, Department of Pharmacology, São Paulo, SP, Brazil (F.B.C.); Hematology and Hemotherapy Center, University of Campinas, Campinas, SP, Brazil (F.F.C.); and The James Buchanan Brady Urological Institute and Department of Urology, The Johns Hopkins School of Medicine, Baltimore, Maryland (A.L.B.)
| | - Fabiano Beraldi Calmasini
- Laboratory of Pharmacology, São Francisco University Medical School, Bragança Paulista, SP, Brazil (D.A.P., F.H.S.); Universidade Federal de São Paulo, Escola Paulista de Medicina, Department of Pharmacology, São Paulo, SP, Brazil (F.B.C.); Hematology and Hemotherapy Center, University of Campinas, Campinas, SP, Brazil (F.F.C.); and The James Buchanan Brady Urological Institute and Department of Urology, The Johns Hopkins School of Medicine, Baltimore, Maryland (A.L.B.)
| | - Fernando Ferreira Costa
- Laboratory of Pharmacology, São Francisco University Medical School, Bragança Paulista, SP, Brazil (D.A.P., F.H.S.); Universidade Federal de São Paulo, Escola Paulista de Medicina, Department of Pharmacology, São Paulo, SP, Brazil (F.B.C.); Hematology and Hemotherapy Center, University of Campinas, Campinas, SP, Brazil (F.F.C.); and The James Buchanan Brady Urological Institute and Department of Urology, The Johns Hopkins School of Medicine, Baltimore, Maryland (A.L.B.)
| | - Arthur L Burnett
- Laboratory of Pharmacology, São Francisco University Medical School, Bragança Paulista, SP, Brazil (D.A.P., F.H.S.); Universidade Federal de São Paulo, Escola Paulista de Medicina, Department of Pharmacology, São Paulo, SP, Brazil (F.B.C.); Hematology and Hemotherapy Center, University of Campinas, Campinas, SP, Brazil (F.F.C.); and The James Buchanan Brady Urological Institute and Department of Urology, The Johns Hopkins School of Medicine, Baltimore, Maryland (A.L.B.)
| | - Fábio Henrique Silva
- Laboratory of Pharmacology, São Francisco University Medical School, Bragança Paulista, SP, Brazil (D.A.P., F.H.S.); Universidade Federal de São Paulo, Escola Paulista de Medicina, Department of Pharmacology, São Paulo, SP, Brazil (F.B.C.); Hematology and Hemotherapy Center, University of Campinas, Campinas, SP, Brazil (F.F.C.); and The James Buchanan Brady Urological Institute and Department of Urology, The Johns Hopkins School of Medicine, Baltimore, Maryland (A.L.B.)
| |
Collapse
|
20
|
Xiang Y, Naik S, Zhao L, Shi J, Ke H. Emerging phosphodiesterase inhibitors for treatment of neurodegenerative diseases. Med Res Rev 2024; 44:1404-1445. [PMID: 38279990 DOI: 10.1002/med.22017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 12/13/2023] [Accepted: 01/09/2024] [Indexed: 01/29/2024]
Abstract
Neurodegenerative diseases (NDs) cause progressive loss of neuron structure and ultimately lead to neuronal cell death. Since the available drugs show only limited symptomatic relief, NDs are currently considered as incurable. This review will illustrate the principal roles of the signaling systems of cyclic adenosine and guanosine 3',5'-monophosphates (cAMP and cGMP) in the neuronal functions, and summarize expression/activity changes of the associated enzymes in the ND patients, including cyclases, protein kinases, and phosphodiesterases (PDEs). As the sole enzymes hydrolyzing cAMP and cGMP, PDEs are logical targets for modification of neurodegeneration. We will focus on PDE inhibitors and their potentials as disease-modifying therapeutics for the treatment of Alzheimer's disease, Parkinson's disease, and Huntington's disease. For the overlapped but distinct contributions of cAMP and cGMP to NDs, we hypothesize that dual PDE inhibitors, which simultaneously regulate both cAMP and cGMP signaling pathways, may have complementary and synergistic effects on modifying neurodegeneration and thus represent a new direction on the discovery of ND drugs.
Collapse
Affiliation(s)
- Yu Xiang
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Swapna Naik
- Department of Pharmacology, Yale Cancer Biology Institute, Yale University, West Haven, Connecticut, USA
| | - Liyun Zhao
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Jianyou Shi
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Hengming Ke
- Department of Biochemistry and Biophysics, The University of North Carolina, Chapel Hill, North Carolina, USA
| |
Collapse
|
21
|
Jain H, Goyal A, Khan AT, Khan NU, Jain J, Chopra S, Sulaiman SA, Reddy MM, Patel K, Khullar K, Daoud M, Sohail AH. Insights into calcific aortic valve stenosis: a comprehensive overview of the disease and advancing treatment strategies. Ann Med Surg (Lond) 2024; 86:3577-3590. [PMID: 38846838 PMCID: PMC11152847 DOI: 10.1097/ms9.0000000000002106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Accepted: 04/14/2024] [Indexed: 06/09/2024] Open
Abstract
Aortic valve stenosis is a disease characterized by thickening and narrowing of the aortic valve (AV), most commonly due to calcification, which leads to left ventricular outflow obstruction called calcific aortic valve disease (CAVD). CAVD presents as a progressive clinical syndrome with cardiorespiratory symptoms, often with rapid deterioration. The modern-day pathophysiology of CAVD involves a complex interplay of genetic factors, chronic inflammation, lipid deposition, and valve calcification, with early CAVD stages resembling atherosclerosis. Various imaging modalities have been used to evaluate CAVD, with a recent trend of using advanced imaging to measure numerous AV parameters, such as peak jet velocity. Significant improvements in mortality have been achieved with transcatheter AV repair, but numerous therapeutics and modalities are being researched to delay the progression of CAVD. This article aims to provide a comprehensive review of CAVD, explore recent developments, and provide insights into future treatments with various novel modalities.
Collapse
Affiliation(s)
- Hritvik Jain
- Department of Internal Medicine, All India Institute of Medical Sciences (AIIMS), Jodhpur
| | - Aman Goyal
- Department of Internal Medicine, Seth Gordhandas Sunderdas (GS) Medical College and King Edward Memorial (KEM) Hospital, Mumbai
| | | | - Noor U. Khan
- Department of Public Health, Health Services Academy, Islamabad, Pakistan
| | - Jyoti Jain
- Department of Internal Medicine, All India Institute of Medical Sciences (AIIMS), Jodhpur
| | - Shrey Chopra
- Department of Internal Medicine, University College of Medical Sciences and Guru Teg Bahadur Hospital, Delhi
| | | | | | - Kush Patel
- Department of Internal Medicine, Baroda Medical College, Gujarat
| | - Kaarvi Khullar
- Department of Internal Medicine, Government Medical College and Hospital, Gondia, Maharashtra, India
| | - Mohamed Daoud
- Department of Internal Medicine, Bogomolets National Medical University, Kyiv, Ukraine
| | - Amir H. Sohail
- Department of Surgery, University of New Mexico Health Sciences, Albuquerque, New Mexico, USA
| |
Collapse
|
22
|
Kintos DP, Salagiannis K, Sgouros A, Nikolaropoulos SS, Topouzis S, Fousteris MA. Identification of new multi-substituted 1H-pyrazolo[3,4-c]pyridin-7(6H)-ones as soluble guanylyl cyclase (sGC) stimulators with vasoprotective and anti-inflammatory activities. Bioorg Chem 2024; 144:107170. [PMID: 38335755 DOI: 10.1016/j.bioorg.2024.107170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 01/24/2024] [Accepted: 01/30/2024] [Indexed: 02/12/2024]
Abstract
Herein, we describe the rational design, synthesis and in vitro functional characterization of new heme-dependent, direct soluble guanylyl cyclase (sGC) agonists. These new compounds bear a 1H-pyrazolo[3,4-c]pyridin-7(6H)-one skeleton, modified to enable efficient sGC binding and stimulation. To gain insights into structure-activity relationships, the N6-alkylation of the skeleton was explored, while a pyrimidine ring, substituted with various C5'-polar groups, was installed at position C3. Among the newly synthesized 1H-pyrazolo[3,4-c]pyridin-7(6H)-ones, derivatives 14b, 15b and 16a display characteristic features of sGC "stimulators" in A7r5 vascular smooth muscle cells in vitro. They strongly synergize with the NO donor, sodium nitroprusside (SNP) in inducing cGMP generation in a manner that requires the presence of a reduced heme moiety associated with sGC, and elevate the cGMP-responsive phosphorylation of the protein VASP at Ser239. In line with their sGC stimulating capacity, docking calculations of derivatives 16a, 15(a-c) on a cryo-EM structure of human sGC (hsGC) in an ΝΟ-activated state indicated the implication of 1H-pyrazolo[3,4-c]pyridin-7(6H)-one skeleton in efficient bonding interactions with the recently identified region that binds known sGC stimulators, while the presence of either a N6-H or N6-methyl group pointed to enhanced binding affinity. Moreover, the in vitro functional effects of our newly identified sGC stimulators were compatible with a beneficial role in vascular homeostasis. Specifically, derivative 14b reduced A7r5 cell proliferation, while 16a dampened the expression of adhesion molecules ICAM-1 and P/E-Selectin in Human Umbilical Vein Endothelial Cells (HUVECs), as well as the subsequent adhesion of U937 leukocytes to the HUVECs, triggered by tumor necrosis factor alpha (TNF-α) or interleukin-1 beta (IL-1β). The fact that these compounds elevate cGMP only in the presence of NO may indicate a novel way of interaction with the enzyme and may make them less prone than other direct sGC agonists to induce characteristic hypotension in vivo.
Collapse
Affiliation(s)
| | - Konstantinos Salagiannis
- Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Patras, GR-26500, Greece
| | - Antonis Sgouros
- Laboratory of Medicinal Chemistry, Department of Pharmacy, University of Patras, Patras, GR-26500, Greece
| | - Sotiris S Nikolaropoulos
- Laboratory of Medicinal Chemistry, Department of Pharmacy, University of Patras, Patras, GR-26500, Greece
| | - Stavros Topouzis
- Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Patras, GR-26500, Greece.
| | - Manolis A Fousteris
- Laboratory of Medicinal Chemistry, Department of Pharmacy, University of Patras, Patras, GR-26500, Greece.
| |
Collapse
|
23
|
Pereira DA, Silveira THR, Calmasini FB, Silva FH. Soluble guanylate cyclase stimulators and activators: new horizons in the treatment of priapism associated with sickle cell disease. Front Pharmacol 2024; 15:1357176. [PMID: 38384294 PMCID: PMC10879333 DOI: 10.3389/fphar.2024.1357176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Accepted: 01/25/2024] [Indexed: 02/23/2024] Open
Abstract
Priapism, defined as a prolonged and often painful penile erection occurring without sexual stimulation or desire, is a common complication in sickle cell disease (SCD), affecting up to 48% of male patients. This condition presents significant clinical challenges and can lead to erectile dysfunction if not properly managed. Current pharmacological treatments for SCD-related priapism are primarily reactive rather than preventative, highlighting a gap in effective medical intervention strategies. A critical factor in developing priapism is the reduced basal bioavailability of nitric oxide (NO) and cyclic guanosine monophosphate (cGMP) in erectile tissues. New prevention strategies should ideally target the underlying pathophysiology of the disease. Compounds that stimulate and activate soluble guanylate cyclase (sGC) emerge as potential therapeutic candidates since these compounds have the property of inducing cGMP production by sGC. This review explores the potential of sGC stimulators and activators in treating priapism associated with SCD. We discuss the advantages of these agents in the face of the challenging pathophysiology of SCD. Additionally, the review underscores the impact of intravascular hemolysis and oxidative stress on priapism pathophysiology in SCD, areas in which sGC stimulators and activators may also have beneficial therapeutic effects.
