1
|
Wang D, Di D, Jiang B, Wang Y, Jiang Z, Jing Y, Wu H, Xin S. Revealing the multiple faces of LRG1: gene expression, structure, function, and therapeutic potential. J Adv Res 2025:S2090-1232(25)00342-X. [PMID: 40368176 DOI: 10.1016/j.jare.2025.05.024] [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: 12/21/2024] [Revised: 05/05/2025] [Accepted: 05/11/2025] [Indexed: 05/16/2025] Open
Abstract
BACKGROUND As the structural characterization of the Leucine-rich alpha-2-glycoprotein 1 (LRG1) protein progresses, its functional diversity has been increasingly unveiled, highlighting its clinical relevance in elucidating disease pathogenesis and identifying potential therapeutic targets. AIM OF REVIEW Grounded in structural biology principles, this review systematically examines the regulatory mechanisms, pathological functions, and intervention strategies associated with LRG1, providing a theoretical foundation for translating these insights into clinical drug therapies. KEY SCIENTIFIC CONCEPTS OF REVIEW LRG1, distinguished by its leucine-rich repeat motifs, plays a pivotal role in various physiological and pathological processes. This review presents a comprehensive analysis of LRG1's multifaceted characteristics and its implications in disease. Initially, the regulatory mechanisms modulating LRG1 gene expression are detailed, encompassing both transcriptional and post-transcriptional controls. The structural attributes and distributions of LRG1 are subsequently outlined, with an emphasis on the functional relevance of its leucine-rich repeat motifs. Furthermore, the review elaborates on the molecular interactions through which LRG1 engages with distinct receptors, triggering downstream signaling pathways involved in pathological processes. Finally, current therapeutic approaches targeting LRG1 and its receptors are summarized, alongside prospective research avenues for innovative therapeutic development.
Collapse
Affiliation(s)
- Ding Wang
- Department of Vascular Surgery, The First Affiliated Hospital, China Medical University, Shenyang City, Liaoning Province 110001, China; Key Laboratory of Pathogenesis, Prevention, and Therapeutics of Aortic Aneurysm in Liaoning Province, Shenyang City, Liaoning Province 110001, China; Regenerative Medicine Research Center of China Medical University, Shenyang City, Liaoning Province 110001, China
| | - Di Di
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang City, Liaoning Province 110122, China; Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang City, Liaoning Province 110032, China
| | - Bo Jiang
- Department of Vascular Surgery, The First Affiliated Hospital, China Medical University, Shenyang City, Liaoning Province 110001, China; Key Laboratory of Pathogenesis, Prevention, and Therapeutics of Aortic Aneurysm in Liaoning Province, Shenyang City, Liaoning Province 110001, China; Regenerative Medicine Research Center of China Medical University, Shenyang City, Liaoning Province 110001, China
| | - Yunlong Wang
- Department of Rehabilitation Medicine, The First Affiliated Hospital, China Medical University, Shenyang City, Liaoning Province 110001, China
| | - Zhenqi Jiang
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang City, Liaoning Province 110122, China
| | - Yuchen Jing
- Department of Vascular Surgery, The First Affiliated Hospital, China Medical University, Shenyang City, Liaoning Province 110001, China; Key Laboratory of Pathogenesis, Prevention, and Therapeutics of Aortic Aneurysm in Liaoning Province, Shenyang City, Liaoning Province 110001, China; Regenerative Medicine Research Center of China Medical University, Shenyang City, Liaoning Province 110001, China
| | - Huizhe Wu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang City, Liaoning Province 110122, China; Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang City, Liaoning Province 110122, China; Scientific Experimental Center, School of Pharmacy, China Medical University, Shenyang City, Liaoning Province 110122, China.
| | - Shijie Xin
- Department of Vascular Surgery, The First Affiliated Hospital, China Medical University, Shenyang City, Liaoning Province 110001, China; Key Laboratory of Pathogenesis, Prevention, and Therapeutics of Aortic Aneurysm in Liaoning Province, Shenyang City, Liaoning Province 110001, China; Regenerative Medicine Research Center of China Medical University, Shenyang City, Liaoning Province 110001, China.
| |
Collapse
|
2
|
Kim JW, Chung DY, Liu FY, Huang Y, Fridayana FR, Vo MN, Cho KS, Ryu JK, Kwon MH, Yin GN. Bone morphogenetic protein 2 rescues neurogenic abnormalities and angiogenic factors in mice with bilateral cavernous nerve injury. J Sex Med 2025:qdaf091. [PMID: 40349205 DOI: 10.1093/jsxmed/qdaf091] [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: 10/25/2024] [Revised: 03/25/2025] [Accepted: 04/07/2025] [Indexed: 05/14/2025]
Abstract
BACKGROUND Bone morphogenetic protein 2 (BMP2), a key isoform within the bone morphogenetic protein family, plays a critical role in promoting angiogenesis and peripheral nerve regeneration, but its specific role in neurogenic erectile dysfunction (ED) remains unclear. AIM This study aimed to explore the therapeutic efficacy of exogenous recombinant BMP2 protein administration in restoring erectile function in a mouse model of cavernous nerve injury (CNI)-induced ED. METHODS Twelve-week-old male C57BL/6 mice were used to evaluate BMP2 expression and erectile function following CNI. Western blotting and immunofluorescence staining were employed to assess BMP2 levels in corpus cavernosum tissues from both sham-operated and CNI-induced ED mice. Erectile function was measured through electrical stimulation of bilateral cavernous nerves, with subsequent intracavernous pressure parameter recordings. Mechanistic investigations included immunofluorescence staining, terminal deoxynucleotidyl transferase dUTP nick-end labeling assay, and western blot analysis. Additionally, ex vivo neurite outgrowth assays were conducted using dorsal root ganglia (DRG) and major pelvic ganglia (MPG) tissues. OUTCOMES In vivo intracavernous pressure, neurovascular regeneration, proliferation, apoptosis, ex vivo neurite sprouting, and survival signaling were measured. RESULTS Bone morphogenetic protein 2 expression was significantly decreased in the corpus cavernosum of CNI mice. Exogenous administration of recombinant BMP2 protein effectively enhanced erectile function in CNI mice, likely through the restoration of endothelial cells, smooth muscle cells, pericytes, and neuronal cells within the corpus cavernosum. Immunofluorescence staining and western blot analysis demonstrated that BMP2 treatment promoted angiogenesis by increasing endothelial cell proliferation and reducing apoptosis in the corpus cavernosum. Furthermore, ex vivo assays revealed that BMP2 promoted neurite sprouting in DRG and MPG tissues exposed to lipopolysaccharide. Mechanistic studies further indicated that BMP2 increased the expression of neurotrophic factors and VEGF, activating the AKT/eNOS signaling pathway. CLINICAL IMPLICATIONS Bone morphogenetic protein 2 may be used as a strategy to treat neurogenic ED or other neurovascular diseases. STRENGTHS AND LIMITATIONS Bone morphogenetic protein 2 has dual roles in vascular and neuronal development. Our study focused on broadly evaluating the role of BMP2 in neurogenic ED. Future studies will evaluate the nerve regeneration effects and novel signaling pathways of BMP2 in a sciatic nerve injury mouse model. In view of its properties as an angiogenic factor, its dose concentration should be strictly controlled to avoid potential side effects. CONCLUSIONS The exogenous administration of recombinant BMP2 protein significantly improved erectile function in CNI mice, suggesting BMP2 as a promising therapeutic candidate for neurogenic ED.
Collapse
Affiliation(s)
- Jong Won Kim
- Department of Urology, College of Medicine, Yonsei University, Seoul 03722, Republic of Korea
- National Research Center for Sexual Medicine and Department of Urology, Inha University School of Medicine, Incheon 22332, Republic of Korea
| | - Doo Yong Chung
- National Research Center for Sexual Medicine and Department of Urology, Inha University School of Medicine, Incheon 22332, Republic of Korea
| | - Fang-Yuan Liu
- National Research Center for Sexual Medicine and Department of Urology, Inha University School of Medicine, Incheon 22332, Republic of Korea
| | - Yan Huang
- National Research Center for Sexual Medicine and Department of Urology, Inha University School of Medicine, Incheon 22332, Republic of Korea
- Program in Biomedical Science & Engineering, Inha University, Incheon 22332, Republic of Korea
| | - Fitri Rahma Fridayana
- National Research Center for Sexual Medicine and Department of Urology, Inha University School of Medicine, Incheon 22332, Republic of Korea
- Program in Biomedical Science & Engineering, Inha University, Incheon 22332, Republic of Korea
| | - Minh Nhat Vo
- National Research Center for Sexual Medicine and Department of Urology, Inha University School of Medicine, Incheon 22332, Republic of Korea
| | - Kang Su Cho
- Department of Urology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06273, Republic of Korea
| | - Ji-Kan Ryu
- National Research Center for Sexual Medicine and Department of Urology, Inha University School of Medicine, Incheon 22332, Republic of Korea
- Program in Biomedical Science & Engineering, Inha University, Incheon 22332, Republic of Korea
| | - Mi-Hye Kwon
- National Research Center for Sexual Medicine and Department of Urology, Inha University School of Medicine, Incheon 22332, Republic of Korea
| | - Guo Nan Yin
- National Research Center for Sexual Medicine and Department of Urology, Inha University School of Medicine, Incheon 22332, Republic of Korea
| |
Collapse
|
3
|
Nageswaran V, Carreras A, Reinshagen L, Beck KR, Steinfeldt J, Henricsson M, Ramezani Rad P, Peters L, Strässler ET, Lim J, Verhaar BJ, Döring Y, Weber C, König M, Steinhagen-Thiessen E, Demuth I, Kränkel N, Leistner DM, Potente M, Nieuwdorp M, Knaus P, Kuebler WM, Ferrell M, Nemet I, Hazen SL, Landmesser U, Bäckhed F, Haghikia A. Gut Microbial Metabolite Imidazole Propionate Impairs Endothelial Cell Function and Promotes the Development of Atherosclerosis. Arterioscler Thromb Vasc Biol 2025; 45:823-839. [PMID: 40143816 PMCID: PMC12017598 DOI: 10.1161/atvbaha.124.322346] [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: 12/18/2024] [Accepted: 03/05/2025] [Indexed: 03/28/2025]
Abstract
BACKGROUND The microbially produced amino acid-derived metabolite imidazole propionate (ImP) contributes to the pathogenesis of type 2 diabetes. However, the effects of ImP on endothelial cell (EC) physiology and its role in atherosclerotic coronary artery disease are unknown. Using both human and animal model studies, we investigated the potential contributory role of ImP in the development of atherosclerosis. METHODS Plasma levels of ImP were measured in patients undergoing elective cardiac angiography (n=831) by ultra-high performance liquid chromatography coupled to tandem mass spectrometry. Odds ratios and corresponding 95% confidence intervals for coronary artery disease were calculated based on the ImP quartiles using both univariable and multivariable logistic regression models. The effects of ImP on functional properties of ECs were assessed using HAECs (human aortic endothelial cells). In a mouse model of carotid artery injury, the impact of ImP on vascular regeneration was examined. Additionally, atheroprone Apoe-/- mice fed a high-fat diet were treated with and without ImP (800 µg), and aortic atherosclerotic lesion area was evaluated after 12 weeks. Next-generation sequencing, Western blot analysis, small interfering RNA-based gene knockdown, and tamoxifen-inducible Cre-loxP experiments were performed to investigate ImP-mediated molecular mechanisms. RESULTS Plasma ImP levels in subjects undergoing cardiac evaluation were associated with increased risk of prevalent coronary artery disease. We found that ImP dose dependently impaired migratory and angiogenic properties of human ECs and promoted an increased inflammatory response. Long-term exposure to ImP compromised the repair potential of the endothelium after an arterial insult. In atheroprone Apoe-/- (apolipoprotein E-/-) mice, ImP increased atherosclerotic lesion size. Mechanistically, ImP attenuated insulin receptor signaling by suppressing the PI3K (phosphoinositide 3-kinase)/AKT pathway leading to sustained activation of the FOXO1 (forkhead box protein O1) transcription factor. Genetic inactivation of endothelial FOXO1 signaling in ImP-treated mice enhanced the angiogenic activity and preserved the vascular repair capacity of ECs after carotid injury. CONCLUSIONS Our findings reveal a hitherto unknown role of the microbially produced histidine-derived metabolite ImP in endothelial dysfunction and atherosclerosis, suggesting that ImP metabolism is a potential therapeutic target in atherosclerotic cardiovascular disease.