Collapse
Affiliation(s)
- Dalila Andrade Pereira
- Laboratory of Pharmacology, São Francisco University Medical School, Bragança Paulista, SP, Brazil
| | | | - Fabiano Beraldi Calmasini
- Department of Pharmacology, Universidade Federal de São Paulo, Escola Paulista de Medicina, São Paulo, SP, Brazil
| | - Fábio Henrique Silva
- Laboratory of Pharmacology, São Francisco University Medical School, Bragança Paulista, SP, Brazil
| |
Collapse
|
24
|
Montfort WR. Per-ARNT-Sim Domains in Nitric Oxide Signaling by Soluble Guanylyl Cyclase. J Mol Biol 2024; 436:168235. [PMID: 37572934 PMCID: PMC10858291 DOI: 10.1016/j.jmb.2023.168235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/01/2023] [Accepted: 08/04/2023] [Indexed: 08/14/2023]
Abstract
Nitric oxide (NO) regulates large swaths of animal physiology including wound healing, vasodilation, memory formation, odor detection, sexual function, and response to infectious disease. The primary NO receptor is soluble guanyly/guanylate cyclase (sGC), a dimeric protein of ∼150 kDa that detects NO through a ferrous heme, leading to a large change in conformation and enhanced production of cGMP from GTP. In humans, loss of sGC function contributes to multiple disease states, including cardiovascular disease and cancer, and is the target of a new class of drugs, sGC stimulators, now in clinical use. sGC evolved through the fusion of four ancient domains, a heme nitric oxide / oxygen (H-NOX) domain, a Per-ARNT-Sim (PAS) domain, a coiled coil, and a cyclase domain, with catalysis occurring at the interface of the two cyclase domains. In animals, the predominant dimer is the α1β1 heterodimer, with the α1 subunit formed through gene duplication of the β1 subunit. The PAS domain provides an extensive dimer interface that remains unchanged during sGC activation, acting as a core anchor. A large cleft formed at the PAS-PAS dimer interface tightly binds the N-terminal end of the coiled coil, keeping this region intact and unchanged while the rest of the coiled coil repacks, and the other domains reposition. This interface buries ∼3000 Å2 of monomer surface and includes highly conserved apolar and hydrogen bonding residues. Herein, we discuss the evolutionary history of sGC, describe the role of PAS domains in sGC function, and explore the regulatory factors affecting sGC function.
Collapse
Affiliation(s)
- William R Montfort
- Department of Chemistry & Biochemistry, University of Arizona, Tucson, AZ 85721, USA.
| |
Collapse
|
25
|
Benza RL, Grünig E, Sandner P, Stasch JP, Simonneau G. The nitric oxide-soluble guanylate cyclase-cGMP pathway in pulmonary hypertension: from PDE5 to soluble guanylate cyclase. Eur Respir Rev 2024; 33:230183. [PMID: 38508664 PMCID: PMC10957071 DOI: 10.1183/16000617.0183-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 01/18/2024] [Indexed: 03/22/2024] Open
Abstract
The nitric oxide (NO)-soluble guanylate cyclase (sGC)-cyclic guanosine monophosphate (cGMP) pathway plays a key role in the pathogenesis of pulmonary hypertension (PH). Targeted treatments include phosphodiesterase type 5 inhibitors (PDE5i) and sGC stimulators. The sGC stimulator riociguat is approved for the treatment of pulmonary arterial hypertension (PAH) and chronic thromboembolic pulmonary hypertension (CTEPH). sGC stimulators have a dual mechanism of action, enhancing the sGC response to endogenous NO and directly stimulating sGC, independent of NO. This increase in cGMP production via a dual mechanism differs from PDE5i, which protects cGMP from degradation by PDE5, rather than increasing its production. sGC stimulators may therefore have the potential to increase cGMP levels under conditions of NO depletion that could limit the effectiveness of PDE5i. Such differences in mode of action between sGC stimulators and PDE5i could lead to differences in treatment efficacy between the classes. In addition to vascular effects, sGC stimulators have the potential to reduce inflammation, angiogenesis, fibrosis and right ventricular hypertrophy and remodelling. In this review we describe the evolution of treatments targeting the NO-sGC-cGMP pathway, with a focus on PH.
Collapse
Affiliation(s)
| | - Ekkehard Grünig
- Pulmonary Hypertension Unit, Thoraxklinik at Heidelberg University Hospital Heidelberg, Heidelberg, Germany
| | - Peter Sandner
- Bayer AG, Wuppertal, Germany
- Institute of Pharmacology, Hannover Medical School, Hannover, Germany
| | - Johannes-Peter Stasch
- Bayer AG, Wuppertal, Germany
- Institute of Pharmacy, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| | - Gérald Simonneau
- Centre de Référence de l'Hypertension Pulmonaire Sévère, CHU Kremlin Bicêtre, Kremlin Bicêtre, France
| |
Collapse
|
26
|
Yin Q, Zheng X, Song Y, Wu L, Li L, Tong R, Han L, Bian Y. Decoding signaling mechanisms: unraveling the targets of guanylate cyclase agonists in cardiovascular and digestive diseases. Front Pharmacol 2023; 14:1272073. [PMID: 38186653 PMCID: PMC10771398 DOI: 10.3389/fphar.2023.1272073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 12/08/2023] [Indexed: 01/09/2024] Open
Abstract
Soluble guanylate cyclase agonists and guanylate cyclase C agonists are two popular drugs for diseases of the cardiovascular system and digestive systems. The common denominator in these conditions is the potential therapeutic target of guanylate cyclase. Thanks to in-depth explorations of their underlying signaling mechanisms, the targets of these drugs are becoming clearer. This review explains the recent research progress regarding potential drugs in this class by introducing representative drugs and current findings on them.
Collapse
Affiliation(s)
- Qinan Yin
- Department of Pharmacy, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Xingyue Zheng
- Department of Pharmacy, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yujie Song
- Department of Pharmacy, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Liuyun Wu
- Department of Pharmacy, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Lian Li
- Department of Pharmacy, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Rongsheng Tong
- Department of Pharmacy, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Lizhu Han
- Department of Pharmacy, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yuan Bian
- Department of Pharmacy, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
27
|
Lawitz EJ, Reiberger T, Schattenberg JM, Schoelch C, Coxson HO, Wong D, Ertle J. Safety and pharmacokinetics of BI 685509, a soluble guanylyl cyclase activator, in patients with cirrhosis: A randomized Phase Ib study. Hepatol Commun 2023; 7:e0276. [PMID: 37889522 PMCID: PMC10615399 DOI: 10.1097/hc9.0000000000000276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 07/29/2023] [Indexed: 10/28/2023] Open
Abstract
BACKGROUND Portal hypertension is a severe complication of cirrhosis. This Phase Ib study (NCT03842761) assessed the safety, tolerability, and pharmacokinetics of soluble guanylyl cyclase activator BI 685509 in patients with mild or moderate hepatic impairment (Child-Pugh [CP] A or B cirrhosis) and healthy volunteers (HVs). METHODS In this single-center, randomized, placebo-controlled study, patients received BI 685509 (maximum doses: 1, 2, or 3 mg, twice daily [BID]) or placebo for 28 days. HVs received one 0.5 mg dose of BI 685509 or placebo. RESULTS In total, 64 participants (CP-A, n=24; CP-B, n=25; HVs, n=15) were included; most commonly with NAFLD (36.7%), alcohol-associated (30.6%), or chronic viral hepatitis-related cirrhosis (28.6%). In patients with CP-A cirrhosis, drug-related adverse events (AEs) occurred in 5.6% of BI 685509-treated patients and 16.7% of placebo recipients. In patients with CP-B cirrhosis, drug-related AEs occurred in 26.3% of BI 685509-treated patients only. No serious AEs occurred in patients with CP-A cirrhosis; in patients with CP-B cirrhosis, serious AEs (not drug-related) occurred in 10.5% of BI 685509-treated patients and 16.7% of patients receiving placebo. BI 685509 was rapidly absorbed; exposure increased with dosage and was similar between etiologies and between patients with CP-A cirrhosis and patients with CP-A cirrhosis but lower in HVs. The mean percentage portal-systemic shunt fraction was measured in patients with CP-A cirrhosis and decreased at the end of treatment in the 2 mg BID (-11.2 ± 11.9%) and 3 mg BID (-14.0 ± 8.4%) BI 685509 dose groups, but not in the placebo group (+1.0 ± 27.3%). CONCLUSION BI 685509 was generally well tolerated, with 3 serious, not drug-related AEs reported in patients with CP-B cirrhosis. In patients with CP-A cirrhosis, portal-systemic shunt fraction in the exploratory efficacy analysis was reduced by 2 mg BID and 3 mg BID BI 685509.
Collapse
Affiliation(s)
- Eric J. Lawitz
- The Texas Liver Institute, University of Texas Health, San Antonio, Texas, USA
| | - Thomas Reiberger
- Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
- Christian Doppler Laboratory for Portal Hypertension and Liver Fibrosis, Medical University of Vienna, Vienna, Austria
| | - Jörn M. Schattenberg
- Metabolic Liver Research Program, I. Department of Medicine, University Medical Center Mainz, Mainz, Rhineland Palatinate, Germany
| | | | | | - Diane Wong
- Translational Medicine and Clinical Pharmacology, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut, USA
| | - Judith Ertle
- Boehringer Ingelheim International GmbH, Ingelheim am Rhein, Germany
| |
Collapse
|
28
|
Bykov VV, Bykova AV, Motov VS, Larchenko VV, Chernysheva GA, Smol'yakova VI, Aliev OI, Khazanov VA, Vengerovskii AI, Udut VV. Pharmacological Effects of a New Soluble Guanylate Cyclase Stimulator in Experimental Ischemic Stroke. Bull Exp Biol Med 2023; 175:749-752. [PMID: 37978152 DOI: 10.1007/s10517-023-05938-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Indexed: 11/19/2023]
Abstract
We studied the action of a new indolinone derivative GRS, acetylsalicylic acid (ASA), and their combination on platelet aggregation, vasodilatory endothelial function, neurological status, and cerebral infarction area in experimental focal cerebral ischemia/reperfusion in rats. GRS compound (10 mg/kg), ASA (10 mg/kg), and their combination in the same doses were administered orally once a day as a suspension in 1% starch solution over 5 days after pathology modeling. Sham-operated and control animals were administered 1% starch solution. On day 5 after pathology modeling, platelet aggregation and brain damage area were studied in a half of rats in each group, and the vasodilatory function of the endothelium was studied in the other half. Neurological deficit was assessed 4 h and 1, 3, and 5 days after pathology modeling. GRS compound and ASA equally effectively prevent platelet aggregation and the development of neurological deficit in rats. GRS compound restores the vasodilatory effects of the endothelium, but only ASA contributes to reduction of the cerebral infarction area. In case of combined administration, GRS and ASA do not exhibit synergy in their antiaggregant effect.
Collapse
Affiliation(s)
- V V Bykov
- IPHAR (Innovative Pharmacology Research, LLC), Tomsk, Russia.
- Siberian State Medical University, Ministry of Health of the Russian Federation, Tomsk, Russia.
| | - A V Bykova
- IPHAR (Innovative Pharmacology Research, LLC), Tomsk, Russia
| | - V S Motov
- IPHAR (Innovative Pharmacology Research, LLC), Tomsk, Russia
| | - V V Larchenko
- IPHAR (Innovative Pharmacology Research, LLC), Tomsk, Russia
- Siberian State Medical University, Ministry of Health of the Russian Federation, Tomsk, Russia
| | - G A Chernysheva
- E. D. Goldberg Research Institute of Pharmacology and Regenerative Medicine, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - V I Smol'yakova
- E. D. Goldberg Research Institute of Pharmacology and Regenerative Medicine, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - O I Aliev
- E. D. Goldberg Research Institute of Pharmacology and Regenerative Medicine, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - V A Khazanov
- IPHAR (Innovative Pharmacology Research, LLC), Tomsk, Russia
| | - A I Vengerovskii
- Siberian State Medical University, Ministry of Health of the Russian Federation, Tomsk, Russia
| | - V V Udut
- E. D. Goldberg Research Institute of Pharmacology and Regenerative Medicine, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| |
Collapse
|
29
|
Olivencia MA, Gil de Biedma-Elduayen L, Giménez-Gómez P, Barreira B, Fernández A, Angulo J, Colado MI, O'Shea E, Perez-Vizcaino F. Oxidized soluble guanylyl cyclase causes erectile dysfunction in alcoholic mice. Br J Pharmacol 2023; 180:2361-2376. [PMID: 37021655 DOI: 10.1111/bph.16087] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 03/13/2023] [Accepted: 04/03/2023] [Indexed: 04/07/2023] Open
Abstract
BACKGROUND AND PURPOSE Alcohol abuse has been associated with erectile dysfunction (ED), but the implicated molecular mechanisms are unresolved. This study analyses the role of alterations in soluble guanylyl cyclase (sGC) in ED. EXPERIMENTAL APPROACH ED was analysed in adult male C57BL/6J mice subjected to the Chronic Intermittent Ethanol (CIE) paradigm. Erectile function was assessed in anaesthetised mice in vivo by evaluating intracavernosal pressure (ICP) and in vitro in isolated mice corpora cavernosa (CC) mounted in a myograph. Protein expression and reactive oxygen species were analysed by western blot and dihydroethidium staining, respectively. KEY RESULTS In CIE mice, we observed a significant decrease in the relaxant response of the CC to stimulation of NO release from nitrergic nerves by electrical field stimulation, to NO release from endothelial cells by acetylcholine, to the PDE5 inhibitor sildenafil, and to the sGC stimulator riociguat. Conversely, the response to the sGC activator cinaciguat, whose action is independent of the oxidation state of sGC, was significantly enhanced in these CC. The responses to adenylyl cyclase stimulation with forskolin were unchanged. We found an increase in reactive oxygen species in the CC from CIE mice as well as an increase in CYP2E1 and NOX2 protein expression. In vivo pre-treatment with tempol prevented alcohol-induced erectile dysfunction. CONCLUSIONS AND IMPLICATIONS Our results demonstrate that alcoholic mice show ED in vitro and in vivo due to an alteration in the redox state of sGC and suggest that sGC activators may be effective in ED associated with alcoholism.