Collapse
Affiliation(s)
- Vanasa Nageswaran
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Campus Benjamin Franklin, Berlin, Germany (V.N., L.R., J.S., P.R.R., E.T.S., N.K., U.L., A.H.)
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Germany (V.N., L.R., P.R.R., L.P., E.T.S., N.K., M.P., W.M.K., U.L., A.H.)
- Friede Springe-Cardiovascular Prevention Center at Charité, Charité-Universitätsmedizin Berlin, Germany (V.N., J.S., E.S.-T., N.K., U.L., A.H.)
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Germany (V.N., P.K.)
- University Hospital St. Josef-Hospital Bochum, Cardiology and Rhythmology, Ruhr University Bochum, Germany (V.N., L.R., A.H.)
| | - Alba Carreras
- Department of Molecular and Clinical Medicine, Wallenberg Laboratory, Sahlgrenska University Hospital, University of Gothenburg, Sweden (A.C., K.R.B., M.H., F.B.)
| | - Leander Reinshagen
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Campus Benjamin Franklin, Berlin, Germany (V.N., L.R., J.S., P.R.R., E.T.S., N.K., U.L., A.H.)
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Germany (V.N., L.R., P.R.R., L.P., E.T.S., N.K., M.P., W.M.K., U.L., A.H.)
- University Hospital St. Josef-Hospital Bochum, Cardiology and Rhythmology, Ruhr University Bochum, Germany (V.N., L.R., A.H.)
| | - Katharina R. Beck
- Department of Molecular and Clinical Medicine, Wallenberg Laboratory, Sahlgrenska University Hospital, University of Gothenburg, Sweden (A.C., K.R.B., M.H., F.B.)
| | - Jakob Steinfeldt
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Campus Benjamin Franklin, Berlin, Germany (V.N., L.R., J.S., P.R.R., E.T.S., N.K., U.L., A.H.)
- Friede Springe-Cardiovascular Prevention Center at Charité, Charité-Universitätsmedizin Berlin, Germany (V.N., J.S., E.S.-T., N.K., U.L., A.H.)
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Germany (J.S., U.L., A.H.)
| | - Marcus Henricsson
- Department of Molecular and Clinical Medicine, Wallenberg Laboratory, Sahlgrenska University Hospital, University of Gothenburg, Sweden (A.C., K.R.B., M.H., F.B.)
| | - Pegah Ramezani Rad
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Campus Benjamin Franklin, Berlin, Germany (V.N., L.R., J.S., P.R.R., E.T.S., N.K., U.L., A.H.)
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Germany (V.N., L.R., P.R.R., L.P., E.T.S., N.K., M.P., W.M.K., U.L., A.H.)
| | - Lisa Peters
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Germany (V.N., L.R., P.R.R., L.P., E.T.S., N.K., M.P., W.M.K., U.L., A.H.)
- Institute of Physiology, Charité-Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany (L.P., W.M.K.)
- Institute of Biology, Freie Universität Berlin, Germany (L.P.)
| | - Elisabeth T. Strässler
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Campus Benjamin Franklin, Berlin, Germany (V.N., L.R., J.S., P.R.R., E.T.S., N.K., U.L., A.H.)
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Germany (V.N., L.R., P.R.R., L.P., E.T.S., N.K., M.P., W.M.K., U.L., A.H.)
| | - Joseph Lim
- Angiogenesis and Metabolism Laboratory, Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Germany (J.L., M.P.)
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (J.L., M.P.)
| | - Barbara J.H. Verhaar
- Department of Internal Medicine-Geriatrics, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center (UMC), the Netherlands (B.J.H.V., M.N.)
- Department of Vascular Medicine, Amsterdam UMC, the Netherlands (B.J.H.V., M.N.)
| | - Yvonne Döring
- Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilian University of Munich, Germany (Y.D., C.W.)
- Division of Angiology, Swiss Cardiovascular Center, Inselspital, Bern University Hospital, University of Bern, Switzerland (Y.D.)
- Department for BioMedical Research (DBMR), University of Bern, Switzerland (Y.D.)
- German Center for Cardiovascular Research (DZHK), Partner Site Munich, Germany (Y.D., C.W.)
| | - Christian Weber
- Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilian University of Munich, Germany (Y.D., C.W.)
- German Center for Cardiovascular Research (DZHK), Partner Site Munich, Germany (Y.D., C.W.)
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University Medical Centre, the Netherlands (C.W.)
- Munich Cluster for Systems Neurology, Germany (C.W.)
| | - Maximilian König
- Department of Internal Medicine D–Geriatrics, University Medicine Greifswald, Germany (M.K.)
| | - Elisabeth Steinhagen-Thiessen
- Friede Springe-Cardiovascular Prevention Center at Charité, Charité-Universitätsmedizin Berlin, Germany (V.N., J.S., E.S.-T., N.K., U.L., A.H.)
- Department of Endocrinology and Metabolic Diseases (including Division of Lipid Metabolism), Charité-Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany (E.S.-T., I.D.)
| | - Ilja Demuth
- Department of Endocrinology and Metabolic Diseases (including Division of Lipid Metabolism), Charité-Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany (E.S.-T., I.D.)
- Charité–Universitätsmedizin Berlin, Berlin Institute of Health Center for Regenerative Therapies, Germany (I.D.)
| | - Nicolle Kränkel
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Campus Benjamin Franklin, Berlin, Germany (V.N., L.R., J.S., P.R.R., E.T.S., N.K., U.L., A.H.)
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Germany (V.N., L.R., P.R.R., L.P., E.T.S., N.K., M.P., W.M.K., U.L., A.H.)
- Friede Springe-Cardiovascular Prevention Center at Charité, Charité-Universitätsmedizin Berlin, Germany (V.N., J.S., E.S.-T., N.K., U.L., A.H.)
| | - David M. Leistner
- German Center for Cardiovascular Research (DZHK), Partner Site Frankfurt Rhine-Main, Germany (D.M.L.)
- Department of Medicine, Cardiology and Angiology, Goethe University Hospital, Frankfurt, Germany (D.M.L.)
| | - Michael Potente
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Germany (V.N., L.R., P.R.R., L.P., E.T.S., N.K., M.P., W.M.K., U.L., A.H.)
- Angiogenesis and Metabolism Laboratory, Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Germany (J.L., M.P.)
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (J.L., M.P.)
| | - Max Nieuwdorp
- Department of Internal Medicine-Geriatrics, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center (UMC), the Netherlands (B.J.H.V., M.N.)
- Department of Vascular Medicine, Amsterdam UMC, the Netherlands (B.J.H.V., M.N.)
| | - Petra Knaus
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Germany (V.N., P.K.)
- Berlin-Brandenburg School for Regenerative Therapies, Germany (P.K.)
- International Max-Planck Research School for Biology and Computation, Berlin, Germany (P.K.)
| | - Wolfgang M. Kuebler
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Germany (V.N., L.R., P.R.R., L.P., E.T.S., N.K., M.P., W.M.K., U.L., A.H.)
- Institute of Physiology, Charité-Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany (L.P., W.M.K.)
- German Center for Lung Research (DZL), Berlin, Germany (W.M.K.)
- Keenan Research Centre for Biomedical Science at St. Michael’s, Toronto, Canada (W.M.K.)
- Departments of Surgery and Physiology, University of Toronto, Canada (W.M.K.)
| | - Marc Ferrell
- Departments of Cardiovascular and Metabolic Sciences, and Cardiovascular Medicine, Cleveland Clinic, OH (M.F., I.N., S.L.H.)
| | - Ina Nemet
- Departments of Cardiovascular and Metabolic Sciences, and Cardiovascular Medicine, Cleveland Clinic, OH (M.F., I.N., S.L.H.)
| | - Stanley L. Hazen
- Departments of Cardiovascular and Metabolic Sciences, and Cardiovascular Medicine, Cleveland Clinic, OH (M.F., I.N., S.L.H.)
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University School of Medicine, OH (S.L.H.)
| | - Ulf Landmesser
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Campus Benjamin Franklin, Berlin, Germany (V.N., L.R., J.S., P.R.R., E.T.S., N.K., U.L., A.H.)
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Germany (V.N., L.R., P.R.R., L.P., E.T.S., N.K., M.P., W.M.K., U.L., A.H.)
- Friede Springe-Cardiovascular Prevention Center at Charité, Charité-Universitätsmedizin Berlin, Germany (V.N., J.S., E.S.-T., N.K., U.L., A.H.)
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Germany (J.S., U.L., A.H.)
| | - Fredrik Bäckhed
- Department of Molecular and Clinical Medicine, Wallenberg Laboratory, Sahlgrenska University Hospital, University of Gothenburg, Sweden (A.C., K.R.B., M.H., F.B.)
- Department of Clinical Physiology, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden (F.B.)
| | - Arash Haghikia
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Campus Benjamin Franklin, Berlin, Germany (V.N., L.R., J.S., P.R.R., E.T.S., N.K., U.L., A.H.)
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Germany (V.N., L.R., P.R.R., L.P., E.T.S., N.K., M.P., W.M.K., U.L., A.H.)
- Friede Springe-Cardiovascular Prevention Center at Charité, Charité-Universitätsmedizin Berlin, Germany (V.N., J.S., E.S.-T., N.K., U.L., A.H.)
- University Hospital St. Josef-Hospital Bochum, Cardiology and Rhythmology, Ruhr University Bochum, Germany (V.N., L.R., A.H.)