Collapse
Affiliation(s)
- Miguel A Olivencia
- Departamento de Farmacologia y Toxicologia, Facultad de Medicina, Universidad Complutense, Madrid, Spain
- CIBER Enfermedades Respiratorias, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Leticia Gil de Biedma-Elduayen
- Departamento de Farmacologia y Toxicologia, Facultad de Medicina, Universidad Complutense, Madrid, Spain
- Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid, Spain
- Red de Investigación en Atención Primaria de Adicciones del Instituto de Salud Carlos III, Madrid, Spain
- Instituto Universitario de Investigación Neuroquímica (IUIN), Facultad de Medicina, Universidad Complutense, Madrid, Spain
| | - Pablo Giménez-Gómez
- Departamento de Farmacologia y Toxicologia, Facultad de Medicina, Universidad Complutense, Madrid, Spain
- Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid, Spain
- Red de Investigación en Atención Primaria de Adicciones del Instituto de Salud Carlos III, Madrid, Spain
- Instituto Universitario de Investigación Neuroquímica (IUIN), Facultad de Medicina, Universidad Complutense, Madrid, Spain
| | - Bianca Barreira
- Departamento de Farmacologia y Toxicologia, Facultad de Medicina, Universidad Complutense, Madrid, Spain
- CIBER Enfermedades Respiratorias, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Argentina Fernández
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
- Servicio de Histología-Investigación, Unidad de Investigación Traslacional en Cardiología (IRYCIS-UFV), Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Javier Angulo
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
- Servicio de Histología-Investigación, Unidad de Investigación Traslacional en Cardiología (IRYCIS-UFV), Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Maria Isabel Colado
- Departamento de Farmacologia y Toxicologia, Facultad de Medicina, Universidad Complutense, Madrid, Spain
- Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid, Spain
- Red de Investigación en Atención Primaria de Adicciones del Instituto de Salud Carlos III, Madrid, Spain
- Instituto Universitario de Investigación Neuroquímica (IUIN), Facultad de Medicina, Universidad Complutense, Madrid, Spain
| | - Esther O'Shea
- Departamento de Farmacologia y Toxicologia, Facultad de Medicina, Universidad Complutense, Madrid, Spain
- Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid, Spain
- Red de Investigación en Atención Primaria de Adicciones del Instituto de Salud Carlos III, Madrid, Spain
- Instituto Universitario de Investigación Neuroquímica (IUIN), Facultad de Medicina, Universidad Complutense, Madrid, Spain
| | - Francisco Perez-Vizcaino
- Departamento de Farmacologia y Toxicologia, Facultad de Medicina, Universidad Complutense, Madrid, Spain
- CIBER Enfermedades Respiratorias, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| |
Collapse
|
30
|
Johnson S, Sommer N, Cox-Flaherty K, Weissmann N, Ventetuolo CE, Maron BA. Pulmonary Hypertension: A Contemporary Review. Am J Respir Crit Care Med 2023; 208:528-548. [PMID: 37450768 PMCID: PMC10492255 DOI: 10.1164/rccm.202302-0327so] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 07/14/2023] [Indexed: 07/18/2023] Open
Abstract
Major advances in pulmonary arterial hypertension, pulmonary hypertension (PH) associated with lung disease, and chronic thromboembolic PH cast new light on the pathogenetic mechanisms, epidemiology, diagnostic approach, and therapeutic armamentarium for pulmonary vascular disease. Here, we summarize key basic, translational, and clinical PH reports, emphasizing findings that build on current state-of-the-art research. This review includes cutting-edge progress in translational pulmonary vascular biology, with a guide to the diagnosis of patients in clinical practice, incorporating recent PH definition revisions that continue emphasis on early detection of disease. PH management is reviewed including an overview of the evolving considerations for the approach to treatment of PH in patients with cardiopulmonary comorbidities, as well as a discussion of the groundbreaking sotatercept data for the treatment of pulmonary arterial hypertension.
Collapse
Affiliation(s)
- Shelsey Johnson
- The Pulmonary Center, Division of Pulmonary, Allergy, Sleep and Critical Care, Boston University School of Medicine, Boston, Massachusetts
- Department of Pulmonary and Critical Care Medicine and
| | - Natascha Sommer
- Excellence Cluster Cardiopulmonary Institute, Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research, Justus Liebig University, Giessen, Germany
| | | | - Norbert Weissmann
- Excellence Cluster Cardiopulmonary Institute, Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research, Justus Liebig University, Giessen, Germany
| | - Corey E. Ventetuolo
- Department of Medicine and
- Department of Health Services, Policy and Practice, Brown University, Providence, Rhode Island
| | - Bradley A. Maron
- Division of Cardiovascular Medicine, Brigham and Women’s Hospital, and Harvard Medical School, Boston, Massachusetts
- Department of Cardiology and Department of Pulmonary, Allergy, Sleep, and Critical Care Medicine, VA Boston Healthcare System, Boston, Massachusetts
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland; and
- The University of Maryland-Institute for Health Computing, Bethesda, Maryland
| |
Collapse
|
31
|
Kraehling JR, Benardeau A, Schomber T, Popp L, Vienenkoetter J, Ellinger-Ziegelbauer H, Pavkovic M, Hartmann E, Siudak K, Freyberger A, Hagelschuer I, Mathar I, Hueser J, Hahn MG, Geiss V, Eitner F, Sandner P. The sGC Activator Runcaciguat Has Kidney Protective Effects and Prevents a Decline of Kidney Function in ZSF1 Rats. Int J Mol Sci 2023; 24:13226. [PMID: 37686032 PMCID: PMC10488129 DOI: 10.3390/ijms241713226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 08/18/2023] [Accepted: 08/23/2023] [Indexed: 09/10/2023] Open
Abstract
Chronic kidney disease (CKD) progression is associated with persisting oxidative stress, which impairs the NO-sGC-cGMP signaling cascade through the formation of oxidized and heme-free apo-sGC that cannot be activated by NO. Runcaciguat (BAY 1101042) is a novel, potent, and selective sGC activator that binds and activates oxidized and heme-free sGC and thereby restores NO-sGC-cGMP signaling under oxidative stress. Therefore, runcaciguat might represent a very effective treatment option for CKD/DKD. The potential kidney-protective effects of runcaciguat were investigated in ZSF1 rats as a model of CKD/DKD, characterized by hypertension, hyperglycemia, obesity, and insulin resistance. ZSF1 rats were treated daily orally for up to 12 weeks with runcaciguat (1, 3, 10 mg/kg/bid) or placebo. The study endpoints were proteinuria, kidney histopathology, plasma, urinary biomarkers of kidney damage, and gene expression profiling to gain information about relevant pathways affected by runcaciguat. Furthermore, oxidative stress was compared in the ZSF1 rat kidney with kidney samples from DKD patients. Within the duration of the 12-week treatment study, kidney function was significantly decreased in obese ZSF1 rats, indicated by a 20-fold increase in proteinuria, compared to lean ZSF1 rats. Runcaciguat dose-dependently and significantly attenuated the development of proteinuria in ZSF1 rats with reduced uPCR at the end of the study by -19%, -54%, and -70% at 1, 3, and 10 mg/kg/bid, respectively, compared to placebo treatment. Additionally, average blood glucose levels measured as HbA1C, triglycerides, and cholesterol were increased by five times, twenty times, and four times, respectively, in obese ZSF1 compared to lean rats. In obese ZSF1 rats, runcaciguat reduced HbA1c levels by -8%, -34%, and -76%, triglycerides by -42%, -55%, and -71%, and cholesterol by -16%, -17%, and -34%, at 1, 3, and 10 mg/kg/bid, respectively, compared to placebo. Concomitantly, runcaciguat also reduced kidney weights, morphological kidney damage, and urinary and plasma biomarkers of kidney damage. Beneficial effects were accompanied by changes in gene expression that indicate reduced fibrosis and inflammation and suggest improved endothelial stabilization. In summary, the sGC activator runcaciguat significantly prevented a decline in kidney function in a DKD rat model that mimics common comorbidities and conditions of oxidative stress of CKD patients. Thus, runcaciguat represents a promising treatment option for CKD patients, which is in line with recent phase 2 clinical study data, where runcaciguat showed promising efficacy in CKD patients (NCT04507061).
Collapse
Affiliation(s)
- Jan R. Kraehling
- Bayer AG, Research and Early Development, Pharma Research Center, 42096 Wuppertal, Germany
| | - Agnes Benardeau
- Bayer AG, Research and Early Development, Pharma Research Center, 42096 Wuppertal, Germany
- Novo Nordisk A/S, Global Drug Discovery, T1D-Kidney Disease, 2760 Måløv, Denmark
| | - Tibor Schomber
- Bayer AG, Research and Early Development, Pharma Research Center, 42096 Wuppertal, Germany
- Vincerx Pharma GmbH, 40789 Monheim, Germany
| | - Laura Popp
- Bayer AG, Research and Early Development, Pharma Research Center, 42096 Wuppertal, Germany
| | - Julia Vienenkoetter
- Bayer AG, Research and Early Development, Pharma Research Center, 42096 Wuppertal, Germany
| | | | - Mira Pavkovic
- Bayer AG, Research and Early Development, Pharma Research Center, 42096 Wuppertal, Germany
| | - Elke Hartmann
- Bayer AG, Research and Early Development, Pharma Research Center, 42096 Wuppertal, Germany
| | - Krystyna Siudak
- Bayer AG, Research and Early Development, Pharma Research Center, 42096 Wuppertal, Germany
| | - Alexius Freyberger
- Bayer AG, Research and Early Development, Pharma Research Center, 42096 Wuppertal, Germany
| | - Ina Hagelschuer
- Bayer AG, Research and Early Development, Pharma Research Center, 42096 Wuppertal, Germany
| | - Ilka Mathar
- Bayer AG, Research and Early Development, Pharma Research Center, 42096 Wuppertal, Germany
| | - Joerg Hueser
- Bayer AG, Research and Early Development, Pharma Research Center, 42096 Wuppertal, Germany
| | - Michael G. Hahn
- Bayer AG, Research and Early Development, Pharma Research Center, 42096 Wuppertal, Germany
| | - Volker Geiss
- Bayer AG, Research and Early Development, Pharma Research Center, 42096 Wuppertal, Germany
| | - Frank Eitner
- Bayer AG, Research and Early Development, Pharma Research Center, 42096 Wuppertal, Germany
- Division of Nephrology and Clinical Immunology, RWTH Aachen University, 52062 Aachen, Germany
| | - Peter Sandner
- Bayer AG, Research and Early Development, Pharma Research Center, 42096 Wuppertal, Germany
- Department of Pharmacology, Hannover Medical School, 30625 Hannover, Germany
| |
Collapse
|
32
|
Vaz-Salvador P, Adão R, Vasconcelos I, Leite-Moreira AF, Brás-Silva C. Heart Failure with Preserved Ejection Fraction: a Pharmacotherapeutic Update. Cardiovasc Drugs Ther 2023; 37:815-832. [PMID: 35098432 PMCID: PMC8801287 DOI: 10.1007/s10557-021-07306-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/17/2021] [Indexed: 02/06/2023]
Abstract
While guidelines for management of heart failure with reduced ejection fraction (HFrEF) are consensual and have led to improved survival, treatment options for heart failure with preserved ejection fraction (HFpEF) remain limited and aim primarily for symptom relief and improvement of quality of life. Due to the shortage of therapeutic options, several drugs have been investigated in multiple clinical trials. The majority of these trials have reported disappointing results and have suggested that HFpEF might not be as simply described by ejection fraction as previously though. In fact, HFpEF is a complex clinical syndrome with various comorbidities and overlapping distinct phenotypes that could benefit from personalized therapeutic approaches. This review summarizes the results from the most recent phase III clinical trials for HFpEF and the most promising drugs arising from phase II trials as well as the various challenges that are currently holding back the development of new pharmacotherapeutic options for these patients.
Collapse
Affiliation(s)
- Pedro Vaz-Salvador
- Department of Surgery and Physiology, Faculty of Medicine, Cardiovascular Research and Development Center - UnIC, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Rui Adão
- Department of Surgery and Physiology, Faculty of Medicine, Cardiovascular Research and Development Center - UnIC, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Inês Vasconcelos
- Department of Surgery and Physiology, Faculty of Medicine, Cardiovascular Research and Development Center - UnIC, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Adelino F. Leite-Moreira
- Department of Surgery and Physiology, Faculty of Medicine, Cardiovascular Research and Development Center - UnIC, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Carmen Brás-Silva
- Department of Surgery and Physiology, Faculty of Medicine, Cardiovascular Research and Development Center - UnIC, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
- Faculty of Nutrition and Food Sciences, University of Porto, Rua Do Campo Alegre, 823 4150-180 Porto, Portugal
| |
Collapse
|
33
|
Olivencia MA, Esposito E, Brancaleone V, Castaldo S, Cirino G, Pérez-Vizcaino F, Sorrentino R, d'Emmanuele di Villa Bianca R, Mitidieri E. Hydrogen sulfide regulates the redox state of soluble guanylate cyclase in CSE -/- mice corpus cavernosum microcirculation. Pharmacol Res 2023; 194:106834. [PMID: 37343646 DOI: 10.1016/j.phrs.2023.106834] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 05/29/2023] [Accepted: 06/19/2023] [Indexed: 06/23/2023]
Abstract
The corpus cavernosum (CC) is a highly vascularized tissue and represents an excellent example of microcirculation. Indeed, erectile dysfunction is considered an early index of cardiovascular disease. Hydrogen sulfide (H2S) at the vascular level is endogenously produced from L-cysteine mainly by the action of cystathionine-γ-lyase (CSE) and plays a role in CC vascular homeostasis. Here we have evaluated the involvement of the endogenous H2S in the regulation of the soluble guanylate cyclase (sCG) redox state. The lack of CSE-derived endogenous H2S, in CSE-/- mice, disrupted the eNOS/NO/sGC/PDE pathway. Indeed, the absence of CSE-derived endogenous H2S caused a significant reduction of the relaxant response to riociguat, an sGC redox-dependent stimulator. Conversely, the response to cinaciguat, an sGC redox-independent activator, was not modified. The relevance of the role played at the redox level of the endogenous H2S was confirmed by the findings that in CC harvested from CSE-/- mice there was a significant reduction of GCβ1 expression coupled with a decrease in CYP5R3, a reductase involved in the regulation of the redox state of sGC. These molecular changes driven by the lack of endogenous H2S translate into a significant reduction in cGMP levels. The replenishment of the lack of H2S with an H2S donor rescued the relaxant response to riociguat in CC of CSE-/- mice. In conclusion, the endogenous CSE-derived H2S plays a physiological key role in the regulation of the redox state of sGC in CC microcirculation.
Collapse
Affiliation(s)
- Miguel A Olivencia
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain; CIBER Enfermedades Respiratorias, Madrid, Spain; Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Erika Esposito
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | | | - Sigismondo Castaldo
- U.O.C. Ricerca Formazione & Cooperazione Internazionale, A.O.R.N." Antonio Cardarelli", Naples, Italy
| | - Giuseppe Cirino
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy; Interdepartmental Centre for Sexual Medicine, University of Naples Federico II, Naples, Italy
| | - Francisco Pérez-Vizcaino
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain; CIBER Enfermedades Respiratorias, Madrid, Spain; Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Raffaella Sorrentino
- Interdepartmental Centre for Sexual Medicine, University of Naples Federico II, Naples, Italy; Department of Molecular Medicine and Medical Biotechnology, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Roberta d'Emmanuele di Villa Bianca
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy; Interdepartmental Centre for Sexual Medicine, University of Naples Federico II, Naples, Italy.
| | - Emma Mitidieri
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| |
Collapse
|
34
|
Theofilis P, Vordoni A, Kalaitzidis RG. Novel therapeutic approaches in the management of chronic kidney disease: a narrative review. Postgrad Med 2023; 135:543-550. [PMID: 37401536 DOI: 10.1080/00325481.2023.2233492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 06/27/2023] [Indexed: 07/05/2023]
Abstract
Chronic kidney disease (CKD) remains a pathologic entity with constantly rising incidence and high rates of morbidity and mortality, which are associated with serious cardiovascular complications. Moreover, the incidence of end-stage renal disease tends to increase. The epidemiological trends of CKD warrant the development of novel therapeutic approaches aiming to prevent its development or retard its progression through the control of major risk factors: type 2 diabetes mellitus, arterial hypertension, and dyslipidemia. Contemporary therapeutics such as sodium-glucose cotransporter-2 inhibitors and second-generation mineralocorticoid receptor antagonists are utilized in this direction. Additionally, experimental and clinical studies present novel drug categories that could be employed in managing CKD, such as aldosterone synthesis inhibitors or activators guanylate cyclase, while the role of melatonin should be further tested in the clinical setting. Finally, in this patient population, the use of hypolipidemic agents may provide incremental benefits.
Collapse
Affiliation(s)
- Panagiotis Theofilis
- Center for Nephrology, "G. Papadakis" General Hospital of Nikaia-Piraeus "Ag. Panteleimon", Athens, Greece
| | - Aikaterini Vordoni
- Center for Nephrology, "G. Papadakis" General Hospital of Nikaia-Piraeus "Ag. Panteleimon", Athens, Greece
| | - Rigas G Kalaitzidis
- Center for Nephrology, "G. Papadakis" General Hospital of Nikaia-Piraeus "Ag. Panteleimon", Athens, Greece
| |
Collapse
|
35
|
Jones AK, Chen H, Ng KJ, Villalona J, McHugh M, Zeveleva S, Wilks J, Brilisauer K, Bretschneider T, Qian HS, Fryer RM. Soluble Guanylyl Cyclase Activator BI 685509 Reduces Portal Hypertension and Portosystemic Shunting in a Rat Thioacetamide-Induced Cirrhosis Model. J Pharmacol Exp Ther 2023; 386:70-79. [PMID: 37230799 DOI: 10.1124/jpet.122.001532] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 04/03/2023] [Accepted: 04/10/2023] [Indexed: 05/27/2023] Open
Abstract
Portal hypertension (PT) commonly occurs in cirrhosis. Nitric oxide (NO) imbalance contributes to PT via reduced soluble guanylyl cyclase (sGC) activation and cGMP production, resulting in vasoconstriction, endothelial cell dysfunction, and fibrosis. We assessed the effects of BI 685509, an NO-independent sGC activator, on fibrosis and extrahepatic complications in a thioacetamide (TAA)-induced cirrhosis and PT model. Male Sprague-Dawley rats received TAA twice-weekly for 15 weeks (300-150 mg/kg i.p.). BI 685509 was administered daily for the last 12 weeks (0.3, 1, and 3 mg/kg p.o.; n = 8-11 per group) or the final week only (Acute, 3 mg/kg p.o.; n = 6). Rats were anesthetized to measure portal venous pressure. Pharmacokinetics and hepatic cGMP (target engagement) were measured by mass spectrometry. Hepatic Sirius Red morphometry (SRM) and alpha-smooth muscle actin (αSMA) were measured by immunohistochemistry; portosystemic shunting was measured using colored microspheres. BI 685509 dose-dependently increased hepatic cGMP at 1 and 3 mg/kg (3.92 ± 0.34 and 5.14 ± 0.44 versus 2.50 ± 0.19 nM in TAA alone; P < 0.05). TAA increased hepatic SRM, αSMA, PT, and portosystemic shunting. Compared with TAA, 3 mg/kg BI 685509 reduced SRM by 38%, αSMA area by 55%, portal venous pressure by 26%, and portosystemic shunting by 10% (P < 0.05). Acute BI 685509 reduced SRM and PT by 45% and 21%, respectively (P < 0.05). BI 685509 improved hepatic and extrahepatic cirrhosis pathophysiology in TAA-induced cirrhosis. These data support the clinical investigation of BI 685509 for PT in patients with cirrhosis. SIGNIFICANCE STATEMENT: BI 685509 is an NO-independent sGC activator that was tested in a preclinical rat model of TAA-induced nodular, liver fibrosis, portal hypertension, and portal systemic shunting. BI 685509 reduced liver fibrosis, portal hypertension, and portal-systemic shunting in a dose-dependent manner, supporting its clinical assessment to treat portal hypertension in patients with cirrhosis.
Collapse
Affiliation(s)
- Amanda K Jones
- Department of Cardiometabolic Diseases Research, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut (A.K.J., H.C., K.J.N., J.V., M.M., S.Z., J.W., H.S.Q., R.M.F.); and Department of Drug Discovery Sciences, Discovery Science Technologies, Boehringer Ingelheim Pharma GmbH & Co., Biberach an der Riss, Germany (K.B., T.B.)
| | - Hongxing Chen
- Department of Cardiometabolic Diseases Research, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut (A.K.J., H.C., K.J.N., J.V., M.M., S.Z., J.W., H.S.Q., R.M.F.); and Department of Drug Discovery Sciences, Discovery Science Technologies, Boehringer Ingelheim Pharma GmbH & Co., Biberach an der Riss, Germany (K.B., T.B.)
| | - Khing Jow Ng
- Department of Cardiometabolic Diseases Research, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut (A.K.J., H.C., K.J.N., J.V., M.M., S.Z., J.W., H.S.Q., R.M.F.); and Department of Drug Discovery Sciences, Discovery Science Technologies, Boehringer Ingelheim Pharma GmbH & Co., Biberach an der Riss, Germany (K.B., T.B.)
| | - Jorge Villalona
- Department of Cardiometabolic Diseases Research, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut (A.K.J., H.C., K.J.N., J.V., M.M., S.Z., J.W., H.S.Q., R.M.F.); and Department of Drug Discovery Sciences, Discovery Science Technologies, Boehringer Ingelheim Pharma GmbH & Co., Biberach an der Riss, Germany (K.B., T.B.)
| | - Mark McHugh
- Department of Cardiometabolic Diseases Research, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut (A.K.J., H.C., K.J.N., J.V., M.M., S.Z., J.W., H.S.Q., R.M.F.); and Department of Drug Discovery Sciences, Discovery Science Technologies, Boehringer Ingelheim Pharma GmbH & Co., Biberach an der Riss, Germany (K.B., T.B.)
| | - Svetlana Zeveleva
- Department of Cardiometabolic Diseases Research, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut (A.K.J., H.C., K.J.N., J.V., M.M., S.Z., J.W., H.S.Q., R.M.F.); and Department of Drug Discovery Sciences, Discovery Science Technologies, Boehringer Ingelheim Pharma GmbH & Co., Biberach an der Riss, Germany (K.B., T.B.)
| | - James Wilks
- Department of Cardiometabolic Diseases Research, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut (A.K.J., H.C., K.J.N., J.V., M.M., S.Z., J.W., H.S.Q., R.M.F.); and Department of Drug Discovery Sciences, Discovery Science Technologies, Boehringer Ingelheim Pharma GmbH & Co., Biberach an der Riss, Germany (K.B., T.B.)
| | - Klaus Brilisauer
- Department of Cardiometabolic Diseases Research, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut (A.K.J., H.C., K.J.N., J.V., M.M., S.Z., J.W., H.S.Q., R.M.F.); and Department of Drug Discovery Sciences, Discovery Science Technologies, Boehringer Ingelheim Pharma GmbH & Co., Biberach an der Riss, Germany (K.B., T.B.)
| | - Tom Bretschneider
- Department of Cardiometabolic Diseases Research, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut (A.K.J., H.C., K.J.N., J.V., M.M., S.Z., J.W., H.S.Q., R.M.F.); and Department of Drug Discovery Sciences, Discovery Science Technologies, Boehringer Ingelheim Pharma GmbH & Co., Biberach an der Riss, Germany (K.B., T.B.)