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Germany (J.S., U.L., A.H.)
| |
Collapse
|
4
|
Vo MN, Kwon MH, Liu FY, Fridayana FR, Huang Y, Hong SS, Kang JH, Yin GN, Ryu JK. Exogenous administration of heparin-binding epidermal growth factor-like growth factor improves erectile function in mice with bilateral cavernous nerve injury. Asian J Androl 2025:00129336-990000000-00305. [PMID: 40247713 DOI: 10.4103/aja2024125] [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: 08/19/2024] [Accepted: 01/02/2025] [Indexed: 04/19/2025] Open
Abstract
Prostate cancer is the second most common malignancy and the sixth leading cause of cancer-related death in men worldwide. Radical prostatectomy (RP) is the standard treatment for localized prostate cancer, but the procedure often results in postoperative erectile dysfunction (ED). The poor efficacy of phosphodiesterase 5 inhibitors after surgery highlights the need to develop new therapies to enhance cavernous nerve regeneration and improve the erectile function of these patients. In the present study, we aimed to examine the potential of heparin-binding epidermal growth factor-like growth factor (HB-EGF) in preserving erectile function in cavernous nerve injury (CNI) mice. We found that HB-EGF expression was reduced significantly on the 1st day after CNI in penile tissue. Ex vivo and in vitro studies showed that HB-EGF promotes major pelvic ganglion neurite sprouting and neuro-2a (N2a) cell migration. In vivo studies showed that exogenous HB-EGF treatment significantly restored the erectile function of CNI mice to 86.9% of sham levels. Immunofluorescence staining showed that mural and neuronal cells were preserved by inducing cell proliferation and reducing apoptosis and reactive oxygen species production. Western blot analysis showed that HB-EGF upregulated protein kinase B and extracellular signal-regulated kinase activation and neurotrophic factor expression. Overall, HB-EGF is a major promising therapeutic agent for treating ED in postoperative RP.
Collapse
Affiliation(s)
- Minh Nhat Vo
- National Research Center for Sexual Medicine and Department of Urology, Inha University School of Medicine, Incheon 22332, Korea
| | - Mi-Hye Kwon
- National Research Center for Sexual Medicine and Department of Urology, Inha University School of Medicine, Incheon 22332, Korea
| | - Fang-Yuan Liu
- National Research Center for Sexual Medicine and Department of Urology, Inha University School of Medicine, Incheon 22332, Korea
| | - Fitri Rahma Fridayana
- National Research Center for Sexual Medicine and Department of Urology, Inha University School of Medicine, Incheon 22332, Korea
- Department of Biomedical Sciences, College of Medicine, Program in Biomedical Science and Engineering, Inha University, Incheon 22332, Korea
| | - Yan Huang
- National Research Center for Sexual Medicine and Department of Urology, Inha University School of Medicine, Incheon 22332, Korea
- Department of Biomedical Sciences, College of Medicine, Program in Biomedical Science and Engineering, Inha University, Incheon 22332, Korea
| | - Soon-Sun Hong
- Department of Biomedical Sciences, College of Medicine, Program in Biomedical Science and Engineering, Inha University, Incheon 22332, Korea
| | - Ju-Hee Kang
- Department of Pharmacology, Medicinal Toxicology Research Center, Inha University College of Medicine, Incheon 22212, Korea
| | - Guo Nan Yin
- National Research Center for Sexual Medicine and Department of Urology, Inha University School of Medicine, Incheon 22332, Korea
| | - Ji-Kan Ryu
- National Research Center for Sexual Medicine and Department of Urology, Inha University School of Medicine, Incheon 22332, Korea
- Department of Biomedical Sciences, College of Medicine, Program in Biomedical Science and Engineering, Inha University, Incheon 22332, Korea
| |
Collapse
|
5
|
Beerkens BLH, IJzerman AP, Heitman LH, van der Es D. Covalent functionalization of G protein-coupled receptors by small molecular probes. RSC Chem Biol 2025; 6:528-538. [PMID: 39957994 PMCID: PMC11827490 DOI: 10.1039/d4cb00294f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Accepted: 02/03/2025] [Indexed: 02/18/2025] Open
Abstract
Roughly one-third of all marketed drugs act by binding to one or more of the >800 human GPCRs, primarily through activation or inhibition via the orthosteric binding site. In addition, novel strategies to alter GPCR functioning are being developed, including allosteric, biased and covalently binding ligands. Molecular probes play an important role in verifying such drug molecules with new modes of action and providing information on all factors involved in GPCR signalling. Various types of molecular probes have been developed, ranging from small molecules to antibodies, each bearing its own advantages and disadvantages. In this mini-review, a closer look is taken at small molecular probes that functionalize GPCRs in a covalent manner, such as through the conjugation of reporter groups like fluorophores or biotin. Covalently bound reporter groups allow the investigation of GPCRs across an increasing range of biochemical assay types, yielding new insights into GPCR signalling pathways. Here, a broad range of recently developed 'functionalized covalent probes' is summarized. Furthermore, the use of these probes in biochemical assays and their applications in the field of GPCR research are discussed. Lastly, a view on possible future applications of these types of small molecular probes is provided.
Collapse
Affiliation(s)
- Bert L H Beerkens
- Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research, Leiden University Einsteinweg 55 2333 CC Leiden The Netherlands
- Oncode Institute, Leiden The Netherlands
| | - Adriaan P IJzerman
- Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research, Leiden University Einsteinweg 55 2333 CC Leiden The Netherlands
| | - Laura H Heitman
- Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research, Leiden University Einsteinweg 55 2333 CC Leiden The Netherlands
- Oncode Institute, Leiden The Netherlands
| | - Daan van der Es
- Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research, Leiden University Einsteinweg 55 2333 CC Leiden The Netherlands
| |
Collapse
|
6
|
Huang Y, Jin HR, Liu FY, Fridayana FR, Vo MN, Ryu JK, Yin GN. Isolation, culture, and characterization of primary endothelial cells and pericytes from mouse sciatic nerve. J Neurosci Methods 2025; 416:110366. [PMID: 39848409 DOI: 10.1016/j.jneumeth.2025.110366] [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: 11/18/2024] [Revised: 01/16/2025] [Accepted: 01/20/2025] [Indexed: 01/25/2025]
Abstract
BACKGROUND The recovery of injured peripheral nerves relies on angiogenesis, where newly formed blood vessels act as pathways guiding Schwann cells across the wound to support axon regeneration. While some research has examined this process, the specific mechanisms of angiogenesis in peripheral nerve healing remain unclear. In vitro models are vital tools to investigate these mechanisms; however, no current in vitro culture methods exist for isolating vascular cells, such as endothelial cells (ECs) and pericytes, specifically from sciatic nerves. NEW METHOD We developed a straightforward and reliable technique for isolating ECs and pericytes from injured sciatic nerves, optimized for use in in vitro studies. Cell types were characterized using specific markers and phenotypic assessments, with flow cytometry confirming cell identity and determining cell purity. RESULTS Our method successfully isolated high-purity ECs and pericytes from injured sciatic nerves. Immunofluorescence analysis showed that primary cultured ECs exhibited strong positive staining for CD31, while pericytes stained strongly for NG2 and PDGFRβ. Flow cytometric analysis confirmed that ECs achieved a purity of 90.22 %, and pericytes reached a purity of 92.01 %. Both cell types were capable of forming organized capillary-like structures, and in co-culture systems, pericytes effectively wrapped around ECs. COMPARISON WITH EXISTING METHODS Current isolation methods for ECs and pericytes from sciatic nerves are limited. Although techniques exist for isolating these cells from other tissues, they often rely on enzymatic digestion, which can damage cell surface proteins and reduce cell viability. Our method allows for the efficient isolation of intact ECs and pericytes from sciatic nerve tissue without such drawbacks, providing a robust platform for in vitro studies. CONCLUSIONS This newly developed method offers an effective approach to isolate ECs and pericytes from the sciatic nerve, contributing a valuable tool for investigating the function and pathology of angiogenesis in the context of sciatic nerve injury recovery.
Collapse
Affiliation(s)
- Yan Huang
- National Research Center for Sexual Medicine and Department of Urology, Inha University College of Medicine, Incheon 22332, Republic of Korea; Program in Biomedical Science & Engineering, Inha University, Incheon 22332, Republic of Korea
| | - Hai-Rong Jin
- Department of Urology, Yantai Yuhuangding Hospital Affiliated to Medical College of Qingdao University, Yantai, Shandong 264000, PR China
| | - Fang-Yuan Liu
- National Research Center for Sexual Medicine and Department of Urology, Inha University College of Medicine, Incheon 22332, Republic of Korea
| | - Fitri Rahma Fridayana
- National Research Center for Sexual Medicine and Department of Urology, Inha University College of Medicine, Incheon 22332, Republic of Korea; Program in Biomedical Science & Engineering, Inha University, Incheon 22332, Republic of Korea
| | - Minh Nhat Vo
- National Research Center for Sexual Medicine and Department of Urology, Inha University College of Medicine, Incheon 22332, Republic of Korea
| | - Ji-Kan Ryu
- National Research Center for Sexual Medicine and Department of Urology, Inha University College of Medicine, Incheon 22332, Republic of Korea; Program in Biomedical Science & Engineering, Inha University, Incheon 22332, Republic of Korea.
| | - Guo Nan Yin
- National Research Center for Sexual Medicine and Department of Urology, Inha University College of Medicine, Incheon 22332, Republic of Korea.
| |
Collapse
|
7
|
Morales DD, Ryu J, Wei C, Hadley JT, Smith MR, Bai J, Lopez-Alvarenga JC, Mummidi S, Duggirala R, Lynch JL, Liu F, Dong LQ. Diet-enhanced LRG1 expression promotes insulin hypersecretion and ER stress in pancreatic beta cells. Diabetologia 2025; 68:615-628. [PMID: 39589509 PMCID: PMC12085111 DOI: 10.1007/s00125-024-06331-0] [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: 04/30/2024] [Accepted: 09/13/2024] [Indexed: 11/27/2024]
Abstract
AIMS/HYPOTHESIS Upregulation of serum leucine-rich α-2-glycoprotein 1 (LRG1) has been implicated in diet-induced obesity and metabolic disorders. However, its specific hormonal actions remain unclear. This study aimed to determine whether diet-enhanced serum LRG1 levels promote hyperinsulinaemia by directly stimulating insulin secretion from pancreatic beta cells. METHODS Human serum samples were obtained from individuals (both male and female) undergoing plastic surgery. Male C57BL/6 wild-type and Lrg1 whole-body knockout (Lrg1KO) mice were fed a 45% high-fat diet, with serum samples collected every 2 weeks to monitor LRG1 and insulin levels throughout diet-induced obesity. MIN6 beta cells were used to investigate the effects of LRG1 on insulin secretion and intracellular Ca2+ release. Antibodies targeting various LRG1 epitopes were used to neutralise LRG1 stimulation in MIN6 cells, and their effectiveness was tested in vivo to assess their ability to prevent LRG1-induced hyperinsulinaemia. RESULTS We observed a significant positive association between human serum LRG1 levels and both age and BMI, with elevated levels observed in individuals with vs without type 2 diabetes. In mice fed a high-fat diet, LRG1 upregulation in serum was associated with hyperinsulinaemia. Lrg1 knockout protected mice from diet-induced islet hyperplasia and the loss of beta cell mass. Furthermore, neutralising LRG1 activity prevented the onset of diet-induced hyperinsulinaemia and preserved glucose tolerance. Mechanistically, LRG1 induces inositol triphosphate (IP3) production and intracellular Ca2+ release from the endoplasmic reticulum (ER) in a phospholipase C (PLC)-dependent manner, leading to excessive insulin secretion and ER stress in MIN6 beta cells. CONCLUSIONS/INTERPRETATION In summary, this study identifies LRG1 as a significant contributor to hyperinsulinaemia and beta cell dysfunction. Targeting LRG1 activity emerges as a promising therapeutic approach for addressing diet-induced beta cell dysfunction and managing type 2 diabetes.