| | - Hu Sheng Qian
- Department of Cardiometabolic Diseases Research, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut (A.K.J., H.C., K.J.N., J.V., M.M., S.Z., J.W., H.S.Q., R.M.F.); and Department of Drug Discovery Sciences, Discovery Science Technologies, Boehringer Ingelheim Pharma GmbH & Co., Biberach an der Riss, Germany (K.B., T.B.)
| | - Ryan M Fryer
- Department of Cardiometabolic Diseases Research, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut (A.K.J., H.C., K.J.N., J.V., M.M., S.Z., J.W., H.S.Q., R.M.F.); and Department of Drug Discovery Sciences, Discovery Science Technologies, Boehringer Ingelheim Pharma GmbH & Co., Biberach an der Riss, Germany (K.B., T.B.)
| |
Collapse
|
36
|
Muminovic A, Chirkov YY, Horowitz JD. Effects of Soluble Guanylate Cyclase Stimulators and Activators on Anti-Aggregatory Signalling in Patients with Coronary Artery Spasm. Int J Mol Sci 2023; 24:9273. [PMID: 37298225 PMCID: PMC10252901 DOI: 10.3390/ijms24119273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 05/16/2023] [Accepted: 05/22/2023] [Indexed: 06/12/2023] Open
Abstract
Impairment of the nitric oxide/soluble guanylate cyclase (NO)/sGC) signalling cascade is associated with many forms of cardiovascular disease, resulting not only in compromised vasodilatation but also loss of anti-aggregatory homeostasis. Myocardial ischaemia, heart failure, and atrial fibrillation are associated with moderate impairment of NO/sGC signalling, and we have recently demonstrated that coronary artery spasm (CAS) is engendered by severe impairment of platelet NO/sGC activity resulting in combined platelet and vascular endothelial damage. We therefore sought to determine whether sGC stimulators or activators might normalise NO/sGC homeostasis in platelets. ADP-induced platelet aggregation and its inhibition by the NO donor sodium nitroprusside (SNP), the sGC stimulator riociguat (RIO), and the sCG activator cinaciguat (CINA) alone or in addition to SNP were quantitated. Three groups of individuals were compared: normal subjects (n = 9), patients (Group 1) with myocardial ischaemia, heart failure and/or atrial fibrillation (n = 30), and patients (Group 2) in the chronic stage of CAS (n = 16). As expected, responses to SNP were impaired (p = 0.02) in patients versus normal subjects, with Group 2 patients most severely affected (p = 0.005). RIO alone exerted no anti-aggregatory effects but potentiated responses to SNP to a similar extent irrespective of baseline SNP response. CINA exerted only intrinsic anti-aggregatory effects, but the extent of these varied directly (r = 0.54; p = 0.0009) with individual responses to SNP. Thus, both RIO and CINA tend to normalise anti-aggregatory function in patients in whom NO/sGC signalling is impaired. The anti-aggregatory effects of RIO consist entirely of potentiation of NO, which is not selective of platelet NO resistance. However, the intrinsic anti-aggregatory effects of CINA are most marked in individuals with initially normal NO/sGC signalling, and thus their magnitude is at variance with extent of physiological impairment. These data suggest that RIO and other sGC stimulators should be evaluated for clinical utility in both prophylaxis and treatment of CAS.
Collapse
Affiliation(s)
| | | | - John D. Horowitz
- Basil Hetzel Institute for Translational Research, University of Adelaide, 37a Woodville Road, Adelaide, SA 5011, Australia; (A.M.); (Y.Y.C.)
| |
Collapse
|
37
|
Reiberger T, Berzigotti A, Trebicka J, Ertle J, Gashaw I, Swallow R, Tomisser A. The rationale and study design of two phase II trials examining the effects of BI 685,509, a soluble guanylyl cyclase activator, on clinically significant portal hypertension in patients with compensated cirrhosis. Trials 2023; 24:293. [PMID: 37095557 PMCID: PMC10123479 DOI: 10.1186/s13063-023-07291-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Accepted: 04/03/2023] [Indexed: 04/26/2023] Open
Abstract
BACKGROUND Clinically significant portal hypertension (CSPH) drives cirrhosis-related complications (i.e. hepatic decompensation). Impaired nitric oxide (NO) bioavailability promotes sinusoidal vasoconstriction, which is the initial pathomechanism of CSPH development. Activation of soluble guanylyl cyclase (sGC), a key downstream effector of NO, facilitates sinusoidal vasodilation, which in turn may improve CSPH. Two phase II studies are being conducted to assess the efficacy of the NO-independent sGC activator BI 685,509 in patients with CSPH due to various cirrhosis aetiologies. METHODS The 1366.0021 trial (NCT05161481) is a randomised, placebo-controlled, exploratory study that will assess BI 685,509 (moderate or high dose) for 24 weeks in patients with CSPH due to alcohol-related liver disease. The 1366.0029 trial (NCT05282121) is a randomised, open-label, parallel-group, exploratory study that will assess BI 685,509 (high dose) alone in patients with hepatitis B or C virus infection or non-alcoholic steatohepatitis (NASH) and in combination with 10 mg empagliflozin in patients with NASH and type 2 diabetes mellitus for 8 weeks. The 1366.0021 trial will enrol 105 patients, and the 1366.0029 trial will enrol 80 patients. In both studies, the primary endpoint is the change from baseline in hepatic venous pressure gradient (HVPG) until the end of treatment (24 or 8 weeks, respectively). Secondary endpoints include the proportion of patients with an HVPG reduction of > 10% from baseline, the development of decompensation events and the change from baseline in HVPG after 8 weeks in the 1366.0021 trial. In addition, the trials will assess changes in liver and spleen stiffness by transient elastography, changes in hepatic and renal function and the tolerability of BI 685,509. DISCUSSION These trials will enable the assessment of the short-term (8 weeks) and longer-term (24 weeks) effects and safety of sGC activation by BI 685,509 on CSPH due to various cirrhosis aetiologies. The trials will use central readings of the diagnostic gold standard HVPG for the primary endpoint, as well as changes in established non-invasive biomarkers, such as liver and spleen stiffness. Ultimately, these trials will provide key information for developing future phase III trials. TRIAL REGISTRATION 1366.0021: EudraCT no. 2021-001,285-38; ClinicalTrials.gov NCT05161481. Registered on 17 December 2021, https://www. CLINICALTRIALS gov/ct2/show/NCT05161481 . 1366.0029: EudraCT no. 2021-005,171-40; ClinicalTrials.gov NCT05282121. Registered on 16 March 2022, https://www. CLINICALTRIALS gov/ct2/show/NCT05282121 .
Collapse
Affiliation(s)
- Thomas Reiberger
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Spitalgasse 23, 1090, Vienna, Austria.
- Vienna Hepatic Hemodynamic Laboratory, Medical University of Vienna, Vienna, Austria.
- Christian-Doppler Laboratory for Portal Hypertension and Liver Fibrosis, Medical University of Vienna, Vienna, Austria.
| | - Annalisa Berzigotti
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Jonel Trebicka
- Department of Internal Medicine B, University of Münster, Münster, Germany
- European Foundation for the Study of Chronic Liver Failure, EFCLIF, Barcelona, Spain
| | - Judith Ertle
- Boehringer Ingelheim International GmbH, Ingelheim Am Rhein, Germany
| | - Isabella Gashaw
- Boehringer Ingelheim Pharma GmbH & Co. KG, Ingelheim Am Rhein, Germany
| | | | | |
Collapse
|
38
|
Balzer MS, Pavkovic M, Frederick J, Abedini A, Freyberger A, Vienenkötter J, Mathar I, Siudak K, Eitner F, Sandner P, Grundmann M, Susztak K. Treatment effects of soluble guanylate cyclase modulation on diabetic kidney disease at single-cell resolution. Cell Rep Med 2023; 4:100992. [PMID: 37023747 PMCID: PMC10140477 DOI: 10.1016/j.xcrm.2023.100992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 01/29/2023] [Accepted: 03/14/2023] [Indexed: 04/08/2023]
Abstract
Diabetic kidney disease (DKD) is the most common cause of renal failure. Therapeutics development is hampered by our incomplete understanding of animal models on a cellular level. We show that ZSF1 rats recapitulate human DKD on a phenotypic and transcriptomic level. Tensor decomposition prioritizes proximal tubule (PT) and stroma as phenotype-relevant cell types exhibiting a continuous lineage relationship. As DKD features endothelial dysfunction, oxidative stress, and nitric oxide depletion, soluble guanylate cyclase (sGC) is a promising DKD drug target. sGC expression is specifically enriched in PT and stroma. In ZSF1 rats, pharmacological sGC activation confers considerable benefits over stimulation and is mechanistically related to improved oxidative stress regulation, resulting in enhanced downstream cGMP effects. Finally, we define sGC gene co-expression modules, which allow stratification of human kidney samples by DKD prevalence and disease-relevant measures such as kidney function, proteinuria, and fibrosis, underscoring the relevance of the sGC pathway to patients.
Collapse
Affiliation(s)
- Michael S Balzer
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, 10117 Berlin, Germany; Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, BIH Charité Clinician Scientist Program, 10117 Berlin, Germany
| | - Mira Pavkovic
- Bayer AG, Research and Early Development, Pharma Research Center, 42096 Wuppertal, Germany
| | - Julia Frederick
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Amin Abedini
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Alexius Freyberger
- Bayer AG, Research and Early Development, Pharma Research Center, 42096 Wuppertal, Germany
| | - Julia Vienenkötter
- Bayer AG, Research and Early Development, Pharma Research Center, 42096 Wuppertal, Germany
| | - Ilka Mathar
- Bayer AG, Research and Early Development, Pharma Research Center, 42096 Wuppertal, Germany
| | - Krystyna Siudak
- Bayer AG, Research and Early Development, Pharma Research Center, 42096 Wuppertal, Germany
| | - Frank Eitner
- Bayer AG, Research and Early Development, Pharma Research Center, 42096 Wuppertal, Germany; Division of Nephrology and Clinical Immunology, RWTH Aachen University, 52062 Aachen, Germany
| | - Peter Sandner
- Bayer AG, Research and Early Development, Pharma Research Center, 42096 Wuppertal, Germany; Department of Pharmacology, Hannover Medical School, 30625 Hannover, Germany
| | - Manuel Grundmann
- Bayer AG, Research and Early Development, Pharma Research Center, 42096 Wuppertal, Germany
| | - Katalin Susztak
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
39
|
Sharina I, Martin E. Cellular Factors That Shape the Activity or Function of Nitric Oxide-Stimulated Soluble Guanylyl Cyclase. Cells 2023; 12:471. [PMID: 36766813 PMCID: PMC9914232 DOI: 10.3390/cells12030471] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 01/29/2023] [Accepted: 01/31/2023] [Indexed: 02/04/2023] Open
Abstract
NO-stimulated guanylyl cyclase (SGC) is a hemoprotein that plays key roles in various physiological functions. SGC is a typical enzyme-linked receptor that combines the functions of a sensor for NO gas and cGMP generator. SGC possesses exclusive selectivity for NO and exhibits a very fast binding of NO, which allows it to function as a sensitive NO receptor. This review describes the effect of various cellular factors, such as additional NO, cell thiols, cell-derived small molecules and proteins on the function of SGC as cellular NO receptor. Due to its vital physiological function SGC is an important drug target. An increasing number of synthetic compounds that affect SGC activity via different mechanisms are discovered and brought to clinical trials and clinics. Cellular factors modifying the activity of SGC constitute an opportunity for improving the effectiveness of existing SGC-directed drugs and/or the creation of new therapeutic strategies.
Collapse
Affiliation(s)
| | - Emil Martin
- Department of Internal Medicine, Cardiology Division, The University of Texas—McGovern Medical School, 1941 East Road, Houston, TX 77054, USA
| |
Collapse
|
40
|
Kosmac K, Ismaeel A, Kim-Shapiro DB, McDermott MM. Praliciguat and Soluble Guanylate Cyclase Stimulators for Peripheral Artery Disease. Circ Res 2023; 132:49-51. [PMID: 36603062 DOI: 10.1161/circresaha.122.322298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Kate Kosmac
- University of Kentucky Center for Muscle Biology, Lexington (K.K., A.I.)
| | - Ahmed Ismaeel
- University of Kentucky Center for Muscle Biology, Lexington (K.K., A.I.)
| | | | - Mary M McDermott
- Department of Medicine and Preventive Medicine, Northwestern University Feinberg School of Medicine (M.M.M.)
| |
Collapse
|
41
|
González-Juanatey JR, Comín-Colet J, Pascual Figal D, Bayes-Genis A, Cepeda JM, García-Pinilla JM, García-Quintana A, Manzano L, Zamorano JL. Optimization of Patient Pathway in Heart Failure with Reduced Ejection Fraction and Worsening Heart Failure. Role of Vericiguat. Patient Prefer Adherence 2023; 17:839-849. [PMID: 36999163 PMCID: PMC10044168 DOI: 10.2147/ppa.s400403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 03/10/2023] [Indexed: 04/01/2023] Open
Abstract
Heart failure (HF) is a progressive condition with periods of apparent stability and repeated worsening HF events. Over time, unless optimization of HF treatment, worsening HF events become more frequent and patients enter into a cycle of recurrent events with high morbidity and mortality. In patients with HF there is an activation of deleterious neurohormonal pathways, such as the renin angiotensin aldosterone system and the sympathetic system, and an inhibition of protective pathways, including natriuretic peptides and guanylate cyclase. Therefore, HF burden can be reduced only through a holistic approach that targets all neurohormonal systems. In this context, vericiguat may play a key role, as it is the only HF drug that activates the nitric oxide-soluble guanylate cyclase-cyclic guanosine monophosphate system. On the other hand, it has been described relevant disparities in the management of HF population. Consequently, it is necessary to homogenize the management of these patients, through an integrated patient-care pathway that should be adapted at the local level. In this context, the development of new technologies (ie, video call, specific platforms, remote control devices, etc.) may be very helpful. In this manuscript, a multidisciplinary group of experts analyzed the current evidence and shared their own experience to provide some recommendations about the therapeutic optimization of patients with recent worsening HF, with a particular focus on vericiguat, and also about how the integrated patient-care pathway should be performed.
Collapse
Affiliation(s)
- José Ramón González-Juanatey
- Cardiology Department, Hospital Clínico Universitario Santiago de Compostela, Centro de investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV), Santiago de Compostela, Spain
- Correspondence: José Ramón González-Juanatey, Email
| | - Josep Comín-Colet
- Cardiology Department, Hospital Universitario de Bellvitge, L’Hospitalet de Llobregat, Barcelona, Spain
| | - Domingo Pascual Figal
- Cardiology Department, Hospital Virgen de la Arrixaca, University of Murcia, Murcia, Spain
| | - Antoni Bayes-Genis
- Cardiology Department, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Jose Maria Cepeda
- Department of Internal Medicine, Hospital Vega Baja, Orihuela, Alicante, Spain
| | - José M García-Pinilla
- Cardiology Department, Hospital Universitario Virgen de la Victoria, IBIMA, Málaga, Spain
- Department of Medicine and Dermatology, Universidad de Málaga, Málaga, Spain
| | - Antonio García-Quintana
- Cardiology Department, Hospital Universitario de Gran Canaria Doctor Negrín, Universidad de Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain
| | - Luis Manzano
- Department of Internal Medicine, Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Universidad de Alcalá, Madrid, Spain
| | - Jose Luis Zamorano
- Cardiology Department, Hospital Universitario Ramón y Cajal, Madrid, Spain
| |
Collapse
|
42
|
Mauersberger C, Sager HB, Wobst J, Dang TA, Lambrecht L, Koplev S, Stroth M, Bettaga N, Schlossmann J, Wunder F, Friebe A, Björkegren JLM, Dietz L, Maas SL, van der Vorst EPC, Sandner P, Soehnlein O, Schunkert H, Kessler T. Loss of soluble guanylyl cyclase in platelets contributes to atherosclerotic plaque formation and vascular inflammation. NATURE CARDIOVASCULAR RESEARCH 2022; 1:1174-1186. [PMID: 37484062 PMCID: PMC10361702 DOI: 10.1038/s44161-022-00175-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 10/27/2022] [Indexed: 07/25/2023]
Abstract
Variants in genes encoding the soluble guanylyl cyclase (sGC) in platelets are associated with coronary artery disease (CAD) risk. Here, by using histology, flow cytometry and intravital microscopy, we show that functional loss of sGC in platelets of atherosclerosis-prone Ldlr-/- mice contributes to atherosclerotic plaque formation, particularly via increasing in vivo leukocyte adhesion to atherosclerotic lesions. In vitro experiments revealed that supernatant from activated platelets lacking sGC promotes leukocyte adhesion to endothelial cells (ECs) by activating ECs. Profiling of platelet-released cytokines indicated that reduced platelet angiopoietin-1 release by sGC-depleted platelets, which was validated in isolated human platelets from carriers of GUCY1A1 risk alleles, enhances leukocyte adhesion to ECs. I mp or ta ntly, p ha rm ac ol ogical sGC stimulation increased platelet angiopoietin-1 release in vitro and reduced leukocyte recruitment and atherosclerotic plaque formation in atherosclerosis-prone Ldlr-/- mice. Therefore, pharmacological sGC stimulation might represent a potential therapeutic strategy to prevent and treat CAD.
Collapse
Affiliation(s)
- Carina Mauersberger
- German Heart Centre Munich, Department of Cardiology, Technical University of Munich, Munich, Germany
- German Centre for Cardiovascular Research, Munich Heart Alliance, Munich, Germany
- These authors contributed equally: Carina Mauersberger, Hendrik B. Sager
| | - Hendrik B. Sager
- German Heart Centre Munich, Department of Cardiology, Technical University of Munich, Munich, Germany
- German Centre for Cardiovascular Research, Munich Heart Alliance, Munich, Germany
- These authors contributed equally: Carina Mauersberger, Hendrik B. Sager
| | - Jana Wobst
- German Heart Centre Munich, Department of Cardiology, Technical University of Munich, Munich, Germany
- German Centre for Cardiovascular Research, Munich Heart Alliance, Munich, Germany
| | - Tan An Dang
- German Heart Centre Munich, Department of Cardiology, Technical University of Munich, Munich, Germany
- German Centre for Cardiovascular Research, Munich Heart Alliance, Munich, Germany
| | - Laura Lambrecht
- German Heart Centre Munich, Department of Cardiology, Technical University of Munich, Munich, Germany
- German Centre for Cardiovascular Research, Munich Heart Alliance, Munich, Germany
| | - Simon Koplev
- Department of Genetics and Genomic Sciences, Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, UK
| | - Marlène Stroth
- German Heart Centre Munich, Department of Cardiology, Technical University of Munich, Munich, Germany
- German Centre for Cardiovascular Research, Munich Heart Alliance, Munich, Germany
| | - Noomen Bettaga
- German Heart Centre Munich, Department of Cardiology, Technical University of Munich, Munich, Germany
| | - Jens Schlossmann
- Department of Pharmacology and Toxicology, University of Regensburg, Regensburg, Germany
| | - Frank Wunder
- Bayer AG, R&D Pharmaceuticals, Wuppertal, Germany
| | - Andreas Friebe
- Institute of Physiology, Julius Maximilian University of Würzburg, Würzburg, Germany
| | - Johan L. M. Björkegren
- Department of Genetics and Genomic Sciences, Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medicine, Neo, Karolinska Institutet, Karolinska Universitetssjukhuset, Huddinge, Sweden
- Department of Cardiac Surgery and The Heart Clinic, Tartu University Hospital and Department of Cardiology, Institute of Clinical Medicine, Tartu University, Tartu, Estonia
| | - Lisa Dietz
- Bayer AG, R&D Pharmaceuticals, Wuppertal, Germany
| | - Sanne L. Maas
- Institute for Molecular Cardiovascular Research and Interdisciplinary Centre for Clinical Research, Rhine-Westphalia Technical University of Aachen, Aachen, Germany
| | - Emiel P. C. van der Vorst
- Institute for Molecular Cardiovascular Research and Interdisciplinary Centre for Clinical Research, Rhine-Westphalia Technical University of Aachen, Aachen, Germany
- Institute for Cardiovascular Prevention, Ludwig Maximilian University of Munich, Munich, Germany
| | | | - Oliver Soehnlein
- German Centre for Cardiovascular Research, Munich Heart Alliance, Munich, Germany
- Institute for Cardiovascular Prevention, Ludwig Maximilian University of Munich, Munich, Germany
- Institute for Experimental Pathology, University of Münster, Münster, Germany
- Department of Physiology and Pharmacology and Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Heribert Schunkert
- German Heart Centre Munich, Department of Cardiology, Technical University of Munich, Munich, Germany
- German Centre for Cardiovascular Research, Munich Heart Alliance, Munich, Germany
- These authors jointly supervised this work: Heribert Schunkert, Thorsten Kessler
| | - Thorsten Kessler
- German Heart Centre Munich, Department of Cardiology, Technical University of Munich, Munich, Germany
- German Centre for Cardiovascular Research, Munich Heart Alliance, Munich, Germany
- These authors jointly supervised this work: Heribert Schunkert, Thorsten Kessler
| |
Collapse
|
43
|
Pang K, Wang W, Qin J, Shi Z, Hao L, Ma Y, Xu H, Wu Z, Pan D, Chen Z, Han C. Role of protein phosphorylation in cell signaling, disease, and the intervention therapy. MedComm (Beijing) 2022; 3:e175. [PMID: 36349142 PMCID: PMC9632491 DOI: 10.1002/mco2.175] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Revised: 08/18/2022] [Accepted: 08/22/2022] [Indexed: 11/06/2022] Open
Abstract
Protein phosphorylation is an important post-transcriptional modification involving an extremely wide range of intracellular signaling transduction pathways, making it an important therapeutic target for disease intervention. At present, numerous drugs targeting protein phosphorylation have been developed for the treatment of various diseases including malignant tumors, neurological diseases, infectious diseases, and immune diseases. In this review article, we analyzed 303 small-molecule protein phosphorylation kinase inhibitors (PKIs) registered and participated in clinical research obtained in a database named Protein Kinase Inhibitor Database (PKIDB), including 68 drugs approved by the Food and Drug Administration of the United States. Based on previous classifications of kinases, we divided these human protein phosphorylation kinases into eight groups and nearly 50 families, and delineated their main regulatory pathways, upstream and downstream targets. These groups include: protein kinase A, G, and C (AGC) and receptor guanylate cyclase (RGC) group, calmodulin-dependent protein kinase (CaMK) group, CMGC [Cyclin-dependent kinases (CDKs), Mitogen-activated protein kinases (MAPKs), Glycogen synthase kinases (GSKs), and Cdc2-like kinases (CLKs)] group, sterile (STE)-MAPKs group, tyrosine kinases (TK) group, tyrosine kinase-like (TKL) group, atypical group, and other groups. Different groups and families of inhibitors stimulate or inhibit others, forming an intricate molecular signaling regulatory network. This review takes newly developed new PKIs as breakthrough point, aiming to clarify the regulatory network and relationship of each pathway, as well as their roles in disease intervention, and provide a direction for future drug development.