Collapse
Affiliation(s)
- Desirae D Morales
- Department of Cell Systems & Anatomy, The University of Texas Health San Antonio, San Antonio, TX, USA
| | - Jiyoon Ryu
- Department of Cell Systems & Anatomy, The University of Texas Health San Antonio, San Antonio, TX, USA
| | - Cong Wei
- Department of Cell Systems & Anatomy, The University of Texas Health San Antonio, San Antonio, TX, USA
| | - Jason T Hadley
- Department of Cell Systems & Anatomy, The University of Texas Health San Antonio, San Antonio, TX, USA
| | - Maia R Smith
- Department of Cell Systems & Anatomy, The University of Texas Health San Antonio, San Antonio, TX, USA
| | - Juli Bai
- Department of Cell Systems & Anatomy, The University of Texas Health San Antonio, San Antonio, TX, USA
| | - Juan C Lopez-Alvarenga
- Division of Population Health & Biostatistics, The University of Texas at Rio Grande Valley, Edinburg, TX, USA
| | - Srinivas Mummidi
- Department of Health and Behavioral Sciences, Texas A&M University-San Antonio, San Antonio, TX, USA
| | - Ravindranath Duggirala
- Department of Health and Behavioral Sciences, Texas A&M University-San Antonio, San Antonio, TX, USA
| | - Jane L Lynch
- Department of Pediatrics, The University of Texas Health San Antonio, San Antonio, TX, USA
| | - Feng Liu
- National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital Central South University, Changsha, Hunan, China
- Metabolic Syndrome Research Center, The Second Xiangya Hospital Central South University, Changsha, Hunan, China
| | - Lily Q Dong
- Department of Cell Systems & Anatomy, The University of Texas Health San Antonio, San Antonio, TX, USA.
- Sam and Ann Barshop Institute for Longevity and Aging Studies, The University of Texas Health San Antonio, San Antonio, TX, USA.
| |
Collapse
|
8
|
Liu FY, Cho YL, Fridayana FR, Niloofar L, Vo MN, Huang Y, Limanjaya A, Kwon MH, Ock J, Lee SJ, Yin GN, Lee NK, Ryu JK. MT-100, a human Tie2-agonistic antibody, improves penile neurovasculature in diabetic mice via the novel target Srpx2. Exp Mol Med 2025; 57:104-117. [PMID: 39741183 PMCID: PMC11799434 DOI: 10.1038/s12276-024-01373-1] [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: 03/18/2024] [Revised: 09/05/2024] [Accepted: 09/30/2024] [Indexed: 01/02/2025] Open
Abstract
Diabetes is an incurable, chronic disease that can lead to many complications, including angiopathy, peripheral neuropathy, and erectile dysfunction (ED). The angiopoietin-Tie2 signaling pathway plays a critical role in blood vessel development, formation, remodeling, and peripheral nerve regeneration. Therefore, strategies for activating the Tie2 signaling pathway have been developed as potential therapies for neurovascular diseases. Here, we developed a human Tie2-agonistic antibody (MT-100) that not only resists Ang-2 antagonism and activates Tie2 signaling but also regulates a novel target, sushi repeat-containing protein X-linked 2 (Srpx2). This regulation led to the survival of vascular and neuronal cells, a reduction in the production of reactive oxygen species (ROS), activation of the PI3K/AKT/eNOS signaling pathway, increased expression of neurotrophic factors, and ultimately alleviation of ED in diabetic mice. Our findings not only provide conclusive evidence that MT-100 is a promising therapeutic strategy for the treatment of diabetic ED but also suggest it has substantial clinical applications for other complications associated with diabetes.
Collapse
Affiliation(s)
- Fang-Yuan Liu
- National Research Center for Sexual Medicine and Department of Urology, Inha University College of Medicine, Incheon, Republic of Korea
| | - Young-Lai Cho
- Environmental Disease Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Fitri Rahma Fridayana
- National Research Center for Sexual Medicine and Department of Urology, Inha University College of Medicine, Incheon, Republic of Korea
- Program in Biomedical Science & Engineering, Inha University, Incheon, Republic of Korea
| | - Lashkari Niloofar
- National Research Center for Sexual Medicine and Department of Urology, Inha University College of Medicine, Incheon, Republic of Korea
- Program in Biomedical Science & Engineering, Inha University, Incheon, Republic of Korea
| | - Minh Nhat Vo
- National Research Center for Sexual Medicine and Department of Urology, Inha University College of Medicine, Incheon, Republic of Korea
| | - Yan Huang
- National Research Center for Sexual Medicine and Department of Urology, Inha University College of Medicine, Incheon, Republic of Korea
- Program in Biomedical Science & Engineering, Inha University, Incheon, Republic of Korea
| | - Anita Limanjaya
- National Research Center for Sexual Medicine and Department of Urology, Inha University College of Medicine, Incheon, Republic of Korea
| | - Mi-Hye Kwon
- National Research Center for Sexual Medicine and Department of Urology, Inha University College of Medicine, Incheon, Republic of Korea
| | - Jiyeon Ock
- National Research Center for Sexual Medicine and Department of Urology, Inha University College of Medicine, Incheon, Republic of Korea
| | - Seon-Jin Lee
- Environmental Disease Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
- Department of Functional Genomics, University of Science and Technology, Daejeon, Republic of Korea
| | - Guo Nan Yin
- National Research Center for Sexual Medicine and Department of Urology, Inha University College of Medicine, Incheon, Republic of Korea.
| | - Nam-Kyung Lee
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea.
- MabTics Co., Ltd., Daejeon, Republic of Korea.
| | - Ji-Kan Ryu
- National Research Center for Sexual Medicine and Department of Urology, Inha University College of Medicine, Incheon, Republic of Korea.
- Program in Biomedical Science & Engineering, Inha University, Incheon, Republic of Korea.
| |
Collapse
|
9
|
Ock J, Yin GN, Liu FY, Huang Y, Fridayana FR, Vo MN, Ryu JK. Ablation of IGFBP5 expression alleviates neurogenic erectile dysfunction by inducing neurovascular regeneration. Investig Clin Urol 2025; 66:74-86. [PMID: 39791587 PMCID: PMC11729225 DOI: 10.4111/icu.20240325] [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: 09/11/2024] [Revised: 11/08/2024] [Accepted: 11/17/2024] [Indexed: 01/12/2025] Open
Abstract
PURPOSE To investigate the therapeutic potential of eliminating insulin-like growth factor-binding protein 5 (IGFBP5) expression in improving erectile function in mice with cavernous nerve injury (CNI)-induced erectile dysfunction (ED). MATERIALS AND METHODS Eight-week-old male C57BL/6 mice were divided into four groups: a sham-operated group and three CNI-induced ED groups. The CNI-induced ED groups were treated with intracavernous injections 3 days before the CNI procedure. These injections included phosphate-buffered saline, scrambled control short hairpin RNA (shRNA), or shRNA targeting mouse IGFBP5 lentiviral particles. One week after CNI, erectile function was evaluated and the penile tissue was then harvested for histological examination and western blot analysis. Additionally, the major pelvic ganglia (MPG) and dorsal root ganglia (DRG) were cultured for ex vivo neurite outgrowth assays. RESULTS Following CNI, IGFBP5 expression in the cavernous tissues significantly increased, reaching its peak at day 7. First, ablation of IGFBP5 expression promotes neurite sprouting in MPG and DRG when exposed to lipopolysaccharide. Second, ablating IGFBP5 expression in CNI-induced ED mice improved erectile function, likely owing to increased neurovascular contents, including endothelial cells, pericytes, and neuronal processes. Third, ablating IGFBP5 expression in CNI-induced ED mice promoted neurovascular regeneration by increasing cell proliferation, reducing apoptosis, and decreasing Reactive oxygen species production. Finally, western blot analysis demonstrated that IGFBP5 ablation attenuated the JNK/c-Jun signaling pathway, activated the PI3K/AKT signaling pathway, and increased vascular endothelial growth factor and neurotrophic factor expression. CONCLUSIONS Ablating IGFBP5 expression enhanced neurovascular regeneration and ultimately improved erectile function in CNI-induced ED mice.
Collapse
Affiliation(s)
- Jiyeon Ock
- National Research Center for Sexual Medicine and Department of Urology, Inha University College of Medicine, Incheon, Korea
| | - Guo Nan Yin
- National Research Center for Sexual Medicine and Department of Urology, Inha University College of Medicine, Incheon, Korea
| | - Fang-Yuan Liu
- National Research Center for Sexual Medicine and Department of Urology, Inha University College of Medicine, Incheon, Korea
| | - Yan Huang
- National Research Center for Sexual Medicine and Department of Urology, Inha University College of Medicine, Incheon, Korea
- Program in Biomedical Science & Engineering, Inha University, Incheon, Korea
| | - Fitri Rahma Fridayana
- National Research Center for Sexual Medicine and Department of Urology, Inha University College of Medicine, Incheon, Korea
- Program in Biomedical Science & Engineering, Inha University, Incheon, Korea
| | - Minh Nhat Vo
- National Research Center for Sexual Medicine and Department of Urology, Inha University College of Medicine, Incheon, Korea
| | - Ji-Kan Ryu
- National Research Center for Sexual Medicine and Department of Urology, Inha University College of Medicine, Incheon, Korea
- Program in Biomedical Science & Engineering, Inha University, Incheon, Korea.
| |
Collapse
|
10
|
Yin GN, Ryu JK. Role of pericytes in regulating penile angiogenesis and nerve regeneration. Asian J Androl 2025; 27:13-19. [PMID: 39162179 PMCID: PMC11784945 DOI: 10.4103/aja202455] [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: 02/18/2024] [Accepted: 05/19/2024] [Indexed: 08/21/2024] Open
Abstract
ABSTRACT Pericytes are multifunctional mural cells that surround the abluminal wall of endothelial cells and are associated with vascular development, vascular permeability, and angiogenesis. Additionally, pericytes demonstrate stem cell-like properties and contribute to neuroinflammatory processes. Pericytes have been extensively studied in the central nervous system. However, specific mechanisms underlying its involvement in various physiological and pathological conditions, especially in erectile dysfunction (ED), remain poorly understood. Advancements in in vitro and in vitro techniques, such as single-cell RNA sequencing, are expanding our understanding of pericytes. Recent studies have shown that pericyte dysfunction is considered an important factor in the pathogenesis of vascular and neurological ED. Therefore, this study aims to analyze the specific role of pericytes in ED, focusing on diabetic and neurogenic ED. This article provides a comprehensive review of research findings on PubMed from 2000 to 2023, concerning pericyte dysfunction in the process of ED, offering valuable insights, and suggesting directions for further research.