Collapse
Affiliation(s)
- Kun Pang
- Department of Urology, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical CollegeThe Affiliated Xuzhou Hospital of Medical College of Southeast UniversityThe Affiliated Xuzhou Center Hospital of Nanjing University of Chinese MedicineXuzhouJiangsuChina
| | - Wei Wang
- Department of Medical CollegeSoutheast UniversityNanjingJiangsuChina
| | - Jia‐Xin Qin
- Department of Urology, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical CollegeThe Affiliated Xuzhou Hospital of Medical College of Southeast UniversityThe Affiliated Xuzhou Center Hospital of Nanjing University of Chinese MedicineXuzhouJiangsuChina
| | - Zhen‐Duo Shi
- Department of Urology, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical CollegeThe Affiliated Xuzhou Hospital of Medical College of Southeast UniversityThe Affiliated Xuzhou Center Hospital of Nanjing University of Chinese MedicineXuzhouJiangsuChina
| | - Lin Hao
- Department of Urology, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical CollegeThe Affiliated Xuzhou Hospital of Medical College of Southeast UniversityThe Affiliated Xuzhou Center Hospital of Nanjing University of Chinese MedicineXuzhouJiangsuChina
| | - Yu‐Yang Ma
- Graduate SchoolBengbu Medical CollegeBengbuAnhuiChina
| | - Hao Xu
- Graduate SchoolBengbu Medical CollegeBengbuAnhuiChina
| | - Zhuo‐Xun Wu
- Department of Pharmaceutical SciencesCollege of Pharmacy and Health SciencesSt. John's University, QueensNew YorkNew YorkUSA
| | - Deng Pan
- Graduate SchoolBengbu Medical CollegeBengbuAnhuiChina
| | - Zhe‐Sheng Chen
- Department of Pharmaceutical SciencesCollege of Pharmacy and Health SciencesSt. John's University, QueensNew YorkNew YorkUSA
| | - Cong‐Hui Han
- Department of Urology, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical CollegeThe Affiliated Xuzhou Hospital of Medical College of Southeast UniversityThe Affiliated Xuzhou Center Hospital of Nanjing University of Chinese MedicineXuzhouJiangsuChina
| |
Collapse
|
44
|
Sevilla-Montero J, Munar-Rubert O, Pino-Fadón J, Aguilar-Latorre C, Villegas-Esguevillas M, Climent B, Agrò M, Choya-Foces C, Martínez-Ruiz A, Balsa E, Muñoz-Calleja C, Gómez-Punter RM, Vázquez-Espinosa E, Cogolludo A, Calzada MJ. Cigarette smoke induces pulmonary arterial dysfunction through an imbalance in the redox status of the soluble guanylyl cyclase. Free Radic Biol Med 2022; 193:9-22. [PMID: 36174878 DOI: 10.1016/j.freeradbiomed.2022.09.026] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 09/05/2022] [Accepted: 09/22/2022] [Indexed: 11/19/2022]
Abstract
Chronic obstructive pulmonary disease (COPD), whose main risk factor is cigarette smoking (CS), is one of the most common diseases globally. Some COPD patients also develop pulmonary hypertension (PH), a severe complication that leads to premature death. Evidence suggests reactive oxygen species (ROS) involvement in COPD and PH, especially regarding pulmonary artery smooth muscle cells (PASMC) dysfunction. However, the effects of CS-driven oxidative stress on the pulmonary vasculature are not completely understood. Herein we provide evidence on the effects of CS extract (CSE) exposure on PASMC regarding ROS production, antioxidant response and its consequences on vascular tone dysregulation. Our results indicate that CSE exposure promotes mitochondrial fission, mitochondrial membrane depolarization and increased mitochondrial superoxide levels. However, this superoxide increase did not parallel a counterbalancing antioxidant response in human pulmonary artery (PA) cells. Interestingly, the mitochondrial superoxide scavenger mitoTEMPO reduced mitochondrial fission and membrane potential depolarization caused by CSE. As we have previously shown, CSE reduces PA vasoconstriction and vasodilation. In this respect, mitoTEMPO prevented the impaired nitric oxide-mediated vasodilation, while vasoconstriction remained reduced. Finally, we observed a CSE-driven downregulation of the Cyb5R3 enzyme, which prevents soluble guanylyl cyclase oxidation in PASMC. This might explain the CSE-mediated decrease in PA vasodilation. These results provide evidence that there might be a connection between mitochondrial ROS and altered vasodilation responses in PH secondary to COPD, and strongly support the potential of antioxidant strategies specifically targeting mitochondria as a new therapy for these diseases.
Collapse
Affiliation(s)
- J Sevilla-Montero
- Departamento de Medicina, Facultad de Medicina, Universidad Autónoma de Madrid. Instituto de Investigación Sanitaria Princesa (IIS-Princesa), Madrid, Spain
| | - O Munar-Rubert
- Departamento de Medicina, Facultad de Medicina, Universidad Autónoma de Madrid. Instituto de Investigación Sanitaria Princesa (IIS-Princesa), Madrid, Spain
| | - J Pino-Fadón
- Departamento de Medicina, Facultad de Medicina, Universidad Autónoma de Madrid. Instituto de Investigación Sanitaria Princesa (IIS-Princesa), Madrid, Spain
| | - C Aguilar-Latorre
- Departamento de Medicina, Facultad de Medicina, Universidad Autónoma de Madrid. Instituto de Investigación Sanitaria Princesa (IIS-Princesa), Madrid, Spain
| | - M Villegas-Esguevillas
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
| | - B Climent
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
| | - M Agrò
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa (CBMSO), Consejo Superior de Investigaciones Científicas (CSIC)-Universidad Autónoma de Madrid, Madrid, Spain
| | - C Choya-Foces
- Unidad de Investigación, Hospital Santa Cristina, Instituto de Investigación Sanitaria Princesa (IIS-Princesa), Madrid, Spain
| | - A Martínez-Ruiz
- Unidad de Investigación, Hospital Santa Cristina, Instituto de Investigación Sanitaria Princesa (IIS-Princesa), Madrid, Spain
| | - E Balsa
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa (CBMSO), Consejo Superior de Investigaciones Científicas (CSIC)-Universidad Autónoma de Madrid, Madrid, Spain
| | - C Muñoz-Calleja
- Departamento de Medicina, Facultad de Medicina, Universidad Autónoma de Madrid. Instituto de Investigación Sanitaria Princesa (IIS-Princesa), Madrid, Spain
| | - R M Gómez-Punter
- Servicio de Neumología, Hospital Universitario La Princesa, Instituto de Investigación Sanitaria Princesa (IIS-Princesa), Madrid, Spain
| | - E Vázquez-Espinosa
- Servicio de Neumología, Hospital Universitario La Princesa, Instituto de Investigación Sanitaria Princesa (IIS-Princesa), Madrid, Spain
| | - A Cogolludo
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
| | - M J Calzada
- Departamento de Medicina, Facultad de Medicina, Universidad Autónoma de Madrid. Instituto de Investigación Sanitaria Princesa (IIS-Princesa), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
45
|
Wu G, Sharina I, Martin E. Soluble guanylyl cyclase: Molecular basis for ligand selectivity and action in vitro and in vivo. Front Mol Biosci 2022; 9:1007768. [PMID: 36304925 PMCID: PMC9592903 DOI: 10.3389/fmolb.2022.1007768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Accepted: 09/27/2022] [Indexed: 01/14/2023] Open
Abstract
Nitric oxide (NO), carbon monoxide (CO), oxygen (O2), hydrogen sulfide (H2S) are gaseous molecules that play important roles in the physiology and pathophysiology of eukaryotes. Tissue concentrations of these physiologically relevant gases vary remarkable from nM range for NO to high μM range of O2. Various hemoproteins play a significant role in sensing and transducing cellular signals encoded by gaseous molecules or in transporting them. Soluble guanylyl cyclase (sGC) is a hemoprotein that plays vital roles in a wide range of physiological functions and combines the functions of gaseous sensor and signal transducer. sGC uniquely evolved to sense low non-toxic levels of NO and respond to elevated NO levels by increasing its catalytic ability to generate the secondary signaling messenger cyclic guanosine monophosphate (cGMP). This review discusses sGC's gaseous ligand selectivity and the molecular basis for sGC function as high-affinity and selectivity NO receptor. The effects of other gaseous molecules and small molecules of cellular origin on sGC's function are also discussed.
Collapse
Affiliation(s)
- Gang Wu
- Hematology-Oncology Division, Department of Internal Medicine, The University of Texas—McGovern Medical School, Houston, TX, United States,*Correspondence: Gang Wu, ; Emil Martin,
| | - Iraida Sharina
- Cardiology Division, Department of Internal Medicine, The University of Texas—McGovern Medical School, Houston, TX, United States
| | - Emil Martin
- Cardiology Division, Department of Internal Medicine, The University of Texas—McGovern Medical School, Houston, TX, United States,*Correspondence: Gang Wu, ; Emil Martin,
| |
Collapse
|
46
|
Becker-Pelster EM, Hahn MG, Delbeck M, Dietz L, Hüser J, Kopf J, Kraemer T, Marquardt T, Mondritzki T, Nagelschmitz J, Nikkho SM, Pires PV, Tinel H, Weimann G, Wunder F, Sandner P, Schuhmacher J, Stasch JP, Truebel HKF. Inhaled mosliciguat (BAY 1237592): targeting pulmonary vasculature via activating apo-sGC. Respir Res 2022; 23:272. [PMID: 36183104 PMCID: PMC9526466 DOI: 10.1186/s12931-022-02189-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 09/16/2022] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Oxidative stress associated with severe cardiopulmonary diseases leads to impairment in the nitric oxide/soluble guanylate cyclase signaling pathway, shifting native soluble guanylate cyclase toward heme-free apo-soluble guanylate cyclase. Here we describe a new inhaled soluble guanylate cyclase activator to target apo-soluble guanylate cyclase and outline its therapeutic potential. METHODS We aimed to generate a novel soluble guanylate cyclase activator, specifically designed for local inhaled application in the lung. We report the discovery and in vitro and in vivo characterization of the soluble guanylate cyclase activator mosliciguat (BAY 1237592). RESULTS Mosliciguat specifically activates apo-soluble guanylate cyclase leading to improved cardiopulmonary circulation. Lung-selective effects, e.g., reduced pulmonary artery pressure without reduced systemic artery pressure, were seen after inhaled but not after intravenous administration in a thromboxane-induced pulmonary hypertension minipig model. These effects were observed over a broad dose range with a long duration of action and were further enhanced under experimental oxidative stress conditions. In a unilateral broncho-occlusion minipig model, inhaled mosliciguat decreased pulmonary arterial pressure without ventilation/perfusion mismatch. With respect to airway resistance, mosliciguat showed additional beneficial bronchodilatory effects in an acetylcholine-induced rat model. CONCLUSION Inhaled mosliciguat may overcome treatment limitations in patients with pulmonary hypertension by improving pulmonary circulation and airway resistance without systemic exposure or ventilation/perfusion mismatch. Mosliciguat has the potential to become a new therapeutic paradigm, exhibiting a unique mode of action and route of application, and is currently under clinical development in phase Ib for pulmonary hypertension.
Collapse
Affiliation(s)
- Eva M Becker-Pelster
- Pharmaceuticals R&D, Pharma Research Center, Bayer AG, Aprather Weg 18A, 42113, Wuppertal, Germany.
| | - Michael G Hahn
- Pharmaceuticals R&D, Pharma Research Center, Bayer AG, Aprather Weg 18A, 42113, Wuppertal, Germany
| | - Martina Delbeck
- Pharmaceuticals R&D, Pharma Research Center, Bayer AG, Aprather Weg 18A, 42113, Wuppertal, Germany
| | - Lisa Dietz
- Pharmaceuticals R&D, Pharma Research Center, Bayer AG, Aprather Weg 18A, 42113, Wuppertal, Germany
| | - Jörg Hüser
- Pharmaceuticals R&D, Pharma Research Center, Bayer AG, Aprather Weg 18A, 42113, Wuppertal, Germany
| | - Johannes Kopf
- Pharmaceuticals R&D, Pharma Research Center, Bayer AG, Aprather Weg 18A, 42113, Wuppertal, Germany
| | - Thomas Kraemer
- Pharmaceuticals R&D, Pharma Research Center, Bayer AG, Aprather Weg 18A, 42113, Wuppertal, Germany
| | - Tobias Marquardt
- Pharmaceuticals R&D, Pharma Research Center, Bayer AG, Aprather Weg 18A, 42113, Wuppertal, Germany
| | - Thomas Mondritzki
- Pharmaceuticals R&D, Pharma Research Center, Bayer AG, Aprather Weg 18A, 42113, Wuppertal, Germany
- Fakultät für Gesundheit, University Witten/Herdecke, Witten, Germany
| | - Johannes Nagelschmitz
- Pharmaceuticals R&D, Pharma Research Center, Bayer AG, Aprather Weg 18A, 42113, Wuppertal, Germany
| | - Sylvia M Nikkho
- Pharmaceuticals R&D, Pharma Research Center, Bayer AG, Aprather Weg 18A, 42113, Wuppertal, Germany
| | - Philippe V Pires
- The Janssen Pharmaceutical Companies of Johnson & Johnson, Allschwil, Switzerland
| | - Hanna Tinel
- Pharmaceuticals R&D, Pharma Research Center, Bayer AG, Aprather Weg 18A, 42113, Wuppertal, Germany
| | - Gerrit Weimann
- Pharmaceuticals R&D, Pharma Research Center, Bayer AG, Aprather Weg 18A, 42113, Wuppertal, Germany
| | - Frank Wunder
- Pharmaceuticals R&D, Pharma Research Center, Bayer AG, Aprather Weg 18A, 42113, Wuppertal, Germany
| | - Peter Sandner
- Pharmaceuticals R&D, Pharma Research Center, Bayer AG, Aprather Weg 18A, 42113, Wuppertal, Germany
- Department of Pharmacology, Hannover Medical School, Hannover, Germany
| | - Joachim Schuhmacher
- Pharmaceuticals R&D, Pharma Research Center, Bayer AG, Aprather Weg 18A, 42113, Wuppertal, Germany
| | - Johannes-Peter Stasch
- Pharmaceuticals R&D, Pharma Research Center, Bayer AG, Aprather Weg 18A, 42113, Wuppertal, Germany
- Institute of Pharmacy, University Halle-Wittenberg, Halle, Germany
| | | |
Collapse
|
47
|
Cui C, Wu C, Shu P, Liu T, Li H, Beuve A. Soluble guanylyl cyclase mediates noncanonical nitric oxide signaling by nitrosothiol transfer under oxidative stress. Redox Biol 2022; 55:102425. [PMID: 35961098 PMCID: PMC9372771 DOI: 10.1016/j.redox.2022.102425] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/20/2022] [Accepted: 07/29/2022] [Indexed: 11/28/2022] Open
Abstract
Soluble guanylyl cyclase (GC1) is an α/β heterodimer producing cGMP when stimulated by nitric oxide (NO). The NO-GC1-cGMP pathway is essential for cardiovascular homeostasis but is disrupted by oxidative stress, which causes GC1 desensitization to NO by heme oxidation and S-nitrosation (SNO) of specific cysteines. We discovered that under these conditions, GC1-α subunit increases cellular S-nitrosation via transfer of nitrosothiols to other proteins (transnitrosation) in cardiac and smooth muscle cells. One of the GC1 SNO-targets was the oxidized form of Thioredoxin1 (oTrx1), which is unidirectionally transnitrosated by GC1 with αC610 as a SNO-donor. Because oTrx1 itself drives transnitrosation, we sought and identified SNO-proteins targeted by both GC1 and Trx1. We found that transnitrosation of the small GTPase RhoA by SNO-GC1 requires oTrx1 as a nitrosothiol relay, suggesting a SNO-GC1→oTrx1→RhoA cascade. The RhoA signaling pathway, which is antagonized by the canonical NO-cGMP pathway, was alternatively inhibited by GC1-α-dependent S-nitrosation under oxidative conditions. We propose that SNO-GC1, via transnitrosation, mediates adaptive responses triggered by oxidation of the canonical NO-cGMP pathway.