Collapse
Affiliation(s)
- Guo Nan Yin
- National Research Center for Sexual Medicine and Department of Urology, Inha University School of Medicine, Incheon 22332, Korea
| | - Ji-Kan Ryu
- National Research Center for Sexual Medicine and Department of Urology, Inha University School of Medicine, Incheon 22332, Korea
- Program in Biomedical Science and Engineering, Inha University, Incheon 22212, Korea
| |
Collapse
|
11
|
Kang M, Lee CS, Son H, Lee J, Lee J, Seo HJ, Kim MK, Choi M, Cho HJ, Kim HS. Latrophilin-2 Deletion in Cardiomyocyte Disrupts Cell Junction, Leading to D-CMP. Circ Res 2024; 135:1098-1115. [PMID: 39421931 DOI: 10.1161/circresaha.124.324670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 09/30/2024] [Accepted: 10/01/2024] [Indexed: 10/19/2024]
Abstract
BACKGROUND Latrophilin-2 (Lphn2), an adhesive GPCR (G protein-coupled receptor), was found to be a specific marker of cardiac progenitors during the differentiation of pluripotent stem cells into cardiomyocytes or during embryonic heart development in our previous studies. Its role in adult heart physiology, however, remains unclear. METHODS The embryonic lethality resulting from Lphn2 deletion necessitates the establishment of cardiomyocyte-specific, tamoxifen-inducible Lphn2 knockout mice, which was achieved by crossing Lphn2 flox/flox mice with mice having MerCreMer (tamoxifen-inducible Cre [Cyclization recombinase] recombinase) under the α-myosin heavy chain promoter. RESULTS Tamoxifen treatment for several days completely suppressed Lphn2 expression, specifically in the myocardium, and induced the dilated cardiomyopathy (D-CMP) phenotype with serious arrhythmia and sudden death in a short period of time. Transmission electron microscopy showed mitochondrial abnormalities, blurred Z-discs, and dehiscent myofibrils. The D-CMP phenotype, or heart failure, worsened during myocardial infarction. In a mechanistic study of D-CMP, Lphn2 knockout suppressed PGC-1α (Peroxisome proliferator-activated receptor gamma coactivator 1-alpha) and mitochondrial dysfunction, leading to the accumulation of reactive oxygen species and the global suppression of junctional molecules, such as N-cadherin (adherens junction), DSC-2 (desmocollin-2; desmosome), and connexin-43 (gap junction), leading to the dehiscence of cardiac myofibers and serious arrhythmia. In an experimental therapeutic trial, activators of p38-MAPK (p38 mitogen-activated protein kinases), which is a downstream signaling molecule of Lphn2, remarkably rescued the D-CMP phenotype of Lphn2 knockout in the heart by restoring PGC-1α and mitochondrial function and recovering global junctional proteins. CONCLUSIONS Lphn2 is a critical regulator of heart integrity by controlling mitochondrial functions and cell-to-cell junctions in cardiomyocytes. Its deficiency leads to D-CMP, which can be rescued by activators of the p38-MAPK pathway.
Collapse
MESH Headings
- Animals
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/pathology
- Mice, Knockout
- Mice
- Cardiomyopathy, Dilated/genetics
- Cardiomyopathy, Dilated/metabolism
- Cardiomyopathy, Dilated/pathology
- Intercellular Junctions/metabolism
- Intercellular Junctions/drug effects
- Receptors, Peptide/genetics
- Receptors, Peptide/metabolism
- Mice, Inbred C57BL
- Receptors, G-Protein-Coupled/metabolism
- Receptors, G-Protein-Coupled/genetics
- Receptors, G-Protein-Coupled/deficiency
- Tamoxifen/pharmacology
- p38 Mitogen-Activated Protein Kinases/metabolism
- Gene Deletion
- Male
- Cells, Cultured
- Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism
- Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics
Collapse
Affiliation(s)
- Minjun Kang
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, College of Medicine or College of Pharmacy, Seoul National University, South Korea (M.K., C.-S.L., H.S., H.J.S., H.-S.K.)
- Biomedical Research Institute (M.K., C.-S.L., H.S., Jaewon Lee, H.J.S., M.-K.K., H.-S.K.), Seoul National University Hospital, South Korea
| | - Choon-Soo Lee
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, College of Medicine or College of Pharmacy, Seoul National University, South Korea (M.K., C.-S.L., H.S., H.J.S., H.-S.K.)
- Biomedical Research Institute (M.K., C.-S.L., H.S., Jaewon Lee, H.J.S., M.-K.K., H.-S.K.), Seoul National University Hospital, South Korea
| | - HyunJu Son
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, College of Medicine or College of Pharmacy, Seoul National University, South Korea (M.K., C.-S.L., H.S., H.J.S., H.-S.K.)
- Biomedical Research Institute (M.K., C.-S.L., H.S., Jaewon Lee, H.J.S., M.-K.K., H.-S.K.), Seoul National University Hospital, South Korea
| | - Jeongha Lee
- Department of Biomedical Sciences, Seoul National University College of Medicine, South Korea (Jeongha Lee, M.C.)
| | - Jaewon Lee
- Biomedical Research Institute (M.K., C.-S.L., H.S., Jaewon Lee, H.J.S., M.-K.K., H.-S.K.), Seoul National University Hospital, South Korea
| | - Hyun Ju Seo
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, College of Medicine or College of Pharmacy, Seoul National University, South Korea (M.K., C.-S.L., H.S., H.J.S., H.-S.K.)
- Biomedical Research Institute (M.K., C.-S.L., H.S., Jaewon Lee, H.J.S., M.-K.K., H.-S.K.), Seoul National University Hospital, South Korea
| | - Moo-Kang Kim
- Department of Internal Medicine (M.-K.K., H.-J.C., H.-S.K.), Seoul National University Hospital, South Korea
- Biomedical Research Institute (M.K., C.-S.L., H.S., Jaewon Lee, H.J.S., M.-K.K., H.-S.K.), Seoul National University Hospital, South Korea
| | - Murim Choi
- Department of Biomedical Sciences, Seoul National University College of Medicine, South Korea (Jeongha Lee, M.C.)
| | - Hyun-Jai Cho
- Department of Internal Medicine (M.-K.K., H.-J.C., H.-S.K.), Seoul National University Hospital, South Korea
| | - Hyo-Soo Kim
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, College of Medicine or College of Pharmacy, Seoul National University, South Korea (M.K., C.-S.L., H.S., H.J.S., H.-S.K.)
- Department of Internal Medicine (M.-K.K., H.-J.C., H.-S.K.), Seoul National University Hospital, South Korea
- Biomedical Research Institute (M.K., C.-S.L., H.S., Jaewon Lee, H.J.S., M.-K.K., H.-S.K.), Seoul National University Hospital, South Korea
| |
Collapse
|
12
|
Fridayana FR, Ock J, Liu FY, Niloofar L, Vo MN, Huang Y, Yin GN, Ryu JK. Heparin-binding epidermal growth factor-like growth factor improves erectile function in streptozotocin-induced diabetic mice. J Sex Med 2024; 21:751-761. [PMID: 39033084 DOI: 10.1093/jsxmed/qdae079] [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: 02/10/2024] [Revised: 06/05/2024] [Accepted: 06/18/2024] [Indexed: 07/23/2024]
Abstract
BACKGROUND Heparin-binding epidermal growth factor-like growth factor (HB-EGF) serves as a pro-angiogenic factor; however, there is to our knowledge currently no reported research on the relationship between HB-EGF and diabetic erectile dysfunction (ED). AIM In this study we aimed to determine whether HB-EGF can improve the erectile function of streptozotocin-induced diabetic mice and to explore the related mechanisms. METHODS Eight-week-old male C57BL/6 mice were used for diabetes induction. Diabetes mellitus (DM) was induced by low-dose injections of streptozotocin (50 mg/kg) for 5 consecutive days. Eight weeks after streptozotocin injections, DM was determined by measuring blood glucose and body weight. Diabetic mice were treated with two intracavernous administrations of phosphate-buffered saline (20 μL) or various doses of HB-EGF (days -3 and 0; 1, 5, and 10 μg in 20 μL of phosphate-buffered saline). The angiogenesis effect of HB-EGF was confirmed by tube formation and migration assays in mouse cavernous endothelial cells and mouse cavernous pericytes under high-glucose conditions. Erectile function was measured by electrical stimulation of the cavernous nerve, as well as histological examination and Western blot analysis for mechanism assessment. OUTCOMES In vitro angiogenesis, cell proliferation, in vivo intracavernous pressure, neurovascular regeneration, cavernous permeability, and survival signaling were the outcomes measured. RESULTS Expression of HB-EGF was reduced under diabetic conditions. Exogenous HB-EGF induced angiogenesis in mouse cavernous endothelial cells and mouse cavernous pericytes under high-glucose conditions. Erectile function was decreased in the DM group, whereas administration of HB-EGF resulted in a significant improvement of erectile function (91% of the age-matched control group) in association with increased neurovascular content, including cavernous endothelial cells, pericytes, and neuronal cells. Histological and Western blot analyses revealed a significant increase in the permeability of the corpus cavernosum in DM mice, which was attenuated by HB-EGF treatment. The protein expression of phospho-Akt Ser473 and phosphorylated endothelial nitric oxide synthase Ser1177 increased after HB-EGF treatment. CLINICAL IMPLICATIONS The use of HB-EGF may be an effective strategy to treat ED associated with DM or other neurovascular diseases. STRENGTHS AND LIMITATIONS Similarly to other pro-angiogenic factors, HB-EGF has dual roles in vascular and neuronal development. Our study focused on broadly evaluating the role of HB-EGF in diabetic ED. In view of the properties of HB-EGF as an angiogenic factor, its dose concentration should be strictly controlled to avoid potential side effects. CONCLUSION In the diabetic ED mouse model in this study erectile function was improved by HB-EGF, which may provide new treatment strategies for patients with ED who do not respond to phosphodiesterase 5 Inhibitors.
Collapse
Affiliation(s)
- Fitri Rahma Fridayana
- National Research Center for Sexual Medicine and Department of Urology, Inha University College of Medicine, Incheon 22332, Republic of Korea
- Program in Biomedical Science & Engineering, Inha University, Incheon 22332, Republic of Korea
| | - Jiyeon Ock
- National Research Center for Sexual Medicine and Department of Urology, Inha University College of Medicine, Incheon 22332, Republic of Korea
| | - Fang-Yuan Liu
- National Research Center for Sexual Medicine and Department of Urology, Inha University College of Medicine, Incheon 22332, Republic of Korea
| | - Lashkari Niloofar
- National Research Center for Sexual Medicine and Department of Urology, Inha University College of Medicine, Incheon 22332, Republic of Korea
- Program in Biomedical Science & Engineering, Inha University, Incheon 22332, Republic of Korea
| | - Minh Nhat Vo
- National Research Center for Sexual Medicine and Department of Urology, Inha University College of Medicine, Incheon 22332, Republic of Korea
| | - Yan Huang
- National Research Center for Sexual Medicine and Department of Urology, Inha University College of Medicine, Incheon 22332, Republic of Korea
- Program in Biomedical Science & Engineering, Inha University, Incheon 22332, Republic of Korea
| | - Guo Nan Yin
- National Research Center for Sexual Medicine and Department of Urology, Inha University College of Medicine, Incheon 22332, Republic of Korea
| | - Ji-Kan Ryu
- National Research Center for Sexual Medicine and Department of Urology, Inha University College of Medicine, Incheon 22332, Republic of Korea
- Program in Biomedical Science & Engineering, Inha University, Incheon 22332, Republic of Korea
| |
Collapse
|
13
|
Huang Y, Yin GN, Liu FY, Fridayana FR, Niloofar L, Vo MN, Ryu JK. Argonaute 2 restored erectile function and corpus cavernosum mitochondrial function by reducing apoptosis in a mouse model of cavernous nerve injury. Investig Clin Urol 2024; 65:400-410. [PMID: 38978220 PMCID: PMC11231665 DOI: 10.4111/icu.20240077] [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: 03/06/2024] [Revised: 04/18/2024] [Accepted: 05/09/2024] [Indexed: 07/10/2024] Open
Abstract
PURPOSE To determine whether the overexpression of the Argonaute RNA-induced silencing complex catalytic component 2 (Ago2) improves erectile function in mice after cavernous nerve injury (CNI). MATERIALS AND METHODS Lentiviruses containing Ago2 open reading frame (ORF) mouse clone (Ago2 O/E) were used to overexpress Ago2, and lentiviruses ORF negative control particles (NC) were used as a negative control. Three days before preparing the CNI model, we injected lentiviruses into the penises of 8-week-old male C57BL/6 mice. Animals were then divided into four groups: the sham operation control group and the CNI+phosphate-buffered saline, CNI+NC, and CNI+Ago2 O/E groups. One week later, erectile function was assessed by electrically stimulating cavernous nerves bilaterally and obtaining intracavernous pressure parameters. Penile tissue was also collected for molecular mechanism studies. RESULTS Ago2 overexpression improved erectile function in mice after CNI-induced erectile dysfunction (ED). Immunofluorescence staining and Western blot analysis showed that under Ago2 overexpressing conditions, the contents of endothelial cells, pericytes, and neuronal cells increased in the penile tissues of CNI mice, and this was attributed to reduced apoptosis and ROS production. In addition, we also found that Ago2 overexpression could restore penile mitochondrial function, thereby improving erectile function in CNI-induced ED mice. CONCLUSIONS Our findings demonstrate that Ago2 overexpression can reduce penile cell apoptosis, restore penile mitochondrial function, and improve erectile function in CNI-induced ED mice.
Collapse
Affiliation(s)
- Yan Huang
- National Research Center for Sexual Medicine and Department of Urology, Inha University College of Medicine, Incheon, Korea
- Program in Biomedical Science & Engineering, Inha University, Incheon, Korea
| | - Guo Nan Yin
- National Research Center for Sexual Medicine and Department of Urology, Inha University College of Medicine, Incheon, Korea
| | - Fang-Yuan Liu
- National Research Center for Sexual Medicine and Department of Urology, Inha University College of Medicine, Incheon, Korea
| | - Fitri Rahma Fridayana
- National Research Center for Sexual Medicine and Department of Urology, Inha University College of Medicine, Incheon, Korea
- Program in Biomedical Science & Engineering, Inha University, Incheon, Korea
| | - Lashkari Niloofar
- National Research Center for Sexual Medicine and Department of Urology, Inha University College of Medicine, Incheon, Korea
- Program in Biomedical Science & Engineering, Inha University, Incheon, Korea
| | - Minh Nhat Vo
- National Research Center for Sexual Medicine and Department of Urology, Inha University College of Medicine, Incheon, Korea
| | - Ji-Kan Ryu
- National Research Center for Sexual Medicine and Department of Urology, Inha University College of Medicine, Incheon, Korea
- Program in Biomedical Science & Engineering, Inha University, Incheon, Korea.
| |
Collapse
|
14
|
Bae SG, Yin GN, Ock J, Suh JK, Ryu JK, Park J. Single-cell transcriptome analysis of cavernous tissues reveals the key roles of pericytes in diabetic erectile dysfunction. eLife 2024; 12:RP88942. [PMID: 38856719 PMCID: PMC11164535 DOI: 10.7554/elife.88942] [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] [Indexed: 06/11/2024] Open
Abstract
Erectile dysfunction (ED) affects a significant proportion of men aged 40-70 and is caused by cavernous tissue dysfunction. Presently, the most common treatment for ED is phosphodiesterase 5 inhibitors; however, this is less effective in patients with severe vascular disease such as diabetic ED. Therefore, there is a need for development of new treatment, which requires a better understanding of the cavernous microenvironment and cell-cell communications under diabetic condition. Pericytes are vital in penile erection; however, their dysfunction due to diabetes remains unclear. In this study, we performed single-cell RNA sequencing to understand the cellular landscape of cavernous tissues and cell type-specific transcriptional changes in diabetic ED. We found a decreased expression of genes associated with collagen or extracellular matrix organization and angiogenesis in diabetic fibroblasts, chondrocytes, myofibroblasts, valve-related lymphatic endothelial cells, and pericytes. Moreover, the newly identified pericyte-specific marker, Limb Bud-Heart (Lbh), in mouse and human cavernous tissues, clearly distinguishing pericytes from smooth muscle cells. Cell-cell interaction analysis revealed that pericytes are involved in angiogenesis, adhesion, and migration by communicating with other cell types in the corpus cavernosum; however, these interactions were highly reduced under diabetic conditions. Lbh expression is low in diabetic pericytes, and overexpression of LBH prevents erectile function by regulating neurovascular regeneration. Furthermore, the LBH-interacting proteins (Crystallin Alpha B and Vimentin) were identified in mouse cavernous pericytes through LC-MS/MS analysis, indicating that their interactions were critical for maintaining pericyte function. Thus, our study reveals novel targets and insights into the pathogenesis of ED in patients with diabetes.
Collapse
Affiliation(s)
- Seo-Gyeong Bae
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST)GwangjuRepublic of Korea
| | - Guo Nan Yin
- National Research Center for Sexual Medicine and Department of Urolog, Inha University School of MedicineIncheonRepublic of Korea
| | - Jiyeon Ock
- National Research Center for Sexual Medicine and Department of Urolog, Inha University School of MedicineIncheonRepublic of Korea
| | - Jun-Kyu Suh
- National Research Center for Sexual Medicine and Department of Urolog, Inha University School of MedicineIncheonRepublic of Korea
| | - Ji-Kan Ryu
- National Research Center for Sexual Medicine and Department of Urolog, Inha University School of MedicineIncheonRepublic of Korea
- Program in Biomedical Science & Engineering, Inha UniversityIncheonRepublic of Korea
| | - Jihwan Park
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST)GwangjuRepublic of Korea
| |
Collapse
|
15
|
Dritsoula A, Camilli C, Moss SE, Greenwood J. The disruptive role of LRG1 on the vasculature and perivascular microenvironment. Front Cardiovasc Med 2024; 11:1386177. [PMID: 38745756 PMCID: PMC11091338 DOI: 10.3389/fcvm.2024.1386177] [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: 02/14/2024] [Accepted: 04/17/2024] [Indexed: 05/16/2024] Open
Abstract
The establishment of new blood vessels, and their subsequent stabilization, is a critical process that facilitates tissue growth and organ development. Once established, vessels need to diversify to meet the specific needs of the local tissue and to maintain homeostasis. These processes are tightly regulated and fundamental to normal vessel and tissue function. The mechanisms that orchestrate angiogenesis and vessel maturation have been widely studied, with signaling crosstalk between endothelium and perivascular cells being identified as an essential component. In disease, however, new vessels develop abnormally, and existing vessels lose their specialization and function, which invariably contributes to disease progression. Despite considerable research into the vasculopathic mechanisms in disease, our knowledge remains incomplete. Accordingly, the identification of angiocrine and angiopathic molecules secreted by cells within the vascular microenvironment, and their effect on vessel behaviour, remains a major research objective. Over the last decade the secreted glycoprotein leucine-rich α-2 glycoprotein 1 (LRG1), has emerged as a significant vasculopathic molecule, stimulating defective angiogenesis, and destabilizing the existing vasculature mainly, but not uniquely, by altering both canonical and non-canonical TGF-β signaling in a highly cell and context dependent manner. Whilst LRG1 does not possess any overt homeostatic role in vessel development and maintenance, growing evidence provides a compelling case for LRG1 playing a pleiotropic role in disrupting the vasculature in many disease settings. Thus, LRG1 has now been reported to damage vessels in various disorders including cancer, diabetes, chronic kidney disease, ocular disease, and lung disease and the signaling processes that drive this dysfunction are being defined. Moreover, therapeutic targeting of LRG1 has been widely proposed to re-establish a quiescent endothelium and normalized vasculature. In this review, we consider the current status of our understanding of the role of LRG1 in vascular pathology, and its potential as a therapeutic target.
Collapse
Affiliation(s)
- Athina Dritsoula
- UCL Institute of Ophthalmology, University College London, London, United Kingdom
| | | | | | | |
Collapse
|
16
|
Mu Y, Luo LB, Wu SJ, Gao Y, Qin XL, Zhao J, Liu Q, Yang J. Bezafibrate alleviates diabetes-induced spermatogenesis dysfunction by inhibiting inflammation and oxidative stress. Heliyon 2024; 10:e28284. [PMID: 38533024 PMCID: PMC10963653 DOI: 10.1016/j.heliyon.2024.e28284] [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: 08/02/2023] [Revised: 03/13/2024] [Accepted: 03/15/2024] [Indexed: 03/28/2024] Open
Abstract
The metabolic disorders caused by diabetes can lead to various complications, including male spermatogenesis dysfunction. Exploring effective therapeutics that attenuate diabetes mellitus (DM)-induced male subfertility is of great importance. Pharmaceuticals targeting PPARα activation such as bezafibrate have been regarded as an important strategy for patients with diabetes. In this study, we use streptozocin (STZ) injection to establish a type 1 DM mice model and use bezafibrate to treat DM mice and evaluate the effects of bezafibrate on the spermatogenic function of the DM male mice. Bezafibrate treatment exhibited protective effects on DM-induced spermatogenesis deficiency, as reflected by increased testis weight, improved histological morphology of testis, elevated sperm parameters, increased serum testosterone concentration as well as increased mRNA levels of steroidogenesis enzymes. Meanwhile, testicular cell apoptosis, inflammation accumulation and oxidative stress status were also shown to be alleviated by bezafibrate compared with the DM group. In vivo and in vitro studies, PPARα specific inhibitor and PPARα knockout mice were further used to investigate the role of PPARα in the protective effects of bezafibrate on DM-induced spermatogenesis dysfunction. Our results indicated that the protection of bezafibrate on DM-induced spermatogenesis deficiency was abrogated by PPARα inhibition or deletion. Our study suggested that bezafibrate administration could ameliorate DM-induced spermatogenesis dysfunction and may represent a novel practical strategy for male infertility.