Collapse
Affiliation(s)
- Chuanlong Cui
- Rutgers School of Graduate Studies, Newark Health Science, Newark, NJ, 07103, USA; Department of Physiology, Pharmacology and Neuroscience, New Jersey Medical School at Rutgers, Newark, NJ, 07103, USA
| | - Changgong Wu
- Thermo Fisher Scientific, Somerset, NJ, 08873, USA
| | - Ping Shu
- Department of Physiology, Pharmacology and Neuroscience, New Jersey Medical School at Rutgers, Newark, NJ, 07103, USA
| | - Tong Liu
- Center for Advanced Proteomics Research, Department of Microbiology, Biochemistry and Molecular Genetics, New Jersey Medical School at Rutgers, Newark, NJ, 07103, USA
| | - Hong Li
- Center for Advanced Proteomics Research, Department of Microbiology, Biochemistry and Molecular Genetics, New Jersey Medical School at Rutgers, Newark, NJ, 07103, USA
| | - Annie Beuve
- Department of Physiology, Pharmacology and Neuroscience, New Jersey Medical School at Rutgers, Newark, NJ, 07103, USA.
| |
Collapse
|
48
|
Jia XY, Liu YM, Wang YF, An JY, Peng KL, Wang H. Bibliometric study of soluble guanylate cyclase stimulators in cardiovascular research based on web of science from 1992 to 2021. Front Pharmacol 2022; 13:963255. [PMID: 36081943 PMCID: PMC9445840 DOI: 10.3389/fphar.2022.963255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 07/28/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Several studies have shown that soluble guanylate cyclase (sGC) stimulators have cardiovascular (CV) benefits. However, few bibliometric analyses have examined this field systematically. Our study aimed to examine the publications to determine the trends and hotspots in CV research on sGC stimulators. Methods: Publications on sGC stimulators in CV research were retrieved from the Web of Science Core Collection. VOSviewer and CiteSpace visualization software were used to analyze publication trends, countries (regions) and institutions, journals and cited journals, authors and cited references, as well as keywords. Results: A total of 1,212 literatures were obtained. From its first appearance in 1992–2021 (based on WOSCC record), the overall volume of publications has shown a gradual increasing trend. Nearly one-third were authored by American scholars, and most were published in Circulation, Circulation Research, and Proceedings of the National Academy of Sciences of the United States of America. Bayer Agency in Germany was the leading driving force, and has a high academic reputation in this field. Stasch JP has published the most related articles and been cited most frequently. Half of the top 10 co-cited references were published in the leading highly co-cited journal Circulation and New England Journal of Medicine. “NO,” “allosteric regulation” and “free radicals” were the focus of previous research, “chronic thromboembolic pulmonary hypertension,” “pulmonary hypertension” and “heart failure” were the main research hotspots. The key words “chronic thromboembolic pulmonary hypertension,” “Pulmonary hypertension,” “preserved ejection fraction” and “heart failure” appeared most recently as research frontiers. Conclusion: The research in the CV field of sGC stimulators was relatively comprehensive, and there was a close relationship among countries, research institutions and authors, but it is still in the exploratory stage in the treatment of CV disease. At present, most studies focus on the results of clinical trials. sGC stimulators in the treatment of heart failure, especially heart failure with preserved ejection fraction, may be the hotpots and Frontier at present and in the future, and should be closely monitored.
Collapse
|
49
|
Escobar C, Luis-Bonilla J, Crespo-Leiro MG, Esteban-Fernández A, Farré N, Garcia A, Nuñez J. Individualizing the treatment of patients with heart failure with reduced ejection fraction: a journey from hospitalization to long-term outpatient care. Expert Opin Pharmacother 2022; 23:1589-1599. [PMID: 35995759 DOI: 10.1080/14656566.2022.2116275] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION : Despite the relevant advances achieved thanks to the traditional step-by-step therapeutic approach, heart failure with reduced ejection fraction (HFrEF) remains associated with considerable morbidity and mortality. The pathogenesis of HFrEF is complex, with the implication of various neurohormonal systems, including activation of deleterious pathways (i.e. renin-angiotensin-aldosterone, sympathetic, and sodium-glucose cotransporter-2 [SGLT2] systems) and the inhibition of protective pathways (i.e. natriuretic peptides and the guanylate cyclase system). Therefore, the burden of HF can only be reduced through a comprehensive approach that involves all evidence-based use of available HF drugs targeting the neurohormonal systems involved. AREAS COVERED : We performed a critical analysis of evidence from recent clinical trials and assessed the effects of HF therapies on hemodynamics and renal function. EXPERT OPINION : HF therapy must be adapted to the clinical profile (i.e. congestion, blood pressure, heart rate, renal function, and electrolytes). Consequently, blood pressure is reduced by beta blockers, renin-angiotensin-aldosterone system inhibitors, sacubitril/valsartan, and, minimally, by SGLT2 inhibitors and vericiguat; heart rate decreases with beta blockers and ivabradine; and renal function is impaired and potassium are levels increased with renin-angiotensin-aldosterone system inhibitors and sacubitril/valsartan. Practical recommendations on how to individualize HF therapy according to patient profile are provided.
Collapse
Affiliation(s)
- Carlos Escobar
- Cardiology Service, Hospital Universitario La Paz, Madrid, Spain
| | | | | | | | | | - Ana Garcia
- Hospital Clinic I Provincial De Barcelona, Barcelona, Spain
| | - Julio Nuñez
- Hospital Clinico de Valencia, Valencia, Spain
| |
Collapse
|
50
|
Mak VW, Patel AM, Yen R, Hanisak J, Lim YH, Bao J, Zheng R, Seganish WM, Yu Y, Healy DR, Ogawa A, Ren Z, Soriano A, Ermakov GP, Beaumont M, Metwally E, Cheng AC, Verras A, Fischmann T, Zebisch M, Silvestre HL, McEwan PA, Barker J, Rearden P, Greshock TJ. Optimization and Mechanistic Investigations of Novel Allosteric Activators of PKG1α. J Med Chem 2022; 65:10318-10340. [PMID: 35878399 DOI: 10.1021/acs.jmedchem.1c02109] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Activation of PKG1α is a compelling strategy for the treatment of cardiovascular diseases. As the main effector of cyclic guanosine monophosphate (cGMP), activation of PKG1α induces smooth muscle relaxation in blood vessels, lowers pulmonary blood pressure, prevents platelet aggregation, and protects against cardiac stress. The development of activators has been mostly limited to cGMP mimetics and synthetic peptides. Described herein is the optimization of a piperidine series of small molecules to yield activators that demonstrate in vitro phosphorylation of vasodilator-stimulated phosphoprotein as well as antiproliferative effects in human pulmonary arterial smooth muscle cells. Hydrogen/deuterium exchange mass spectrometry experiments with the small molecule activators revealed a mechanism of action consistent with cGMP-induced activation, and an X-ray co-crystal structure with a construct encompassing the regulatory domains illustrated a binding mode in an allosteric pocket proximal to the low-affinity cyclic nucleotide-binding domain.
Collapse
Affiliation(s)
- Victor W Mak
- Discovery Chemistry, Merck & Co., Inc., South San Francisco, California 94080, United States
| | - Akash M Patel
- Discovery Chemistry, Merck & Co., Inc., South San Francisco, California 94080, United States
| | - Rose Yen
- Discovery Chemistry, Merck & Co., Inc., South San Francisco, California 94080, United States
| | - Jennifer Hanisak
- Discovery Chemistry, Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - Yeon-Hee Lim
- Discovery Chemistry, Merck & Co., Inc., South San Francisco, California 94080, United States
| | - Jianming Bao
- Discovery Chemistry, Merck & Co., Inc., South San Francisco, California 94080, United States.,Ionova Life Science, Shenzhen 518122, Guangdong, China
| | - Rong Zheng
- IDSU, Wuxi AppTec Co., Ltd, Shanghai 200131, China
| | - W Michael Seganish
- Discovery Chemistry, Merck & Co., Inc., South San Francisco, California 94080, United States
| | - Yang Yu
- Discovery Chemistry, Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - David R Healy
- Discovery Biology, Merck & Co., Inc., Boston, Massachusetts 02115, United States
| | - Aimie Ogawa
- Quantitative Biosciences, Merck & Co., Inc., South San Francisco, California 94080, United States
| | - Zhao Ren
- Quantitative Biosciences, Merck & Co., Inc., South San Francisco, California 94080, United States
| | - Aileen Soriano
- Mass Spectrometry and Biophysics, Computation and Structural Chemistry, Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - Grigori P Ermakov
- PPDM Discovery Bioanalytics, Merck & Co., Inc., South San Francisco, California 94080, United States
| | - Maribel Beaumont
- PPDM Discovery Bioanalytics, Merck & Co., Inc., South San Francisco, California 94080, United States
| | - Essam Metwally
- Computational and Structural Chemistry, Merck & Co., Inc., South San Francisco, California 94080, United States
| | - Alan C Cheng
- Computational and Structural Chemistry, Merck & Co., Inc., South San Francisco, California 94080, United States
| | - Andreas Verras
- Schrodinger Inc., 120 West 45th Street, 17th Floor, New York, New York 10036-4041, United States.,Computational and Structural Chemistry, Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - Thierry Fischmann
- Computational and Structural Chemistry, Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - Matthias Zebisch
- Evotec (UK) Ltd, 114 Innovation Drive, Milton Park, Abingdon, Oxfordshire OX14 4RZ, U.K
| | - H Leonardo Silvestre
- Evotec (UK) Ltd, 114 Innovation Drive, Milton Park, Abingdon, Oxfordshire OX14 4RZ, U.K
| | - Paul A McEwan
- Evotec (UK) Ltd, 114 Innovation Drive, Milton Park, Abingdon, Oxfordshire OX14 4RZ, U.K
| | - John Barker
- Evotec (UK) Ltd, 114 Innovation Drive, Milton Park, Abingdon, Oxfordshire OX14 4RZ, U.K
| | - Paul Rearden
- DMPK, Recursion Pharmaceuticals, Salt Lake City, Utah 84101, United States.,PPDM, Merck & Co., Inc., South San Francisco, California 94080, United States
| | - Thomas J Greshock
- Discovery Chemistry, Merck & Co., Inc., South San Francisco, California 94080, United States
| |
Collapse
|