Collapse
Affiliation(s)
- Yang Mu
- Reproductive Medicine Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Ling-Bo Luo
- Reproductive Medicine Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Shu-juan Wu
- Reproductive Medicine Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yue Gao
- Reproductive Medicine Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Xiao-lin Qin
- Reproductive Medicine Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Jing Zhao
- Reproductive Medicine Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Qian Liu
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Jing Yang
- Reproductive Medicine Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| |
Collapse
|
17
|
Scharf MM, Humphrys LJ, Berndt S, Di Pizio A, Lehmann J, Liebscher I, Nicoli A, Niv MY, Peri L, Schihada H, Schulte G. The dark sides of the GPCR tree - research progress on understudied GPCRs. Br J Pharmacol 2024. [PMID: 38339984 DOI: 10.1111/bph.16325] [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: 08/25/2023] [Revised: 11/24/2023] [Accepted: 01/08/2024] [Indexed: 02/12/2024] Open
Abstract
A large portion of the human GPCRome is still in the dark and understudied, consisting even of entire subfamilies of GPCRs such as odorant receptors, class A and C orphans, adhesion GPCRs, Frizzleds and taste receptors. However, it is undeniable that these GPCRs bring an untapped therapeutic potential that should be explored further. Open questions on these GPCRs span diverse topics such as deorphanisation, the development of tool compounds and tools for studying these GPCRs, as well as understanding basic signalling mechanisms. This review gives an overview of the current state of knowledge for each of the diverse subfamilies of understudied receptors regarding their physiological relevance, molecular mechanisms, endogenous ligands and pharmacological tools. Furthermore, it identifies some of the largest knowledge gaps that should be addressed in the foreseeable future and lists some general strategies that might be helpful in this process.
Collapse
Affiliation(s)
- Magdalena M Scharf
- Karolinska Institutet, Dept. Physiology & Pharmacology, Sec. Receptor Biology & Signaling, Stockholm, Sweden
| | - Laura J Humphrys
- Institute of Pharmacy, University of Regensburg, Regensburg, Germany
| | - Sandra Berndt
- Rudolf Schönheimer Institute for Biochemistry, Molecular Biochemistry, University of Leipzig, Leipzig, Germany
| | - Antonella Di Pizio
- Leibniz Institute for Food Systems Biology at the Technical University of Munich, Freising, Germany
- Chemoinformatics and Protein Modelling, Department of Molecular Life Science, School of Life Science, Technical University of Munich, Freising, Germany
| | - Juliane Lehmann
- Rudolf Schönheimer Institute for Biochemistry, Molecular Biochemistry, University of Leipzig, Leipzig, Germany
| | - Ines Liebscher
- Rudolf Schönheimer Institute for Biochemistry, Molecular Biochemistry, University of Leipzig, Leipzig, Germany
| | - Alessandro Nicoli
- Leibniz Institute for Food Systems Biology at the Technical University of Munich, Freising, Germany
- Chemoinformatics and Protein Modelling, Department of Molecular Life Science, School of Life Science, Technical University of Munich, Freising, Germany
| | - Masha Y Niv
- The Institute of Biochemistry, Food Science and Nutrition, Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Lior Peri
- The Institute of Biochemistry, Food Science and Nutrition, Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Hannes Schihada
- Institute of Pharmaceutical Chemistry, Philipps-University Marburg, Marburg, Germany
| | - Gunnar Schulte
- Karolinska Institutet, Dept. Physiology & Pharmacology, Sec. Receptor Biology & Signaling, Stockholm, Sweden
| |
Collapse
|
18
|
Ock J, Liu FY, Fridayana FR, Niloofar L, Vo MN, Huang Y, Piao S, Zhou T, Guonan Y. MicroRNA-148a-3p in pericyte-derived extracellular vesicles improves erectile function in diabetic mice by promoting cavernous neurovascular regeneration. BMC Urol 2023; 23:209. [PMID: 38104056 PMCID: PMC10725581 DOI: 10.1186/s12894-023-01378-4] [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: 01/06/2023] [Accepted: 11/23/2023] [Indexed: 12/19/2023] Open
Abstract
BACKGROUND To investigate the regulatory role of microRNA (miR)-148a-3p in mouse corpus cavernous pericyte (MCPs)-derived extracellular vesicles (EVs) in the treatment of diabetes-induced erectile dysfunction (ED). METHODS Mouse corpus cavernous tissue was used for MCP primary culture and EV isolation. Small-RNA sequencing analysis was performed to assess the type and content of miRs in MCPs-EVs. Four groups of mice were used: control nondiabetic mice and streptozotocin-induced diabetic mice receiving two intracavernous injections (days - 3 and 0) of phosphate buffered saline, MCPs-EVs transfected with reagent control, or MCPs-EVs transfected with a miR-148a-3p inhibitor. miR-148a-3p function in MCPs-EVs was evaluated by tube-formation assay, migration assay, TUNEL assay, intracavernous pressure, immunofluorescence staining, and Western blotting. RESULTS We extracted EVs from MCPs, and small-RNA sequencing analysis showed miR-148a-3p enrichment in MCPs-EVs. Exogenous MCPs-EV administration effectively promoted mouse cavernous endothelial cell (MCECs) tube formation, migration, and proliferation, and reduced MCECs apoptosis under high-glucose conditions. These effects were significantly attenuated in miR-148a-3p-depleted MCPs-EVs, which were extracted after inhibiting miR-148a-3p expression in MCPs. Repetitive intracavernous injections of MCPs-EVs improved erectile function by inducing cavernous neurovascular regeneration in diabetic mice. Using online bioinformatics databases and luciferase report assays, we predicted that pyruvate dehydrogenase kinase-4 (PDK4) is a potential target gene of miR-148a-3p. CONCLUSIONS Our findings provide new and reliable evidence that miR-148a-3p in MCPs-EVs significantly enhances cavernous neurovascular regeneration by inhibiting PDK4 expression in diabetic mice.
Collapse
Affiliation(s)
- Jiyeon Ock
- Department of Urology and National Research Center for Sexual Medicine, Inha University School of Medicine, 7-206, 3rd ST, Shinheung-Dong, Jung-Gu, Incheon, 22332, Republic of Korea
| | - Fang-Yuan Liu
- Department of Urology and National Research Center for Sexual Medicine, Inha University School of Medicine, 7-206, 3rd ST, Shinheung-Dong, Jung-Gu, Incheon, 22332, Republic of Korea
| | - Fitri Rahma Fridayana
- Department of Urology and National Research Center for Sexual Medicine, Inha University School of Medicine, 7-206, 3rd ST, Shinheung-Dong, Jung-Gu, Incheon, 22332, Republic of Korea
- Program in Biomedical Science & Engineering, Inha University, Incheon, South Korea
| | - Lashkari Niloofar
- Department of Urology and National Research Center for Sexual Medicine, Inha University School of Medicine, 7-206, 3rd ST, Shinheung-Dong, Jung-Gu, Incheon, 22332, Republic of Korea
- Program in Biomedical Science & Engineering, Inha University, Incheon, South Korea
| | - Minh Nhat Vo
- Department of Urology and National Research Center for Sexual Medicine, Inha University School of Medicine, 7-206, 3rd ST, Shinheung-Dong, Jung-Gu, Incheon, 22332, Republic of Korea
| | - Yan Huang
- Department of Urology and National Research Center for Sexual Medicine, Inha University School of Medicine, 7-206, 3rd ST, Shinheung-Dong, Jung-Gu, Incheon, 22332, Republic of Korea
- Program in Biomedical Science & Engineering, Inha University, Incheon, South Korea
| | - Shuguang Piao
- Department of Urology, Changhai Hospital Affiliated with the Naval Medicine University, Shanghai, 200433, People's Republic of China.
| | - Tie Zhou
- Department of Urology, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, No. 1279 Sanmen Road, Shanghai, 200434, China.
| | - Yin Guonan
- Department of Urology and National Research Center for Sexual Medicine, Inha University School of Medicine, 7-206, 3rd ST, Shinheung-Dong, Jung-Gu, Incheon, 22332, Republic of Korea.
| |
Collapse
|
19
|
Chung DY, Ryu JK, Yin GN. Regenerative therapies as a potential treatment of erectile dysfunction. Investig Clin Urol 2023; 64:312-324. [PMID: 37417556 DOI: 10.4111/icu.20230104] [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: 03/20/2023] [Revised: 05/04/2023] [Accepted: 05/22/2023] [Indexed: 07/08/2023] Open
Abstract
Erectile dysfunction (ED) is the most common sexual dysfunction disease in adult males. ED can be caused by many factors, such as vascular disease, neuropathy, metabolic disturbances, psychosocial causes, and side effects of medications. Although current oral phosphodiesterase type 5 inhibitors can achieve a certain effect, they cause temporary dilatation of blood vessels with no curative treatment effects. Emerging targeted technologies, such as stem cell therapy, protein therapy, and low-intensity extracorporeal shock wave therapy (Li-ESWT), are being used to achieve more natural and long-lasting effects in treating ED. However, the development and application of these therapeutic methods are still in their infancy, and their pharmacological pathways and specific mechanisms have not been fully discovered. This article reviews the preclinical basic research progress of stem cells, proteins, and Li-ESWT therapy, as well as the current status of clinical application of Li-ESWT therapy.
Collapse
Affiliation(s)
- Doo Yong Chung
- National Research Center for Sexual Medicine and Department of Urology, Inha University College of Medicine, Incheon, Korea
| | - Ji-Kan Ryu
- National Research Center for Sexual Medicine and Department of Urology, Inha University College of Medicine, Incheon, Korea
- Program in Biomedical Science & Engineering, Inha University, Incheon, Korea
| | - Guo Nan Yin
- National Research Center for Sexual Medicine and Department of Urology, Inha University College of Medicine, Incheon, Korea.
| |
Collapse
|
20
|
Ock J, Wu J, Liu FY, Fridayana FR, Niloofar L, Vo MN, Hong SS, Kang JH, Suh JK, Yin GN, Jin HR, Ryu JK. Heme-binding protein 1 delivered via pericyte-derived extracellular vesicles improves neurovascular regeneration in a mouse model of cavernous nerve injury. Int J Biol Sci 2023; 19:2663-2677. [PMID: 37324943 PMCID: PMC10266087 DOI: 10.7150/ijbs.81809] [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: 12/13/2022] [Accepted: 05/05/2023] [Indexed: 06/17/2023] Open
Abstract
As a peripheral nerve injury disease, cavernous nerve injury (CNI) caused by prostate cancer surgery and other pelvic surgery causes organic damage to cavernous blood vessels and nerves, thereby significantly attenuating the response to phosphodiesterase-5 inhibitors. Here, we investigated the role of heme-binding protein 1 (Hebp1) in erectile function using a mouse model of bilateral CNI, which is known to promote angiogenesis and improve erection in diabetic mice. We found a potent neurovascular regenerative effect of Hebp1 in CNI mice, demonstrating that exogenously delivered Hebp1 improved erectile function by promoting the survival of cavernous endothelial-mural cells and neurons. We further found that endogenous Hebp1 delivered by mouse cavernous pericyte (MCP)-derived extracellular vesicles promoted neurovascular regeneration in CNI mice. Moreover, Hebp1 achieved these effects by reducing vascular permeability through regulation of claudin family proteins. Our findings provide new insights into Hebp1 as a neurovascular regeneration factor and demonstrate its potential therapeutic application to various peripheral nerve injuries.
Collapse
Affiliation(s)
- Jiyeon Ock
- National Research Center for Sexual Medicine and Department of Urology, Inha University College of Medicine, Incheon, 22332, Republic of Korea
| | - Jitao Wu
- Department of Urology, Yantai Yuhuangding Hospital Affiliated to Medical College of Qingdao University, Yantai, Shandong 264000, People's Republic of China
| | - Fang-Yuan Liu
- National Research Center for Sexual Medicine and Department of Urology, Inha University College of Medicine, Incheon, 22332, Republic of Korea
| | - Fitri Rahma Fridayana
- National Research Center for Sexual Medicine and Department of Urology, Inha University College of Medicine, Incheon, 22332, Republic of Korea
| | - Lashkari Niloofar
- National Research Center for Sexual Medicine and Department of Urology, Inha University College of Medicine, Incheon, 22332, Republic of Korea
| | - Minh Nhat Vo
- National Research Center for Sexual Medicine and Department of Urology, Inha University College of Medicine, Incheon, 22332, Republic of Korea
| | - Soon-Sun Hong
- Department of Biomedical Sciences, College of Medicine, Program in Biomedical Science & Engineering, Inha University, Incheon, Korea
| | - Ju-Hee Kang
- Department of Pharmacology, Medicinal Toxicology Research Center, Inha University College of Medicine, Incheon, Korea
| | - Jun-Kyu Suh
- National Research Center for Sexual Medicine and Department of Urology, Inha University College of Medicine, Incheon, 22332, Republic of Korea
| | - Guo Nan Yin
- National Research Center for Sexual Medicine and Department of Urology, Inha University College of Medicine, Incheon, 22332, Republic of Korea
| | - Hai-Rong Jin
- Department of Urology, Yantai Yuhuangding Hospital Affiliated to Medical College of Qingdao University, Yantai, Shandong 264000, People's Republic of China
| | - Ji-Kan Ryu
- National Research Center for Sexual Medicine and Department of Urology, Inha University College of Medicine, Incheon, 22332, Republic of Korea
| |
Collapse
|
21
|
Lin L, Guo Z, He E, Long X, Wang D, Zhang Y, Guo W, Wei Q, He W, Wu W, Li J, Wo L, Hong D, Zheng J, He M, Zhao Q. SIRT2 regulates extracellular vesicle-mediated liver-bone communication. Nat Metab 2023; 5:821-841. [PMID: 37188819 PMCID: PMC10229428 DOI: 10.1038/s42255-023-00803-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 04/11/2023] [Indexed: 05/17/2023]
Abstract
The interplay between liver and bone metabolism remains largely uncharacterized. Here, we uncover a mechanism of liver-bone crosstalk regulated by hepatocyte SIRT2. We demonstrate that hepatocyte SIRT2 expression is increased in aged mice and elderly humans. Liver-specific SIRT2 deficiency inhibits osteoclastogenesis and alleviates bone loss in mouse models of osteoporosis. We identify leucine-rich α-2-glycoprotein 1 (LRG1) as a functional cargo in hepatocyte-derived small extracellular vesicles (sEVs). In SIRT2-deficient hepatocytes, LRG1 levels in sEVs are upregulated, leading to increased transfer of LRG1 to bone-marrow-derived monocytes (BMDMs), and in turn, to inhibition of osteoclast differentiation via reduced nuclear translocation of NF-κB p65. Treatment with sEVs carrying high levels of LRG1 inhibits osteoclast differentiation in human BMDMs and in mice with osteoporosis, resulting in attenuated bone loss in mice. Furthermore, the plasma level of sEVs carrying LRG1 is positively correlated with bone mineral density in humans. Thus, drugs targeting hepatocyte-osteoclast communication may constitute a promising therapeutic strategy for primary osteoporosis.
Collapse
Affiliation(s)
- Longshuai Lin
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Shanghai Frontiers Science Center of Cellular Homeostasis and Human Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Pathology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Zengya Guo
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Enjun He
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xidai Long
- Department of Pathology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Difei Wang
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Shanghai Frontiers Science Center of Cellular Homeostasis and Human Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yingting Zhang
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Shanghai Frontiers Science Center of Cellular Homeostasis and Human Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weihong Guo
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qian Wei
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Shanghai Frontiers Science Center of Cellular Homeostasis and Human Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei He
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Shanghai Frontiers Science Center of Cellular Homeostasis and Human Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wanying Wu
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Shanghai Frontiers Science Center of Cellular Homeostasis and Human Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jingchi Li
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Shanghai Frontiers Science Center of Cellular Homeostasis and Human Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lulu Wo
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Shanghai Frontiers Science Center of Cellular Homeostasis and Human Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dengli Hong
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Shanghai Frontiers Science Center of Cellular Homeostasis and Human Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Junke Zheng
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Shanghai Frontiers Science Center of Cellular Homeostasis and Human Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ming He
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Shanghai Frontiers Science Center of Cellular Homeostasis and Human Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Department of Pathology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China.
| | - Qinghua Zhao
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
22
|
Yang J, Yin GN, Kim DK, Han AR, Lee DS, Min KW, Fu Y, Yun J, Suh JK, Ryu JK, Kim HM. Crystal structure of LRG1 and the functional significance of LRG1 glycan for LPHN2 activation. Exp Mol Med 2023:10.1038/s12276-023-00992-4. [PMID: 37121976 DOI: 10.1038/s12276-023-00992-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Accepted: 02/21/2023] [Indexed: 05/02/2023] Open
Abstract
The serum glycoprotein leucine-rich ɑ-2-glycoprotein 1 (LRG1), primarily produced by hepatocytes and neutrophils, is a multifunctional protein that modulates various signaling cascades, mainly TGFβ signaling. Serum LRG1 and neutrophil-derived LRG1 have different molecular weights due to differences in glycosylation, but the impact of the differential glycan composition in LRG1 on its cellular function is largely unknown. We previously reported that LRG1 can promote both angiogenic and neurotrophic processes under hyperglycemic conditions by interacting with LPHN2. Here, we determined the crystal structure of LRG1, identifying the horseshoe-like solenoid structure of LRG1 and its four N-glycosylation sites. In addition, our biochemical and cell-biological analyses found that the deglycosylation of LRG1, particularly the removal of glycans on N325, is critical for the high-affinity binding of LRG1 to LPHN2 and thus promotes LRG1/LPHN2-mediated angiogenic and neurotrophic processes in mouse tissue explants, even under normal glucose conditions. Moreover, the intracavernous administration of deglycosylated LRG1 in a diabetic mouse model ameliorated vascular and neurological abnormalities and restored erectile function. Collectively, these data indicate a novel role of LRG1 glycans as molecular switches that can tune the range of LRG1's cellular functions, particularly the LRG1/LPHN2 signaling axis.
Collapse
Affiliation(s)
- Jimin Yang
- Center for Biomolecular and Cellular Structure, Institute for Basic Science (IBS), Daejeon, 34126, Republic of Korea
| | - Guo Nan Yin
- National Research Center for Sexual Medicine and Department of Urology, Inha University School of Medicine, Incheon, 22332, Republic of Korea
| | - Do-Kyun Kim
- Center for Biomolecular and Cellular Structure, Institute for Basic Science (IBS), Daejeon, 34126, Republic of Korea
- Korea Zoonosis Research Institute, Jeonbuk National University, Iksan, 54596, Republic of Korea
| | - Ah-Reum Han
- Center for Biomolecular and Cellular Structure, Institute for Basic Science (IBS), Daejeon, 34126, Republic of Korea
| | - Dong Sun Lee
- Center for Biomolecular and Cellular Structure, Institute for Basic Science (IBS), Daejeon, 34126, Republic of Korea
| | - Kwang Wook Min
- Center for Biomolecular and Cellular Structure, Institute for Basic Science (IBS), Daejeon, 34126, Republic of Korea
| | - Yaoyao Fu
- Center for Biomolecular and Cellular Structure, Institute for Basic Science (IBS), Daejeon, 34126, Republic of Korea
| | - Jeongwon Yun
- Graduate School of Medical Science & Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Jun-Kyu Suh
- National Research Center for Sexual Medicine and Department of Urology, Inha University School of Medicine, Incheon, 22332, Republic of Korea.
| | - Ji-Kan Ryu
- National Research Center for Sexual Medicine and Department of Urology, Inha University School of Medicine, Incheon, 22332, Republic of Korea.
| | - Ho Min Kim
- Center for Biomolecular and Cellular Structure, Institute for Basic Science (IBS), Daejeon, 34126, Republic of Korea.
- Graduate School of Medical Science & Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea.
| |
Collapse
|
23
|
Argonaute 2 Restores Erectile Function by Enhancing Angiogenesis and Reducing Reactive Oxygen Species Production in Streptozotocin (STZ)-Induced Type-1 Diabetic Mice. Int J Mol Sci 2023; 24:ijms24032935. [PMID: 36769259 PMCID: PMC9918048 DOI: 10.3390/ijms24032935] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/22/2023] [Accepted: 02/01/2023] [Indexed: 02/05/2023] Open
Abstract
Severe vascular and nerve damage from diabetes is a leading cause of erectile dysfunction (ED) and poor response to oral phosphodiesterase 5 inhibitors. Argonaute 2 (Ago2), a catalytic engine in mammalian RNA interference, is involved in neurovascular regeneration under inflammatory conditions. In the present study, we report that Ago2 administration can effectively improve penile erection by enhancing cavernous endothelial cell angiogenesis and survival under diabetic conditions. We found that although Ago2 is highly expressed around blood vessels and nerves, it is significantly reduced in the penis tissue of diabetic mice. Exogenous administration of the Ago2 protein restored erectile function in diabetic mice by reducing reactive oxygen species production-signaling pathways (inducing eNOS Ser1177/NF-κB Ser536 signaling) and improving cavernous endothelial angiogenesis, migration, and cell survival. Our study provides new evidence that Ago2 mediation may be a promising therapeutic strategy and a new approach for diabetic ED treatment.
Collapse
|
24
|
Functional and Immunofluorescence Evaluations of Vascular and Neural Integrities in Urinary Bladder of Streptozotocin-Induced Diabetic Mice. Int Neurourol J 2022; 26:201-209. [PMID: 36203252 PMCID: PMC9537429 DOI: 10.5213/inj.2244152.076] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 08/29/2022] [Indexed: 12/16/2022] Open
Abstract
Purpose To assess functional and structural changes in vascular and neural structures associated with diabetic bladder dysfunction (DBD) in the bladders of streptozotocin (STZ)-induced diabetic mice. Methods Eight-week-old C57BL/6 mice were injected with STZ at 50 mg/kg daily for 5 consecutive days. Catheters were inserted 12 weeks later, and 5 days after catheter placement bladder functions were assessed by conscious cystometry. Neurovascular and extracellular matrix marker changes in harvested urinary bladders were investigated by immunofluorescent staining. Body weights and fasting and postprandial blood glucose levels were measured 12 weeks after STZ injection. Results STZ-induced diabetic mice had significantly lower body weights and significantly higher blood glucose levels. Assessment of bladder function in STZ-induced diabetic mice revealed a nearly 3-fold increase in bladder capacity and intercontractile interval compared to controls. However, basal pressure, maximal bladder pressure, and threshold pressure were not significantly different. Morphological and structural analysis showed that STZ-induced diabetic mice had significantly reduced microvascular density in lamina propria (33% of the nondiabetic control values), and severely decreased nerve contents in the detrusor region (42% of the nondiabetic control values). Conclusions STZ-induced diabetic mice exhibit functional and structural derangements in urinary bladder. The present study provides a foundation and describes a useful means of evaluating the efficacies of therapeutic targets and exploring the detailed mechanism of DBD.
Collapse
|